HK1263036A1 - Intranasal testosterone bio-adhesive gel formulations and use thereof for treating male hypogonadism - Google Patents
Intranasal testosterone bio-adhesive gel formulations and use thereof for treating male hypogonadism Download PDFInfo
- Publication number
- HK1263036A1 HK1263036A1 HK19123001.0A HK19123001A HK1263036A1 HK 1263036 A1 HK1263036 A1 HK 1263036A1 HK 19123001 A HK19123001 A HK 19123001A HK 1263036 A1 HK1263036 A1 HK 1263036A1
- Authority
- HK
- Hong Kong
- Prior art keywords
- testosterone
- gel formulation
- gel
- formulation
- treatment
- Prior art date
Links
Description
The invention is a divisional application of Chinese patent application with application number 201280035150.5, application date 2012, 5, 15 and entitled "intranasal testosterone bioadhesive gel preparation and use thereof for treating male hypogonadism".
Technical Field
The present invention relates to 4.0% and 4.5% intranasal testosterone bioadhesive gels for providing sustained intranasal delivery of testosterone to men; and intranasal treatment methods for safely providing sustained release of testosterone to treat men with hypogonadism. In particular, the present invention relates to improved Testosterone Replacement Therapy (TRT) and sustained intranasal testosterone gel formulations for the treatment of male hypogonadism. The present invention also relates to a system for intranasally dispensing precise dosages of such gels in small volumes at optimal anatomical locations within each nostril of a male, such that an effective amount of testosterone is deposited at the optimal anatomical location within each nostril, for use in TRT, including effective treatment of testosterone deficiency, such as hypogonadism, in a male subject.
Background
Androgens are a group of C19 steroids that cause virilization of the reproductive tract and development and maintenance of male secondary sex characteristics. It also contributes to male muscle mass, bone mass, libido and sexual function. Testosterone is the main androgen, which is secreted by the leydig cells of the testis, and whose production is increased during puberty. See, for example, Tietz of Clinical chemistry and Molecular Diagnostics,4th edition, Editors: Burtis CA, Ashwood ER, and BrunsDE (2006.). Androgen deficiency is now recognized as a relatively common condition in elderly men. See, e.g., 2.Wang C, Swerdloff R.S. Androgen replacement therapy. Ann Med,29: 365-; andropause, namely, clinical indications of the decimine in seriumTestosterone levels with the imaging in men.J. Gerontol A Med Sci,57: M76-M99 (2002); and Haren Mtet al.Andropause: a quality-of-life issue in adhesives, Med Clin North Am,90: 1005-. Testosterone hormone therapy is proposed for replacement therapy and treatment of men with conditions associated with a deficiency or absence of endogenous testosterone, such as treatment of male hypogonadism. This can lead to sexual dysfunction, muscle loss, increased fat, infertility, loss of beard and body hair, and other symptoms.
Hypogonadism was identified as testosterone deficiency. Male hypogonadism may be congenital or it may be triggered later in life by, for example, injury, trauma, surgery, infection, disease, medication and/or aging. In general, childhood-onset male hypogonadism has minimal effect and is generally not diagnosed until delayed puberty. Symptoms or signs associated with childhood onset male hypogonadism, if not treated, include muscular and body hair dysplasia, including beard, pubic hair, chest hair and axillary hair dysplasia, elevated voice tones, overgrowth of the arms and legs relative to the body trunk, small scrotum, abnormal growth of the penis and testes, and other growth problems, such as the growth and maturation of the prostate and seminal vesicles. In adult onset male hypogonadism, symptoms may include sperm production deficiency, osteoporosis, muscle loss or body muscles, altered fat distribution, fatigue and energy loss, weakness, anemia, mood swings such as depression and anger, decline in cognitive abilities including memory loss and attention deficit, sleep disturbances, gynecomastia, loss of beard and body hair, impotence, erectile dysfunction; reduced ejaculatory volume, infertility, decreased libido (loss of libido) and other secondary sexual characteristics deterioration.
Male hypogonadism is defined as primary hypogonadism, which results from testicular disorders; or central or secondary hypogonadism, which results from a disorder of the hypothalamic pituitary axis. In primary hypogonadism, testosterone production is deficient in the testes because the testes do not respond to FSH and LH. Thus, an elevation of the two hormones FSH and LH is observed in primary male hypogonadism. The most common cause of primary male hypogonadism is Klinefelter syndrome. Other congenital causes of primary hypogonadism may include, for example, bilateral congenital anemarrhena, leydig dysplasia (leydig hypoplasia), cremaster descent (cryptorchidism), Noonan syndrome, Myotonic Dystrophy (MD), and testosterone enzyme synthesis deficiency. Causes of adult onset primary hypogonadism may include aging, autoimmune disease, surgery, chemotherapy, radiation, infection, illness, surgery, alcohol abuse, drug therapy, and the administration of recreational drugs.
In secondary or central hypogonadism, insufficient amounts of FSH and LH are produced in the hypothalamus. Reproductive causes of secondary or central hypogonadism include, for example, Kallmann's syndrome, Prader-Willi syndrome (PWS), Dandy-Walker malformation, Luteinizing Hormone (LH) deficiency singularity, and Idiopathic Hypogonadotropic Hypogonadism (IHH). Causes of secondary or central hypogonadism of the adult-onset type may include aging, illness, infection, tumor, bleeding, nutritional deficiencies, alcohol abuse, cirrhosis, obesity, weight loss, Cushing's syndrome, hypopituitarism, hyperprolactinemia, hemochromatosis (hemochromatosis), surgery, trauma, drug therapy, and the administration of recreational drugs.
In primary male hypogonadism, lower than normal testosterone levels are observed, but overall FSH and LH levels are higher than normal. In secondary or central male hypogonadism, lower than normal testosterone, FSH and LH levels are observed. Thus, the diagnosis of primary or secondary male hypogonadism is generally determined by hormone levels, and in the test, the blood levels of testosterone for primary and secondary hypogonadism are characterized as low and should be replaced. Treatment generally varies with etiology, but generally involves testosterone replacement therapy. In the united states, testosterone may be administered as an intramuscular injection, a transdermal patch, or a transdermal gel. In other countries, oral formulations of testosterone are available.
In view of the fact that millions of men in the united states and around the world suffer from hypogonadism, there is a real and urgent need for a pharmaceutical treatment that can treat this condition effectively and conveniently, so that the quality of life of these subjects can be improved. One therapeutic goal of such treatment to address this urgent need may be to restore male testosterone levels to levels of young adults with the desire to reduce symptoms that may be commonly associated with hypogonadism due to testosterone deficiency.
Disclosure of Invention
The present invention overcomes limitations and deficiencies associated with current Testosterone Replacement Therapy (TRT) and current testosterone therapy approaches to specifically treat hypogonadism in male subjects by discovering novel transnasal testosterone gels and methods for TRT and treatment of hypogonadism. In particular, the present invention overcomes the limitations and drawbacks of currently available testosterone dosing options by discovering a new and improved dosage strength testosterone gel formulation specifically designed for intranasal administration to deliver a therapeutically effective amount of testosterone to treat men suffering from and/or already diagnosed with testosterone deficiency, including hypogonadism.
The term "therapeutically effective amount" means an amount of testosterone sufficient to elicit a therapeutic or prophylactic effect for use in testosterone replacement or supplemental therapy to treat male testosterone deficiency, i.e., male hypogonadism.
Thus, in general, the present invention provides novel and improved, substantially non-irritating, novel dosage strength testosterone gel formulations formulated with the following amounts of testosterone: between about 4% and 8.0% by weight, preferably between about 4.0% and about 4.5% by weight, more preferably about 4.0%, about 4.5% and 8.0% by weight for nasal administration to deliver a therapeutically effective amount of testosterone to effectively treat men diagnosed with testosterone deficiency, including hypogonadism.
According to the invention, the rate of diffusion of testosterone in the intranasal gel of the invention through the Franz cell membrane, as contemplated by the invention, is between about 28 and 100 slope (slope)/mgT%, preferably about 30 and 95 slope/mgT%. For those intranasal gels formulated with between about 4.0% and 4.5% testosterone, the preferred rate of testosterone diffusion is between about 28 and 35 slope/mgT%.
The present invention also relates to a novel method of nasally administering testosterone gels to the nasal cavity. In general, the new methods involve the local deposition of intranasal testosterone gels into the nasal cavity of each nostril to deliver a therapeutically effective amount of testosterone in a smaller volume over the life of the medicament, providing a constant effective testosterone brain and/or blood level for TRT, particularly for effective treatment of men in need of testosterone to treat hypogonadism.
More particularly, the present invention relates to bioavailable intranasal testosterone gel formulations suitable for nasal administration, for TRT and for the treatment of hypogonadal subjects. According to the invention, and as an example. The invention considers:
treatment with a unit dose device pre-filled with 125 μ Ι _ of 4.0% testosterone gel, delivering about 5.0mg testosterone per nostril (intranasal), given for example three times a day (total dose 30 mg/day);
treatment with a unit dose device pre-filled with about 150 μ Ι _ of 4.5% gel, delivering about 6.75mg testosterone per nostril (intranasal), given, for example, twice daily (total dose 27.0 mg/day); and/or
Treatment with a unit dose device pre-filled with approximately 125 μ Ι _ of 4.5% gel, delivering approximately 5.625mg testosterone per nostril (intranasal), given for example three times a day (total dose 33.75 mg/day).
In general, the intranasal testosterone gel formulations of the present invention are formulated with about 4% and 4.5% testosterone by weight, and testosterone is well absorbed when such gel formulations are administered nasally to hypogonadal subjects. More specifically, testosterone is rapidly absorbed after nasal administration, with peak concentrations reached within 36 minutes to 1 hour 6 minutes (mean Tmax) after nasal administration, and maximum serum concentrations reached after about 1-2 hours after nasal administration. The maximum testosterone concentration during the 24 hour interval was observed in about 57% to 71% of hypogonadal men during the first dose (0-10 hours), while about 29% to 43% of subjects had their maximum 24-h testosterone concentration during the subsequent dose.
Formulations comprising 4% and 4.5% testosterone by weight provide surprising properties. Importantly, the solubility of testosterone in pure castor oil was a maximum of 3.6%, with 4% Labrafil dropping to about 3.36%. Addition of fumed silica gel (Aerosil, CabOsil) increased the solubility of testosterone in castor oil up to 4.5%, even with 4.0% Labrafil. This is counter-intuitive to those skilled in the art. However, without wishing to be bound by any particular theory, it is believed that this increase in solubility in the presence of silica is due, at least in part, to SiO2The fact that about 10% of testosterone is absorbed.
According to the novel method of the invention, intranasal testosterone gel is deposited locally on the outer lateral outer wall (opposite the nasal septum) within the nasal cavity of each nostril, preferably about the middle to about the upper part of the outer lateral outer wall (opposite the nasal septum), directly below the cartilaginous part of the outer lateral outer wall within the nasal cavity of each nostril. After the deposition of gel within each nostril of the nose is complete, the subject then gently and carefully squeezes and/or rubs the lateral nose so that the deposited gel remains in contact with the mucosa within the nasal cavity for sustained release of testosterone during the life of the medicament. Typical nasal administration deposits testosterone gel doses are between about 50 to about 150 microliters per nostril, preferably about 125 to about 150 microliters per nostril.
In practicing the methods of the invention, an intranasal testosterone gel of the invention of between approximately about 50 microliters and about 150 microliters is administered to each nostril of a subject once daily or twice daily or three times daily, e.g., one, two, three, four, or more consecutive weeks, or two, three, four, five, or six consecutive days or more, or intermittently such as once, two, or three times every other day or week, or as needed once or twice daily, as a TRT or to treat male testosterone deficiency, including male hypogonadism.
Furthermore, the invention contemplates pharmaceutically equivalent, therapeutically equivalent, bioequivalent and/or interchangeable testosterone gel formulations for nasal administration, regardless of the method selected to demonstrate equivalence or bioequivalence, such as pharmacokinetic methods, microdialysis, in vitro and in vivo methods and/or clinical endpoints described herein. Thus, the present invention contemplates bioequivalent, pharmaceutically equivalent and/or therapeutically equivalent testosterone gel formulations for nasal administration, in particular testosterone gel formulations for nasal administration, which are a gel formulation of 0.15% testosterone by weight, a gel formulation of 0.45% testosterone by weight and a gel formulation of 0.6% testosterone by weight-when the treatment means according to the invention is used for the treatment of anorgasmia and/or HSDD by intranasal administration. Thus, the present invention contemplates: (a) a pharmaceutically equivalent testosterone gel formulation for nasal administration comprising the same amount of testosterone in the same dosage form; (b) a bioequivalent testosterone gel formulation for nasal administration that is chemically equivalent and that yields comparable bioavailability when administered to the same individual with the same dosage regimen; (c) a therapeutically equivalent testosterone gel formulation for nasal administration that provides substantially the same efficacy and/or toxicity when administered to the same individual with the same dosage regimen; and (d) interchangeable testosterone gel formulations for nasal administration of the present invention that are pharmaceutically, bioequivalent, and therapeutically equivalent.
Although the intranasal testosterone gel of the present invention is the preferred pharmaceutical formulation when practicing the novel method of the present invention, it is to be understood that the novel topical intranasal gel formulation and method of the present invention also contemplates nasal administration of any suitable active ingredient, such as neurosteroids or sex hormones (e.g., androgens and progestins, similar testosterone, estradiol, estrogens, estrones, progestins, and the like), neurotransmitters (e.g., acetylcholine, epinephrine, norepinephrine, dopamine, 5-hydroxytryptamine, melatonin, histamine, glutamate (esters), gamma-aminobutyric acid, aspartate, glycine, adenosine, ATP, GTP, oxytocin, vasopressin, endorphin, nitric oxide, pregnenolone, and the like), prostaglandins, benzodiazepine like diazepam (benzodiazepines like diazepam), Midazolam (midazolam), lorazepam (lorazepam), etc., and PDEF inhibitors like sildenafil (sildenafil), tadalafil (tadalafil), vardenafil (vardenafil), etc., in any suitable pharmaceutical formulation, such as a liquid, cream, ointment, salve, or gel. Examples of additional topical formulations for use in the practice of the novel methods according to the present invention include topical nasal formulations disclosed in: for example, U.S. patent nos. 5,578,588, 5,756,071, and 5,756,071 and U.S. patent publication nos. 2005/0100564, 2007/0149454, and 2009/0227550, the entire contents of which are incorporated herein by reference.
The present invention also relates to encapsulated medicaments comprising the novel and improved testosterone gel formulations of the present invention for nasal administration. For example, the present invention contemplates a prefilled single or multi-dose applicator system for nasal administration to strategically and uniquely deposit nasal testosterone gel at a preferred location within the nasal cavity to practice the novel methods and teachings of the present invention. In general, the applicator system of the present invention is, for example, an airless liquid, a dip tube fluid dispensing system, a pump, a prefilled unit dose syringe, or any other system suitable for practicing the methods of the present invention. The applicator system or pump includes, for example, a chamber pre-filled with a single or multiple doses of the intranasal testosterone gel of the present invention, which is closed by an actuator nozzle or cap. The actuator nozzle may include an outlet channel and a tip, wherein the actuator nozzle is shaped to conform to the inner surface of a user's nares to (a) consistently deliver a balanced dose of an intranasal testosterone gel of the present invention during intranasal administration within the nasal cavity, and (b) deposit at a specified location within each of the patient's nares, as contemplated by the novel methods and teachings of the present invention. Examples of prefilled multi-dose applicator systems include, for example, (a) COMOD systems available from Ursatec, Verpackung-GmbH, Schillerstr.4, 66606St. Wendel, Germany, (b) Albion or digital airless applicator systems available from Airlessystems, RD 14927380Charlev, France or 250 th Route 303 Congers, NY 10950, (c) nasal applicators available from Neopac, The Tube, Hoffmann Neopac AG, Burgdorfssse 22, Postfach, 3672 Oberdilessbach, Switzerland or (d) syringes described in The description examples below.
A nasal multi-dose dispenser device according to an embodiment of the present invention, such as an Albion or Digital airless applicator system, available from airless systems, is composed of a fluid container and a dispensing pump for delivering multiple doses of a gel or other topical formulation. In one embodiment of the invention, the nasal multi-dose dispenser device is adapted for use in an airless liquid dispensing system. In another embodiment of the invention, the nasal multidose dispenser device is adapted for use in a dip tube fluid dispensing system.
One example of an airless system contemplated by the present invention is one such airless system: it will deliver liquids, including gels, without the need for a pressurized gas or air pump to contact the liquid (or gel). Generally, the airless system of the present invention comprises a flexible pouch containing a liquid, a rigid cylindrical container, a moving piston, a suction pump, an administration valve, and a delivery nozzle, as shown, for example, in fig. 1-4. See also fig. 7A, 7B, 8A, 8B, 9A, 9B, 10A, 10B and 11.
The multi-dose dispenser 100 of FIG. 1 is configured with a fluid container 120, a dispensing pump 140, and a cap 102 in accordance with the present invention.
Fluid container 120 includes a container body 122, a base 124, and a neck 126. The dispensing pump 140 is secured to the neck body by the sleeve 128. The top end of the container body 122 is closed by a dispensing pump 140. The sleeve 128 abuts the top end of the container body 122 and tightly grips the neck liner 150. The container body 122 forms a vacuum and contains the fluid to be dispensed.
The dispensing pump 140 is closed by its actuator nozzle 130, which retains a stem 144 at the tip of the stem. The actuator nozzle 130 includes an outlet passage 132 and a tip 134.
The actuator nozzle 130 is shaped to conform to the inner surface of the user's nares. The actuator nozzle 130 is movable between a downward opening position and an upward closing position. The user removes the cap 102 and inserts the actuator nozzle 130 into the user's nostril. When the user pushes the actuator nozzle 130 downward to the open position, fluid in the dosing chamber 180 is drawn through the dispensing pump 140 and exits the tip 134 via the outlet passage 132 of the actuator nozzle 130.
Fig. 2 shows a cross-sectional view of the dispensing pump 140.
The dispensing pump has a body 142 configured with a bottom inlet having an inlet valve 160 with a ball 162 as its valve component. The ball 162 is held in place by a retainer 164 and a return spring 170.
The rod 144 carries a spring cap 172 at its bottom end. The piston 174 is located above the spring cap 172. Rod 144 passes through an axial bore in piston base 176.
The sidewall of piston 174 forms a seal against dispensing pump body 142 by a flange. Sleeve 128 grips stem gasket 152 tightly against stem collar 146, dispensing pump body 142 and the top of piston 174.
A precompression spring 178 is located between the piston base 176 and the rod collar 146. The pre-compressed spring 178 biases the actuator nozzle 130 to the closed position via the stem 144.
A return spring 170, which returns the piston 174 upward, is compressed between two opposing seats on the cage 164 and spring cover 172.
The dispensing pump 140 has an administration chamber 180, the administration chamber 180 being formed between the retainer 164 and the piston 174. When the user pushes the actuator nozzle downward to the open position, fluid in the dosing chamber is drawn through the dispensing pump 140 and dispensed from the tip of the actuator nozzle 130.
When the user releases the actuator nozzle 130 upward to the closed position, fluid in the container body 122 is drawn into the dosing chamber 180 by the dispensing pump 140. Thus, one dose of fluid is ready for the next actuation of the actuator nozzle by the user.
In another embodiment of the present invention, the dispenser 200 of FIG. 3 is configured with a fluid container 220, a dispensing pump 240, and a cap 202.
The fluid container 220 includes a container body 222, a base 224, and a neck 226. The dispensing pump 240 is secured to the neck body by the sleeve 228. The top end of the container body 222 is closed by a dispensing pump 240. The sleeve 228 abuts the top end of the container body 222 and tightly grips the neck liner 250. The container body 222 contains a fluid to be dispensed.
The dispensing pump 240 is closed by its actuator nozzle 230, which actuator nozzle 230 retains the rod 244 at the rod tip. The actuator nozzle 230 includes an outlet passage 232 and a tip 234. The actuator nozzle 230 is shaped to conform to the inner surface of the user's nares. The actuator nozzle 230 is movable between a downward opening position and an upward closing position. The user removes the cap 202 and inserts the actuator nozzle 230 into the user's nostril. When the user pushes the actuator nozzle 230 downward to the open position, fluid in the administration chamber 280 is drawn through the dispensing pump 240 and exits the tip 234 via the outlet passage 232 of the actuator nozzle 230.
Fig. 4 shows a cross-sectional view of the dispensing pump 240.
The dispensing pump has a body 242 provided with a bottom inlet having an inlet valve 260 with a ball 262 as a valve component thereof. The ball 262 is held in place by a retainer 264 and a return spring 270. Optionally, a dip tube 290 may extend downward from the inlet valve 260 and dip into the liquid contained in the container body.
The rod 244 carries a spring cap 272 at its bottom end. The piston 274 is located above the spring cover 272. The rod 244 passes through an axial bore of the piston base 276.
The sidewall of the piston 274 forms a seal against the dispensing pump body 242 via a flange. Sleeve 228 grips stem gasket 252 tightly against stem collar 246, dispensing pump body 242 and the top of piston 274.
Precompression spring 278 is located between piston base 276 and rod collar 246. Precompression spring 278 biases actuator nozzle 230 to the closed position via rod 244.
A return spring 270, which returns the piston 274 upward, is compressed between two opposing seats on the cage 264 and the spring cover 272. The dispensing pump 240 has an administration chamber 280, the administration chamber 280 being formed between the retainer 264 and the piston 274. When the user pushes the actuator nozzle downward to the open position, air enters the dosing chamber 280, which forces the fluid in the dosing chamber to be extracted by the dispensing pump 240 and dispensed from the tip of the actuator nozzle 230.
When the user releases the actuator nozzle 230 upward to the closed position, the air contained in the dosing chamber 280 forces the fluid in the container body 222 to be drawn into the dosing chamber 280. Thus, one dose of fluid is ready for the next actuation of the actuator nozzle by the user.
The amount of fluid drawn into the dosing chamber by the dispensing pump may be a fixed volume. The dispensing pump may be of various sizes to accommodate a range of delivery volumes. For example, the dispensing pump may have a delivery volume of 140 μ l.
The dispenser of the invention may dispense-preferably nasally-a topical intranasal gel or other topical intranasal formulation comprising an alternative or additional active ingredient such as a neurosteroid or sex hormone (e.g. androgens and progestins, similar testosterone, estradiol, estrogens, estrones, progestins, etc.), a neurotransmitter (e.g. acetylcholine, epinephrine, norepinephrine, dopamine, 5-hydroxytryptamine, melatonin, histamine, glutamate, gamma-aminobutyric acid, aspartate, glycine, adenosine, ATP, GTP, oxytocin, vasopressin, endorphin, nitric oxide, pregnenolone, etc.), a prostaglandin, benzodiazepine diazepam, midazolam, lorazepam, etc., and a PDEF inhibitor like sildenafil, tadalafil, vardenafil, etc., in the form of a liquid, cream, Ointment, salve or gel. The dispenser may be adapted for cosmetic, dermatological or pharmaceutical applications. Examples of topical intranasal formulations for topical nasal administration that may be dispensed according to the present invention include the nasal testosterone gels or other intranasal topical gels of the present invention wherein testosterone is replaced by or combined with an effective amount of another active ingredient, such as those described herein above. In addition, other testosterone formulations suitable and contemplated for dispensing from a dispenser and/or according to the methods of the present invention include those disclosed in: for example, U.S. Pat. nos. 5,578,588, 5,756,071 and 5,756,071 and U.S. patent publication nos. 2005/0100564, 2007/0149454 and 2009/0227550, the entire contents of which are incorporated herein by reference.
It will be appreciated by those skilled in the art that the amount of testosterone in the lower dose strength intranasal testosterone gel of the present invention that will be therapeutically effective in a particular condition will depend on the following: dosing regimen, site of administration, specific gel formulation, drug lifetime, and condition being treated. Thus, it is often impractical to determine a specific dosage amount herein; however, it is believed that one skilled in the art will be able to determine the appropriate therapeutically effective amount based on the guidance provided herein, information available in the art regarding testosterone replacement therapy, and routine testing.
It should be further understood that the above summary of the present invention is not intended to describe each disclosed embodiment or every implementation of the present invention. The specification further exemplifies illustrative embodiments. Guidance is provided throughout the specification by way of examples, which examples may be applied in different combinations. In each example, the instances merely serve as representative groups and should not be construed as exclusive examples.
Drawings
The foregoing and other objects, advantages and features of the invention and the manner in which the same are practiced will become more apparent from the following detailed description when taken in conjunction with the accompanying drawings and examples of exemplary embodiments, wherein:
FIG. 1 is a side view of a first embodiment of the present invention;
FIG. 2 is a side cross-sectional view of a dispensing pump according to a first embodiment of the present invention;
FIG. 3 is a side view of a second embodiment of the present invention;
FIG. 4 is a side cross-sectional view of a dispensing pump according to a second embodiment of the present invention;
FIG. 5 is a side view of a second embodiment of the present invention in relation to a cylinder-less assembly of the present invention;
FIG. 6 is a side view of a second embodiment of the present invention with respect to a digital actuator and dome;
figure 7A shows the right nostril of subject #1 after single dose syringe administration;
figure 7B shows the left nostril of subject #1 after administration of a multi-dose dispenser
Figure 8A shows the right nostril of subject #2 after single dose syringe administration;
figure 8B shows the left nostril of subject #2 after administration from a multi-dose dispenser;
figure 9A shows the right nostril of subject #3 after single dose syringe administration;
figure 9B shows the left nostril of subject #3 after administration from a multi-dose dispenser;
FIGS. 10A and 10B illustrate the use of a multi-dose dispenser according to the present invention;
FIG. 11 illustrates a multi-dose dispenser according to the present invention;
FIG. 12 shows Franz cell device location layout for comparative testing according to example 5;
figure 13 is a graph showing the change over time in serum testosterone levels of a 4.5% testosterone bioadhesive gel administered twice daily in each nostril of a hypogonadal male according to the invention as compared to the normal testosterone pharmacokinetics of a young healthy adult male, such as over river MJ et al: clinical Endocrinology,58: 710-;
FIG. 14 shows a comparison between TBS1A 8% (part I);
FIG. 15 shows a comparison between TBS1A 8% (part I);
FIG. 16 shows a comparison between 6-hour and 24-hour runs (RD11101 and RD 11102);
FIG. 17 shows a comparison between TBS1A 4% (part I);
FIG. 18 shows a comparison between TBS1A 4% (part II);
FIG. 19 shows a comparison between TBS1A 4% (part III);
FIG. 20 shows comparative slow diffusion;
FIG. 21 shows a comparison between 6-hour and 24-hour runs (RD11063 and RD 11085); and
FIG. 22 shows a comparison between 400mg and 1 g of gel (RD 11063).
Detailed Description
By way of example and to provide a more complete understanding of the present invention and its attendant advantages, the following detailed description and examples are given with respect to the novel lower dosage strength intranasal testosterone gel, administration device and method of the present invention.
As used in the description of the invention and the appended claims, the singular forms "a", "an" and "the" are used interchangeably and are intended to include the plural forms as well, unless the context clearly indicates otherwise. Also as used herein, "and/or" means and includes any and all possible combinations of one or more of the listed items, as well as no combinations when interpreted in the alternative ("or").
Also as used herein, "at least one" means "one or more" of the listed elements.
Singular forms are intended to include plural forms and are also used interchangeably herein where appropriate and to fall within the meaning unless otherwise explicitly stated.
Except where noted otherwise, capitalized and non-capitalized forms of all terms fall within each meaning.
Unless otherwise indicated, it is to be understood that all numbers expressing quantities, proportions, and numerical properties of ingredients, reaction conditions, and so forth, used in the specification and claims are to be understood as being modified in all instances by the term "about".
All parts, percentages, ratios, etc., are herein by weight unless otherwise indicated.
As used herein, "bioequivalence" or "bioequivalence" means a pharmaceutically equivalent nasally administered testosterone gel formulation or drug product, and its bioavailability (rate and extent of absorption) after administration in the same molar dose or amount is somewhat similar: i.e., its therapeutic effect, is substantially the same with respect to safety and efficacy. In other words, bioequivalent or bioequivalent means that there is no significant difference in the rate and extent to which testosterone can be utilized at the site of action from such a formulation when administered at the same molar dose under similar conditions-e.g., the rate at which testosterone can leave such a formulation and the rate at which testosterone can be absorbed and/or utilized at the site of action to affect TRT, including hypogonadism. In other words, the bioavailability of two nasally administered testosterone gel formulation drug products (same galenic form) at the same molar dose are highly similar and are unlikely to produce clinically relevant differences in therapeutic effect or adverse effect or both. The terms "bioequivalence" and "drug (pharmaceutical) equivalence" and "therapeutic equivalence" are also defined and used herein as follows: (a) FDA, (b) Code of Federal Regulations ("c.f.r."), Title 21, (c) Health Canada (Ministry of Health, Canada), (d) European Medicines Agency (EMEA), and/or (e) Japanese minimum of Health and Welfare (Ministry of Health, japan). Thus, it is to be understood that the present invention contemplates nasal administration of a testosterone gel formulation or a pharmaceutical product: testosterone gel formulations or pharmaceutical products can be administered nasally bioequivalent to other nasal cavities of the invention. As an example, according to the present invention, administration of a testosterone gel formulation or pharmaceutical product by a first nasal cavity is bioequivalent to administration of a testosterone gel formulation or pharmaceutical product by a second nasal cavity when a measurement of at least one pharmacokinetic parameter(s), such as Cmax, Tmax, AUC, etc., of the testosterone gel formulation or pharmaceutical product administered by the first nasal cavity differs by no more than about ± 25% from a measurement of the same pharmacokinetic parameter of the testosterone gel formulation or pharmaceutical product administered by the second nasal cavity of the present invention.
As used herein, "bioavailability" or "bioavailable" means generally the rate and extent of testosterone absorption into the systemic circulation, and more specifically, is intended to reflect the rate or measurement of the rate and extent at which testosterone is available at the site of action or is available for absorption from the drug product at the site of action. In other words, and by way of example, the time-concentration profile of testosterone in the systemic circulation reflects the extent and rate of absorption of testosterone from the lower dose strength gel formulation administered nasally in accordance with the present invention.
As used herein, the term "pharmaceutical equivalence" or "pharmaceutically equivalent" means that the nasally administered testosterone gel formulations or pharmaceutical products of the invention comprise the same amount of testosterone, but not necessarily the same inactive ingredients, in the same dosage form, for the same route of administration and in compliance with the same or comparable pharmacopoeia or other applicable standards of identity, strength, quality and purity, including potency and, where appropriate, content consistency and/or stability. Thus, it is to be understood that the present invention contemplates nasal administration of a testosterone gel formulation or pharmaceutical product that is pharmaceutically equivalent to other nasal administration of testosterone gel formulations or pharmaceutical products used in accordance with the present invention.
As used herein, "therapeutic equivalence" or "therapeutically equivalent" means those of the following formulations or pharmaceutical products for nasal administration of testosterone gel: (a) the same clinical efficacy and safety profile will result when a testosterone pharmaceutical product is used in accordance with the present invention for TRT and for the treatment of testosterone deficiency, including hypogonadism in male subjects; and (b) a pharmaceutical equivalent, e.g., which comprises testosterone in the same dosage form, which has the same route of administration; and it has the same testosterone intensity. In other words, therapeutic equivalence means that chemical equivalents of the lower dose strength testosterone formulations of the invention (i.e., comprising the same amount of testosterone in the same dosage form when administered to the same subject in the same dosage regimen) will provide substantially the same potency and toxicity.
As used herein, "nasally administering a testosterone gel formulation" means a formulation of testosterone that includes a combination of a solvent, a humectant, and a viscosity increasing agent.
As used herein, "plasma testosterone level" means the level of testosterone in the plasma of a subject. Plasma testosterone levels are determined by methods known in the art.
"diagnosis" or "prognosis," as used herein, means the use of information (e.g., biological or chemical information from biological samples, signs and symptoms, physical examination results, psychological examination results, etc.) to predict the most likely outcome, time frame, and/or response to a particular treatment for a given disease, disorder or condition based on comparison of multiple subjects possessing common symptoms, signs, family history, or other data that is relevant to consider the health of the patient or to determine the subject's affliction, e.g., testosterone deficiency, including hypogonadism.
A "subject," according to some embodiments, is a subject whose signs and symptoms, physical examination results, and/or psychological examination results will be determined and documented in connection with the subject's condition (i.e., disease or illness state) and/or in response to a drug candidate or treatment.
"subject", as used herein, is preferably, but not necessarily, limited to a human subject. The subject may be male or female, preferably female, and may be of any race or ethnic group, including, but not limited to, caucasian, african, asian, hispanic, indian, and the like. As used herein, a subject may also include animals, in particular, mammals such as dogs, cats, cows, goats, horses, sheep, pigs, rodents (e.g., rats and mice), lagomorphs, primates (including non-human primates), and the like, which may be treated according to the methods of the invention or screened for veterinary or pharmaceutical drug development purposes. Subjects according to some embodiments of the invention include patients, humans or others in need of a therapeutic treatment for testosterone deficiency, including hypogonadism.
"treatment," as used herein, includes any drug, drug product, method, procedure, lifestyle change, or other modification introduced in an attempt to cause a particular change in the health (i.e., related to a particular disease, disorder, or condition) of a subject.
"drug" or "drug substance", as used herein, means an active ingredient, such as a chemical entity or a biological entity or a combination of chemical entities and/or biological entities, suitable for administration to a male subject for the treatment of testosterone deficiency, including hypogonadism. According to the invention, the drug or drug substance is testosterone or a pharmaceutically acceptable salt or ester thereof.
The term "drug product," as used herein, is synonymous with the terms "drug," medicament, "" therapeutic intervention, "or" pharmaceutical product. Most preferably, the pharmaceutical product is approved for use in accordance with the methods of the present invention by a governmental agency. The pharmaceutical product according to the invention is an intranasal gel formulated with a pharmaceutical substance, namely testosterone.
"diseases," "disorders," and "conditions" are well known in the art and indicate the presence of signs and/or symptoms in a subject or patient that are generally considered abnormal and/or undesirable. The disease or disorder can be diagnosed and classified based on pathological changes. The Disease or condition may be selected from the Disease types listed in standard texts such as Harrison's Principles of Internal Medicine,1997 or Robbinspathetic Basis of Disease, 1998.
As used herein, a patient or subject "diagnosed" or "determined" for a subject having a testosterone deficiency, such as hypogonadism, refers to the process of determining whether the subject has a testosterone deficiency, such as hypogonadism.
As used herein, "control subject" means a subject that has not been diagnosed as having testosterone deficiency or hypogonadism and/or not exhibiting any detectable symptoms associated with these disorders. By "control subject" is also meant a subject that is not at risk of developing testosterone deficiency or hypogonadism, as defined herein.
The testosterone gel formulations of the present invention are viscous and thixotropic oil-based formulations comprising testosterone solutions intended for intranasal administration. The non-irritating formulation is designed to adhere to the nose. Furthermore, it acts as a control matrix, thus allowing for sustained drug delivery through the nasal mucosa.
Other pharmacologically inactive ingredients in the testosterone intranasal gel are castor oil USP, oleoyl polyoxylglycerides EP and colloidal silicon dioxide NF. These excipients are of no human or animal origin. All excipients are well known and listed in the FDA-issued "inactive ingredient" list of approved pharmaceutical products.
The steroid hormone testosterone is the active ingredient in the testosterone gel formulation of the present invention. The preparation of pharmaceutical substances presents no potential danger to humans; the synthetic pathway is well characterized.
Table 1: testosterone nomenclature
Structural formula (I)
Molecular formula
C19H28O2
Relative molecular mass
288.4
Physical and chemical characteristics
The physicochemical properties of testosterone are listed in table 2.
Table 2: general characteristics of Testosterone
The testosterone of the testosterone gel formulation of the invention appears as white or slightly milky crystals or crystalline powder. It is readily soluble in methanol and ethanol, soluble in acetone and isopropanol and insoluble in n-heptane. It can also be considered insoluble in water (S)20℃=2.41×10-2g/L±0.04×10-2g/L); its n-octanol/water partition coefficient (log P)OWDetermined by HPLC) was 2.84. The solubility of testosterone in oil was determined to be 0.8% in isopropyl myristate, 0.5% in peanut oil, 0.6% in soybean oil, 0.5% in corn oil, 0.7% in cottonseed oil, and up to 4% in castor oil.
Since testosterone is sufficiently solubilized in the formulation of the invention, the physical characteristics of the drug substance do not affect the performance of the drug product, the testosterone gel formulation of the invention. However, the manufacturability of the testosterone gel formulations of the present invention is affected by the testosterone particle size. The solubility of the drug substance in the matrix is particularly advantageous when particle sizes of 50% ≦ 25 microns, 90% ≦ 50 microns are used.
According to the invention, when formulated into the intranasal testosterone gel of the present invention, the testosterone drug may be, for example, in crystalline, noncrystalline, micronized, non-micronized, powder, small particle or large particle form. Exemplary ranges of testosterone particle size include about 0.5 microns to about 200 microns. Preferably, the testosterone particle size is in the range of about 5 microns to about 100 microns, and testosterone is in crystalline or amorphous and non-micronized or micronized form. Preferably, testosterone is in crystalline or amorphous micronized form.
The molecular structure of testosterone does not contain functional groups that can be protonated or deprotonated at physiological pH-ranges. Thus, testosterone will be considered to be a neutral molecule with no pKa value in the range 1-14. Because it is neutral, testosterone is compatible with excipients.
The testosterone gel formulations of the present invention are viscous and thixotropic oil-based formulations comprising testosterone solutions intended for intranasal administration. The non-irritating formulation is designed to adhere to the nose. Furthermore, it acts as a control matrix, thus allowing for sustained drug delivery through the nasal mucosa.
Other pharmacologically inactive ingredients in the testosterone intranasal gel are castor oil USP, oleoyl polyoxylglycerides EP and colloidal silicon dioxide NF. These excipients are of no human or animal origin. All excipients are well known and listed in the FDA-issued "inactive ingredient" list of approved pharmaceutical products.
According to the "Handbook of Pharmaceutical Additives", oleoyl polyoxylglycerides are used as hydrophilic oils for topical, injectable and nasal agents. In FDA-approved pharmaceutical products, they are used as co-emulsifiers in topical and vaginal lotions/creams. In France, such excipients are approved for use in nasal preparations such as "Rhino-Sulforman" (Laboratoire Jolly-Jatel, France; containing 10% oleoyl polyoxylglycerides) and "HuileGomenolee 2% (" Laboratoire GomLe nol, France; containing 10% oleoyl polyoxylglycerides). Thus, like castor oil, oleoyl polyoxylglycerides are inferred to be suitable for routes of administration where safety and tolerability are of highest importance (e.g. injections and nasal or vaginal preparations).
Oleoyl polyoxylglycerides are also known as Labrafil M1944 CS, almond oil PEG-6 ester, polyethylene glycol-5-oleate, mixtures of glycerides and polyvinyl esters. Castor oil, which is used as a solvent for the testosterone gel formulation of the present invention, is a fixed oil. Such oils have the advantage of being non-volatile or diffusive (as opposed to volatile oils or liquid paraffin), but have the disadvantage of being hydrophobic. The nasal mucosa contains 95-97% water. In the absence of an oil acyl polyglycol-glyceride, castor oil containing the active ingredient will form a non-interacting layer on the mucosa. To achieve sufficient contact between the castor oil layer and the mucosal membrane, a hydrophilic oleoyl polyethylene glycol-glyceride oil is added to the formulation to form an emulsion between the castor oil and the mucosal fluid.
Depending on the application, oleoyl polyoxylglycerides are applied as a semi-solid in a concentration range of about 3 to 20%. The amount of oleoyl macrogol-glyceride in the testosterone gel formulation of the invention is high enough to allow better contact of the carrier oil with the mucosa, and low enough to have minimal effect on the amount of testosterone that can be incorporated into the carrier oil. An advantageous concentration of oleoyl polyethylene glycol-glyceride in the testosterone gel formulation of the invention was found to be 4% of the formulation.
According to the "Handbook of Pharmaceutical Additives", colloidal silica is used as oil absorbent, heat stabilizer and gelling agent. In FDA-approved pharmaceutical products, they are used in dental gels, sublingual tablets, endocervical gels, suppositories, vaginal creams/tablets/tampons and inhalation capsules. In addition, it was used as an excipient in "Testoderm with adhesive" (Alza Corporation, approved in 1996) testosterone transdermal patches. Thus, it can be concluded that colloidal silica is suitable for administration routes where safety and tolerability are of the highest importance (e.g. inhalants, endocervical, vaginal or rectal formulations).
For clinical trial delivery, testosterone intranasal gel was delivered in a unit dose syringe consisting of: a syringe body made of polypropylene; a plunger molded from polyethylene; and a syringe cap made of high density polyethylene. The syringes were packaged in aluminum foil as a secondary package. The prefilled unit dose syringe according to the study application in the examples was filled as follows: (a) 4% testosterone intranasal bioadhesive gel-148 microliters and 5.92mgs testosterone; (b) 4.5% testosterone intranasal bioadhesive gel-148 microliters and 6.66mgs testosterone; and (c) 4.5% testosterone intranasal bioadhesive gel-148 microliters and 7.785mgs testosterone.
The oil in the testosterone gel formulation of the present invention is thickened with colloidal silicon dioxide, which acts as a gel former. Such compounds are commonly used in hardened oleogels.
Contemplated dosage forms of the testosterone gel formulations of the present invention are semi-solid, non-liquid. The formulation is thickened with colloidal silicon dioxide. Colloidal silica is believed to contribute to the thixotropic properties of the gel, simplifying drug delivery to the nostrils.
Colloidal silica is generally an inert material that is well tolerated as an excipient in mucosal applications such as suppositories. Colloidal silica is typically used in these formulations at concentrations ranging from about 0.5 to 10%. The concentration of colloidal silicon dioxide in the testosterone gel formulation of the invention is high enough to achieve gel formation, but at a level that has minimal effect on testosterone incorporation into the carrier oil.
Preferably, the intranasal testosterone gels of the present invention typically have a viscosity ranging between about 3,000cps and about 27,000 cps. It will be understood by those skilled in the art that while the above viscosity ranges are considered to be preferred viscosity ranges, any suitable viscosity or viscosity range that does not defeat the purpose of the invention is contemplated.
A detailed description of the testosterone gel formulation batches of the present invention is shown in table 3.
Table 3: composition of the Testosterone gel formulations of the invention
The testosterone gel formulations of the present invention are stored at room temperature (20-25 ℃ or 68 to 77 ° f). Temperature excursions of 15 to 30 ℃ or 59 to 86 ° f may be allowed for the testosterone gel formulations of the present invention. Stability data supports a 12month shelf life. The unit dose syringe is selected for primary packaging of clinical material for clinical trials described below, enabling ease of medication, the ability to generate multiple doses by varying fill volume, and consistency in delivered dose. The syringe is composed of a syringe body, a plunger, and a syringe cap. The syringe body is molded from polypropylene, the plunger is molded from polyethylene, and the cap is made from HDPE. These syringes are designed and prepared to deliver low volumes of sterile and non-sterile solutions, liquids and gels. With respect to additional environmental (i.e., exposure to dust, light, moisture, and oxygen) barrier protection, the syringe is enclosed in a foil laminated overwrap pouch.
The syringe and cap are designed for clinical settings and comply with EU medical devices Directive 93/42/EEC, 6, 14, 1993 and modified regulations. Since this container closure is only intended for this part of the clinical protocol, no further studies have been made on syringes and syringe assemblies.
With regard to further protection elements, the two syringes are contained in a secondary package consisting of an aluminum foil pouch. The two syringes were enclosed in aluminum foil pouches and each pouch was sealed.
The pouch is comprised of a flexible 3-layer-foil-laminate wherein: a) polyester 12 microns, b) aluminum 12 microns and c) polyethylene 75 microns. It is produced by Floeter Flexibles GmbH and is supplied under the name "CLIMAPAC II 12-12-75".
The present invention provides an intranasal bioadhesive gel formulation of testosterone to be administered intranasally, wherein the formulation dose is about 4.0% or 4.5% testosterone by weight of the gel.
The methods and treatments of the present invention are suitable for male TRT, and in particular for treating testosterone deficient male subjects, such as those diagnosed with hypogonadism.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
Examples
Having now generally described the invention, it will be more readily understood by reference to the following examples, which are provided as illustration and are not intended to limit the invention unless otherwise indicated.
The following examples are provided for illustrative purposes only and are not intended to limit the scope of what the inventors regard as their invention.
Example 1
Description and composition of Testosterone gel formulations of the invention
The composition of the three different concentrations of drug product administered in this clinical trial is provided in the table below.
Description of the dosage forms
The testosterone gel formulations of the present invention are viscous and thixotropic oil-based formulations comprising solubilized testosterone that are intended for intranasal administration. The medicine product is prepared from the following non-active ingredients in pharmacopoeia: castor oil, oleoyl polyoxylglycerides, and colloidal silica.
Two different doses of the testosterone gel formulation of the invention were administered intranasally: 0.4% w/w and 0.45% w/w. Each syringe was added in excess to account for the gel remaining in the syringe after administration. This excess remained consistent at 23 μ l regardless of the gel volume in the syringe.
4.0% and 4.5% intranasal Testosterone composition
Table 1: ingredient, quantity, quality standard and function-0.4% testosterone gel formulation of the invention
TABLE 6 ingredients, amounts, quality standards and Functions-0.6% Testosterone gel formulations of the invention
Table 2: components, quantity, quality standards and functions, TBS-1: 5.6 mg/125. mu.l/syringe (4.5% gel)
Table 3: components, quantity, quality standards and functions, TBS-1: 6.75 mg/150. mu.l/syringe (4.5% gel)
Container with a lid
The testosterone gel formulation of the present invention is supplied in a unit dose polypropylene syringe. The two syringes for each dose were enclosed in a protective aluminum foil pouch.
Example 2
Intranasal testosterone gel formulations
The testosterone gel formulation of the present invention is a testosterone formulation of testosterone in an intranasal gel that is proposed for evaluating the pharmacokinetics of two different doses of the testosterone gel formulation of the present invention in hypogonadal men.
The active ingredient, testosterone, from Bayer Schering.
Challenges with nasal delivery include:
particles need to be larger than for pulmonary administration (i.e. only particles >10 μm are heavy enough to avoid entering the respiratory tract);
the concentration must be higher because smaller volumes can be administered;
rapid clearance of the therapeutic agent from the site of deposition results in a shorter time available for absorption;
there may be local tissue irritation; and
limited formulation manufacturing possibilities that alter drug delivery characteristics.
Testosterone is indicated for TRT in men with testosterone deficiency for any number of reasons, including hypogonadism. Currently available testosterone administration options are oral, buccal, injectable, implantable, and transdermal (patches and gels).
Intranasal testosterone (3.2%) gels were developed for The treatment of Male hypogonadism and have been administered to hypogonadism men in several clinical trials, see, e.g., matern, c.et al, 2008The Aging Male11(4):171-178(Dec 2008, The entire contents of which are incorporated by reference in phase II study NCT00975650, which was performed in The united states, testosterone deficient men and supplemented with matern et al, Romanian study reported above, 3.2% intranasal gels, such as matern et al, reported above, did not achieve The FDA-required plasma levels of testosterone to support TRT efficacy in testosterone deficient men.
Example 3
Excess of
[ Testosterone gel preparation of the invention]
No excess was added to the formulation. An excess was added to each syringe to account for the gel remaining in the syringe after administration. This excess remained consistent at 23 μ l regardless of the gel volume in the syringe. The theoretical fill and dispense amounts of the testosterone gel formulations of the present invention are provided below.
Example 4
Physicochemical and biological Properties
[ Testosterone gel preparation of the invention]
The testosterone bioadhesive gel formulation of the present invention has a viscosity ranging from 3,000 to 10,000mPa x sec. Viscosity is very important because it helps the gel in the nasal cavity to remain in contact with the nasal mucosa. When the viscosity is less than about 3,000mPa × sec (i.e., 3,000 centipoise), the gel tends to be pulled out of the nasal cavity by gravity.
Example 5
Batch formulation
[ Testosterone gel preparation of the invention]
Three different concentrations of testosterone gel formulations of the invention, 0.15%, 0.45% and 0.6%, were prepared for the proposed clinical trials. The batch formulations for these batches are shown in table 5 below.
Table 5: 200KG batch formulation, 4.0% and 4.5% bioadhesive testosterone gel formulation of the invention, 8KG batch size
Example 6
Preparation method and process control
[Testosterone gel formulations of the invention]
The material was prepared according to the following method.
Flow chart of preparation process
Mixed ingredient-batch gel
The premix was prepared by: sufficient testosterone was mixed with castor oil portion 1 for 10 minutes using a propeller mixer. Mixture I was prepared by: add premix to remaining castor oil and mix for 60 minutes. The product temperature was kept below 50 ℃ throughout the mixing process.
Oleoyl polyoxylglycerides were preheated to 40-50 ℃ and mixed for 10 minutes before being added to mixture I. It was identified as mixture II. It was mixed for 45 minutes while keeping the product temperature below 50 ℃. Mixture II was then screened through a screen to remove any undissolved testosterone aggregates.
Mixture III was prepared by: colloidal silica was added to mixture II and mixed for 15 minutes while maintaining the product temperature below 50 ℃. Visual inspection was performed after this step to ensure the gel was transparent.
Upon completion of mixing, the gel was stirred and cooled to a product temperature below 30 ℃. The product was then discharged into a stainless steel drum and a bulk gel sample was extracted for analytical testing.
Filling and packaging-clinical supply
After release of the final gel mixture by the quality control laboratory, the filling and encapsulation process was performed by: fill a predetermined volume into the syringe and then cap the syringe cap. The two syringes were enclosed in foil pouches.
The syringe was filled with a pipette, with the gel taken from a storage tank. After filling the syringe, the pipette tip is discarded and the syringe cap is capped. Each syringe was individually labeled.
After the mark is applied, the two syringes are enclosed in a preformed foil pouch and the pouch is sealed. Each pocket is marked.
Example 7
The pharmaceutical product, TBS-1, is a viscous and thixotropic oil-based formulation comprising solubilized testosterone, intended for intranasal administration, for use in the treatment of male hypogonadism.
The pharmaceutical product is formulated with the following pharmacopoeia inactive ingredients: castor oil, oleoyl polyoxylglycerides, and colloidal silica.
To allow different doses to be administered in a phase II regimen, the syringe is used as a unit dose container for clinical supply.
The syringe intended for clinical procedures is needle-free, with a twist-off cap applied to the syringe end. The syringe is composed of a syringe barrel and a plunger. The syringe barrel is formed of polypropylene. The plunger is formed of polyethylene. The syringe cap is formed from High Density Polyethylene (HDPE).
A new dose TBS-1 formulation was prepared for clinical study TBS-1-2010-01 (submitted to the institution at 07/28/2010, serial No. 0019). The amount of testosterone in these formulations was 4.0% and 4.5% with the concomitant regulation of castor oil mass. The precise formulations are listed in tables 1,2 and 3. TBS-1 was concentrated so that the same dose was given intranasally in a smaller volume.
Three different concentrations of TBS-1 gel will be administered in this clinical trial 5.0 mg/125. mu.l/syringe (4.0% gel), 5.6 mg/125. mu.l/syringe (4.5% gel) and 6.75 mg/150. mu.l/syringe (4.5% gel). An excess was added to each syringe to account for the gel remaining in the syringe after administration. This excess remained consistent regardless of the volume of gel in the syringe.
Composition of
The composition of the three different concentrations of drug product to be administered in this clinical trial is provided in tables 1,2 and 3.
Table 1: components, quantity, quality standards and functions, TBS-1: 5.0 mg/125. mu.l/syringe (4.0% gel)
Table 2: components, quantity, quality standards and functions, TBS-1: 5.6 mg/125. mu.l/syringe (4.5% gel)
Table 3: components, quantity, quality standards and functions, TBS-1: 6.75 mg/150. mu.l/syringe (4.5% gel)
Container with a lid
TBS-1 gel was supplied in a unit dose polypropylene syringe. Two syringes for each dose were enclosed in a protective aluminum foil pouch.
Control of pharmaceutical products [ TBS-1, gel ]
Provisions [ TBS-1, gel ]
TBS-1 batch gels were tested as per the following batch release protocol.
Table 1: TBS-1 batch gel specification
The finished TBS-1 gel, packaged in a unit dose syringe, was examined according to the following batch release specifications.
Table 2: specification of TBS-1 gel-Encapsulated Unit dose Syringe
TAMC-Total aerobic microbial count
TYMC-Total combination Yeast/mold count
Batch analysis [ TBS-1, gel ]
TBS-1 has been produced in one preliminary batch (batch No. 100304), four pilot scale batches (batch Nos. ED 187, ED188, ED 189 and ED 014), two pilot non-GMP batches (NA 090811-1 and NA090723-1) and three industrial scale batches (batches 9256, 0823 and 0743). Data for new lots 0823 and 0743 are described in tables 4 and 5.
Table 3: TBS-1 batch description
Batch 0743, batch 4.5% testosterone gel, was filled to two different dose strengths, 5.6mg (batch 0943) and 6.75mg (batch 0744), by varying the gel weight in the finished syringe. Batch 0823, batch 4.0% testosterone gel, filled to a dose strength of 5.0mg (batch 0942).
Table 4: batch analysis-TBS-1 batches 0743 and 0823
TAMC-Total aerobic microbial count
TYMC-Total combination Yeast/mold count
Table 5: batch analysis-TBS-1 batches 00744, 0942 and 0943
Stability [ TBS-1, gel ]
Stability summarization and summarization [ TBS-1, gel ]
This section has been modified to include additional data on ongoing stability studies on primary stability batches and to provide stability data on drug products in syringes for phase II clinical studies. Only the update part and new information have been included for review.
All stability studies of TBS-1 gels have been performed by ACC GmbH Analytical Clinical Concepts,9-11,63849 Laidelsbach/Ashofburgh, Germany. Stability studies are ongoing that meet ICH requirements.
Table 1: stability studies supporting TBS-1 implementation
In general, this portion of the provided stability data is summarized as supporting the stability of the polymer at controlled room temperature conditions [ i.e., 25 ℃ (77 ° f); transport TBS-1 stored at 15-30 deg.C (59-86F.) ] for a 24 month "use" period. The data also show that the pharmaceutical product does not require special storage conditions. The packaging configuration is sufficient to protect the drug product from light, which does not physically degrade or change after exposure to temperature cycling stresses.
When conditioned at controlled room temperature [ i.e., 25 ℃ (77 ° f); when shipped at 15-30 ℃ (59-86 ° f), the clinical supply is eligible for a 1-year review period to reflect the validity period of the trial and available data. If additional data is available, the review period will be extended appropriately.
Stability data [ TBS-1, gel ]
In this section, an updated stability data table is provided for commercial size batch lots 9256, 0743, and 0823 and finished lot lots 9445, 9446, 9447, 0943.
A real-time stability protocol for 6 months on an industrial scale batch (bulk) (batch 9256) is being implemented. A 36 month real time and 6 month accelerated stability protocol was performed for three different dose batches 9256 packaged in 1ml syringes: batch 94454.0 mg (3.2% gel), batch 94465.5 mg (3.2% gel), batch 94477.0 mg (3.2% gel).
A 6 month real time stability protocol was performed for industrial scale batch batches 0743 (4.5% gel) and 0823 (4.0% gel). A 36 month real time and 6 month accelerated stability protocol was performed for batch 0943 (batch 0743 filled in a 1ml syringe).
Table 2: stability program for commercial-Scale batches of TBS-1 gel and finished product filled in 1ml syringes
Table 3: stability data, TBS-1 batch 9256 (3.2% batch gel) prepared 7 months 2009, stored at ambient temperature
Table 4: stability data, 4.0mg TBS-1 batch 9445 (3.2% gel) 1ml syringe (25. + -. 2 ℃, 60. + -. 5% RH, horizontal)
Table 5: stability data, 4.0mg TBS-1 batch 9445 (3.2% gel) 1ml syringe, (40. + -. 2 ℃, 75. + -. 5% RH, horizontal)
Table 6: stability data, 5.5mg TBS-1 batch 9446 (3.2% gel) 1ml syringe (25. + -. 2 ℃, 60. + -. 5% RH, horizontal)
Table 7: stability data, 5.5mg TBS-1 batch 9446 (3.2% gel) 1ml syringe (40. + -. 2 ℃, 75. + -. 5% RH, horizontal)
Table 8: stability data, 7.0mg TBS-1 batch 9447 (3.2% gel) 1ml syringe (25. + -. 2 ℃, 60. + -. 5% RH, horizontal)
Table 9: stability data, 7.0mg TBS-1 batch 9447 (3.2% gel) 1ml syringe (40. + -. 2 ℃, 75. + -. 5% RH., level)
Table 10: stability data, 5.6mg TBS-1 batch 0943 (4.5% gel) 1ml syringe (25. + -. 2 ℃, 60. + -. 5% RH, horizontal)
Table 11: stability data, 5.6mg TBS-1 batch 0943 (4.5% gel) 1ml syringe (40. + -. 2 ℃, 75. + -. 5% RH, horizontal)
Table 12: stability data, TBS-1 batch 0743 (4.5% gel) batch, stored at ambient temperature
14: stability data, TBS-1 batch 0823 (4.5% gel) batch, stored at ambient temperature
Example 8
This was a phase 2 study designed to investigate intranasal absorption of 4% drug three times a day and 4.5% drug given twice a day and three times a day, and to compare absorption from previous studies in the same subjects responding to a 3.2% testosterone gel. In previous studies, 4.0mg, 5.5mg and 7.0mg testosterone were delivered intranasally using Nasobol-01-2009, 3.2% testosterone gels using gel volumes of 125. mu.L, 172. mu.L and 219. mu.L, respectively. In this study, 5.0mg, 5.65mg and 6.75mg testosterone were administered in gel volumes of 125 μ L, 125 μ L and 150 μ L, respectively. This study allowed the study of the delivery of similar testosterone amounts in much smaller volumes.
In this open-label study, subjects were fairly randomly selected into three treatment groups. Treatment was given in a parallel design for one week. At the end of one week, the three treatments were compared by performing a 24 hour pharmacokinetic study of systemic absorption of the drug product testosterone and its two physiological metabolites dihydrotestosterone and estradiol.
8. Purpose of study
8.1 first purpose
The first objective of this study was to determine bioavailability by PK analysis of 4% TBS-1 gel (administered three times a day) and 4.5% TBS-1 gel (administered two times a day and three times a day) in hypogonadal men.
8.2 second purpose
The second objective of the study was to establish the safety profile of TBS-1.
9. Study plan
9.1 Overall study design and plan description
This is an open label, randomized, balanced, triple-treatment (4.0% t.i.d., 4.5% b.i.d., and 4.5% t.i.d.) parallel design pharmacokinetic study of TBS-1 administered intranasally. Serum concentrations of total testosterone, dihydrotestosterone and estradiol were measured using a validated LC/MS method.
Subjects with hypogonadism were required to visit the clinic in three (3) cases, one of which (1) visit (visit 3) required overnight retention to achieve the aforementioned 24-hour pharmacokinetic profile.
All subjects determined the following pharmacokinetic parameters:
calculate AUC over 24 hours0-τ、Cavg、Cmin、Cmax、tmaxPTF and PTS mean and standard error of the mean.
C calculation of Testosterone, Dihydrotestosterone and estradiolavgA percentage of objects below, within, and above the reference range for each analyte.
Erythrocytosis, anemia, and infection were monitored by measuring whole blood cell counts at screening and cessation of visits.
Approximately 30 subjects are planned to be recruited. Twenty-two (22) subjects completed the study. Study participation ranged from 2 to 3 weeks.
9.2 study design discussion
Testosterone therapy in hypogonadal men should correct clinical abnormalities of testosterone deficiency, including sexual dysfunction. Testosterone reduces body fat and increases lean muscle mass and bone density with minimal side effects.
There are several testosterone replacement products available which can be administered intramuscularly, orally, in buccal tablets to the gums or topically in patches or gels. Current replacement therapies have certain drawbacks. Testosterone injections showed wide fluctuations in serum testosterone levels, usually at values above the reference range (5). Testosterone patches have a high skin irritation rate (6, 7). Testosterone gels, while commonly used in north america, are not necessarily convenient and risk skin-to-skin transfer to family members (8, 9). Oral testosterone undecanoate needs to be given with a high fat meal and the levels obtained are usually very low (10-12).
Intranasal administration of a novel testosterone formulation (TBS-1) has been shown to be efficiently absorbed and to show excellent ability as a therapeutic product for the treatment of male hypogonadism (13). The nasal mucosa provides an alternative route of administration that is not first-pass, is highly osmotic and easy to administer, and is rapidly absorbed into the systemic circulation, resulting in high plasma levels-similar to those observed following intravenous administration.
The advantages of testosterone nasal gel when compared to other formulations are as follows: convenient administration forms-allowing unobtrusive use, much smaller amounts of active ingredient required by the subject, and known that this type of administration is less likely to contaminate other family members (wife and child).
Several studies have indicated the applicability of testosterone administration using nasal gels. Previous studies were conducted in 2009 to demonstrate the efficacy of TBS-1 in treating hypogonadal men in need of testosterone replacement therapy. Efficacy was determined by: establishment of optimal pharmacokinetic profiles of serum testosterone levels according to the multi-dose b.i.d. dosing profile of TBS-1, with three different strengths of testosterone (8.0mg, 11.0mg and 14.0mg) and comparison with the active controlAnd (6) comparing. The second objective of this study was to establish the security features of TBS-1. This will be achieved by: adverse and severe adverse events were monitored throughout the study, and various subsequent safety parameters were compared to those obtained at baseline. These safety parameters consist of vital signs, complete blood counts, chemical characteristics, endocrine characteristics, and urinalysis. In addition, subsequent nasal mucosal and prostate changes were compared to baseline.
An important advantage of the dose-finding design drive of this study is that it minimizes subject selection bias and often observes different host groups in sequential study design.
Schiff & Company monitors three clinical sites to ensure subject safety and clinical study performance according to ICH E6 and FDA guidance.
The central laboratory was used for hematology and biochemistry parameter analysis to obtain consistent and fair laboratory results. The second central laboratory was used for PK analysis.
The following are specific activities in the study design during the visit of the subjects:
physical examination was only at screening and day 8.
Informed consent was signed before screening visit 1, In/Ex phase: a selection and exclusion period.
2If the subject received a prior normal prostate examination at Nasobol-01-2009, it would not need to be performed.
3Chemical overview: Na/K, glucose, urea, creatine, total bilirubin, albumin, calcium, phosphate, uric acid, AST, ALT, ALP, GGT, and CK.
4Complete blood cell count and differential.
5Urine dipsticks (no microscope).
6Cocaine, cannabinoids, opioids, benzodiazepines.
7Urine alcohol, through the dip sheet.
8Serum testosterone, dihydrotestosterone and estradiol for T and DHT will be measured by the reference laboratory using a validated LC-MS/MS method, and for estradiol, using a validated LC-MS/MS or immunoassay method.
Screening visit 1
Subjects, interviewed by a clinical researcher or his/her designated physician/nurse practitioner after having voluntarily signed an informed consent, who acquired medical and physical history, recorded demographic data, and performed routine physical examinations. Weight and height were measured and BMI was calculated. Vital signs (sitting for 5 minutes) were measured (blood pressure, heart rate, respiration rate and body temperature).
If the subject had a normal digital rectal examination of the prostate in the last Nasobol-01-2009 trial, it was not repeated.
Clinical investigators evaluated subject study eligibility based on inclusion/exclusion criteria, and that a subject eligible for testosterone replacement therapy required a washout period; the stored product is administered intramuscularly for four (4) weeks (e.g., testosterone heptanoate 200mg/mL), and the product is administered orally or topically (patch, gel, or buccal cavity) for two (2) weeks. At the end of the washout period, subjects returned to measure their serum testosterone.
No treatment subjects needed no washout period.
Under fasting conditions, blood was drawn for serum testosterone at 0900h ± 30 min. Serum testosterone levels must be >150ng/dL, and <300 ng/dL.
Blood was drawn for clinical laboratory studies after an overnight fast (8-10 hour fast) and included the following:
o complete blood cell count (hemoglobin, hematocrit, MCV, MCHC, RBC, WBC & Difference)
Clinical chemistry Profile (Na/K, glucose, Urea, creatine, Total bilirubin, Albumin, calcium, phosphate, uric acid, AST, ALT, ALP, GGT, and CK)
o serum PSA
o test for HBV, HCV and HIV (hepatitis B surface antigen, hepatitis C antibody, HIV antibody)
o whole blood sample for hemoglobin A1c
Urine o for dipstick urinalysis
o urine was used for drug screening (cocaine, cannabis, opioids and benzodiazepines). Subjects who tested positive were not recruited unless the positive test was due to intervention by a physician-specified drug
Urine for alcohol test
The otorhinolaryngological nasal endoscopy was done by ENT specialist.
Subjects meeting all inclusion and exclusion criteria were enrolled into the study and randomly selected to one of three treatment groups (A, B or C).
Visit 2 (day 1)
Subjects arrived at the clinic at 2000 or earlier under fasting conditions (6-8 hour fasting).
Instructions for appropriate skill to administer to the subject for intranasal administration of TBS-1.
Blood was drawn at 2045 for baseline serum testosterone, dihydrotestosterone and estradiol concentrations.
Vital signs (sitting for 5 minutes) (blood pressure, heart rate, respiration rate and body temperature) were measured to establish a baseline.
Giving the subject a weekly supply of bags: treatment A was 18 pockets, treatment B was 12 pockets, and treatment C was 18 pockets. The pockets required for dosing during the pharmacokinetic profile remain with the clinical investigator. Each pouch for treatment A, B or C contained two syringes pre-filled with TBS-1 gel.
TBS-1 was administered to the subject at 2100 in accordance with its treatment group at its first dose.
At 2200 vital signs are measured and the subject and his bag supply for his treatment group are sent home.
Telephone inspection (day 4)
On day 4, all subjects were called to check compliance with study drug administration, 48 hour abstinence compliance and to record any adverse events that may have occurred. The reminder subject carries all syringes for counting at visit 3.
Visit 3 (day 7)
Subjects arrived at the clinic at 2000 or earlier under fasting conditions (6-8 hour fasting).
Blood was drawn at 2045 for baseline serum testosterone, dihydrotestosterone and estradiol concentrations.
Subject carries out a 24 hour pharmacokinetic profile immediately after dosing at 2100. Vital signs were recorded hourly two hours after dosing.
Record security parameters.
Subjects remained fasted for two hours after dosing and were then given a dinner. After dinner, subjects fasted again overnight and remained fasted until day 8 at 0900. Lunch and dinner on day 8 were performed at regular times and were not subjected to fasting conditions.
Pharmacokinetic blood draw
Administration of the drug should occur at the indicated times (2100 h and 0700h for b.i.d. medication, and 2100h, 0700h and 1300h for t.i.d. medication) ± 5 minutes.
Blood draw should be within the specified time ± 5 minutes when the blood draw interval is ≤ 30 minutes, and within the specified time ± 15 minutes when the blood draw is >30 minutes.
Treatment A: blood draw for serum testosterone, dihydrotestosterone and estradiol measurements: t.i.d. blood draw on medication was completed at 2100 following time after drug administration; 0.33, 0.66, 1.0, 1.5, 2.0, 3.0, 6.0, 9.0, 9.75, 10.33, 10.66, 11.0, 11.5, 12.0, 13.0, 14.0, 15.75, 16.33, 16.66, 17.0, 17.5, 18.0, 20.0, 22.0 and 24.0 hours (total blood draw; 25+ baseline).
Treatment B: blood draw for serum testosterone, dihydrotestosterone and estradiol measurements: b.i.d. blood draw on medication was completed at 2100 following time after drug administration; 0.33, 0.66, 1.0, 1.5, 2.0, 3.0, 6.0, 9.0, 9.75, 10.33, 10.66, 11.0, 11.5, 12.0, 13.0, 16.0, 19.0, 22.0, and 24.0 hours (total blood draw; 19+ baseline).
Treatment C: blood draw for serum testosterone, dihydrotestosterone and estradiol measurements: t.i.d. blood draw on medication was completed at 2100 following time after drug administration; 0.33, 0.66, 1.0, 1.5, 2.0, 3.0, 6.0, 9.0, 9.75, 10.33, 10.66, 11.0, 11.5, 12.0, 13.0, 14.0, 15.75, 16.33, 16.66, 17.0, 17.5, 18.0, 20.0, 22.0 and 24.0 hours (total blood draw; 25+ baseline).
The last blood draw in the pharmacokinetic profile includes enough blood to measure the clinical laboratory safety parameters required at the time of cessation (Close-out).
Visit 3 (day 8), stop visit
Subjects underwent the following evaluations:
routine physical examination, including vital signs (blood pressure, heart rate, respiratory rate, and body temperature).
Otolaryngological nasal examination.
Blood samples for complete blood cell count (hemoglobin, hematocrit, RBC, WBC and differential, MCV, MCHC) were collected.
Blood samples were collected for chemical profiles (Na/K, glucose, urea, creatine, calcium, phosphate, uric acid, total bilirubin, albumin, AST, ALT, ALP, GGT, and CK).
Blood samples for PSA.
Urine samples for dipstick urinalysis.
9.3 study population selection
Subjects were enrolled in the study according to the following inclusion/exclusion criteria:
9.3.1 inclusion criteria
High dose intranasal testosterone responder men in a Nasobol-01-2009 trial.
2. Written informed consent.
3. Males between the ages of 18 and 80.
4. A male having primary or secondary hypogonadism and having a serum testosterone level >150ng/dL and <300ng/dL in the morning (0900h + -30 min) in blood drawn under fasting conditions.
BMI in the range of 18.5-35kg/m2In the meantime.
6. All clinical laboratory assessments at the screening visit were from blood or collected urine drawn after an overnight fast (10 hours) and were within ± 15% of the clinical laboratory reference range, except serum testosterone.
7. Normal otorhinolaryngological nasal endoscopy. See appendix 16.1.1 for exclusion criteria for endoscopy.
8. Previous normal prostate examination (non-perceptible prostate mass) from the Nasobol-01-2009 test.
9. The PSA of the serum is less than or equal to 4.0 ng/mL.
9.3.2 exclusion criteria
1. Any type of significant intercurrent disease, in particular liver, kidney or heart diseases, diabetes or psychosis in any form.
2. Limitation of walking power, defined as walking two blocks on a horizontal surface or climbing level 10 difficulty
3. Hematocrit > 54% at screening.
4. History of cancer, excluding skin cancer.
5. History of nasal surgery, in particular turbidoplasty, septal plasty, rhinoplasty, "nasal augmentation" or sinus surgery.
6. A subject having an anterior nasal fracture.
7. Subjects with active allergies, such as rhinitis, rhinorrhea, and nasal congestion.
8. Subjects suffering from muco-inflammatory diseases, in particular pemphigus and Sjogren's syndrome.
9. A subject suffering from a paranasal sinus condition, in particular acute sinusitis, chronic sinusitis or fungal allergic rhinosinusitis.
10. A history of nasal disorders (e.g., polyposis, recurrent nasal bleeding (> 1 nosebleeds per month), abuse of nasal decongestants), or sleep apnea.
11. Subjects employing any form of intranasal drug delivery, specifically nasal corticosteroids and oxymetazoline, comprise a nasal spray (e.g., a drisan 12 hour nasal spray).
12. History of severe adverse drug reactions or leukopenia.
13. Abnormal bleeding tendency or history of thrombophlebitis not associated with venipuncture or intravenous intubation.
14. Hepatitis B, hepatitis C or HIV positive test.
15. History of asthma and ongoing asthma treatment.
16. History of sleep problems.
17. Smokers (10 cigarettes daily).
18. Regular drinkers who have more than four (4) units of alcohol per day (1 unit-300 ml beer, 1 glass of wine, 1 glass of spirits) or those who have difficulty giving up alcohol during the 48 hours preceding the 24 hour blood sampling visit.
19. History or current confirmation of alcohol abuse or any legitimate or illegitimate drug substance; or positive urine drug and alcohol screening for drugs of abuse and alcohol.
20. Current treatments are with androgens (e.g., dehydroepiandrostenedione, androstenedione) or anabolic steroids (e.g., testosterone, dihydrotestosterone).
21. Treatment with estrogen, GnRH antagonist or growth hormone during the first 12 months.
22. Treatment with drugs that interfere with testosterone metabolism, such as Anastrozole (Anastrozole), Clomiphene (Clomiphene), Dutasteride (Dutasteride), Finasteride (Finasteride), Flutamide (Flutamide), Ketoconazole (Ketoconazole), Spironolactone (Spironolactone), and Spironolactone (Testolactone).
23. Androgen therapy (intramuscular, topical, oral, etc.) over the past four weeks.
24. Poor compliance history or failure to maintain attending subjects.
25. Any other investigational study, except Nasobol-01-2009, was enrolled during the study or 30 days before the study began.
26. Blood donations (typically 550mL) were made at any time during the study and during the 12 week period before the study began.
9.3.3 removal of subjects from treatment or evaluation
The subject knows that he is free to leave the study at any time without giving the reason for his leaving and without consequences for his future medical care. It is queried to inform the direct researcher of his decision. Subject participation in the study may be discontinued based on any of the following reasons:
subject personal desire.
Apparently not subject to the study protocol and procedure.
Intercurrent disease, interfering with the progress of the study.
Intolerable adverse events, including clinically significant abnormal laboratory findings, where these hamper subject safety in the opinion of clinical researchers.
Clinical investigator's decision: departure from the study is the best benefit of the subject.
Clinical researchers have the right to terminate the study prematurely for safety reasons after having informed and negotiated with the sponsor. The sponsor has the authority to terminate the study prematurely if the clinical observations collected during the course of the study indicate that it may not be reasonable to continue or for other reasons (e.g., administrative, etc.) that are described based on the contract between the sponsor and the clinical site. But this is not necessary. There was no premature termination or discontinuation in the study.
9.4 treatment
9.4.1 administration of a drug
Subjects were selected centrally and randomly to the following treatment groups, such that the number in the groups was fairly balanced at three centers:
therapy a (n ═ 10): TBS-1 syringe pre-filled with 125 μ L of 4.0% gel, so that 5.0mg testosterone was delivered per nostril (intranasal) given at t.i.d. at 2100, 0700, and 1300. (Total dose 30 mg/day)
Therapy B (n ═ 10): TBS-1 syringe pre-filled with 150 μ L4.5% gel, so that 6.75mg testosterone was delivered per nostril (intranasal) for b.i.d. administration at 2100 and 0700. (Total dose 27.0 mg/day)
Therapy C (n ═ 10): TBS-1 syringe pre-filled with 125 μ L of 4.5% gel, delivering 5.625mg testosterone per nostril (intranasal) given at 2100, 0700, and 1300 at t.i.d. (Total dose 33.75 mg/day)
9.4.2 identity of research products
Name of drug: TBS-1 (pre-filled syringes to contain 5.0mg, 5.625mg and 6.75mg testosterone per syringe).
Pharmaceutical forms: the gel is administered nasally.
Content (wt.): active ingredients: testosterone.
Excipient: the amount of silica, castor oil,
modes of administration: nasally, in single doses to each nostril.
Manufacturer of the product:Haupt Pharma Amareg。
Batch number: 0744. 0942 and 0943.
Storage conditions: 20-25 deg.C.
Package with a metal layer
TBS-1 study drug was delivered to the clinical trial site in foil pouches using ready syringes (two syringes per pouch). Examples of syringe and pouch labels are described in appendix 4 of the protocol.
9.4.3 method of dispensing a subject for treatment
And (3) enabling the objects meeting the inclusion criteria to be divided into 1: 1: the 1 basis was randomly assigned to one of the three treatment groups. In screening, each object is assigned an object number by position in a sequential order. The object number consists of 5 digits. The first 2 numbers represent the site number assigned to the investigator, followed by a 3-digit object number. For example, 01-001 represents site (01) and first object (001).
The object number is used to identify the object throughout the study and is entered into the entire archive. The same object number is not assigned to more than one object.
9.4.4 dose selection in the study
In the previous study, Nasobol-01-2009, 4.0mg, 5.5mg, and 7.0mg testosterone were delivered intranasally using 3.2% testosterone gels using gel volumes of 125. mu.L, 172. mu.L, and 219. mu.L, respectively. In this study, 5.0mg, 5.65mg and 6.75mg testosterone were administered in gel volumes of 125 μ L, 125 μ L and 150 μ L, respectively. This study allowed the investigation of similar testosterone amounts delivered in smaller volumes.
9.4.5 dosage selection and timing for each subject
This is based on the results of previous studies.
9.4.6 cause blindness
Blindness does not exist because this is an open label study. A non-blinding explanation is that a quantitative rather than a qualitative analytical endpoint is measured and that it does not suffer from prejudices by the subject or the researcher.
9.4.7 Pre-and concomitant therapy
The following drugs were banned during the study:
the subject takes any form of intranasal drug delivery, specifically nasal corticosteroids and oxymetazoline, including nasal sprays s (e.g., drisan 12 hour nasal spray).
Current treatments are with androgens (e.g., dehydroepiandrostenedione, androstenedione) or anabolic steroids (e.g., testosterone, dihydrotestosterone).
Treatment with estrogen, GnRH antagonist or growth hormone occurs within the first 12 months.
Treatment with drugs that interfere with testosterone metabolism, e.g.; anastrozole, clomiphene, dutasteride, finasteride, flutamide, ketoconazole, spironolactone, and spironolactone.
Androgen therapy over the past four weeks (intramuscular, topical, oral, etc.).
9.4.8 treatment compliance
All medications were dispensed according to the protocol. It is the responsibility of the primary investigator to ensure that an accurate record of medication problems and returns is maintained. At the end of the study, the used original packaging was returned to the sponsor for destruction. Drug accountability was verified by monitors during the study and before the remaining study drug was destroyed.
During visit 2, subjects were given a week of bag supply; treatment A was 18 pockets, treatment B was 12 pockets, and treatment C was 18 pockets. Each pouch contained two syringes pre-filled with TBS-1 gel for treatment A, B or C. The subject was instructed how to administer the gel and was also given a diary to illustrate the time of administration at his home.
9.5 efficacy and safety variables
9.5.1 efficacy and safety measures of evaluation
The primary efficacy parameter was the AUC obtained within 24 hours after TBS-1 administration. Calculation of 24 hours C from AUCavg。
The area under the concentration curve (AUC) of b.i.d. and t.i.d. doses for the 0 to 24 hour time interval was determined using the trapezoidal rule.
The mean concentration (C) in the dosing interval was calculated from AUC using the following formulaavg):Cavg=AUC0-τAnd/τ, wherein τ is the dosing interval.
Calculate peak-to-valley fluctuation (PTF) and peak-to-valley steering (PTS) as follows:
o PTF=(Cmax-Cmin)/Cavg
o PTS=(Cmax-Cmin)/Cmin
·Cmin、Cmaxand tmaxTaken from actual measurements. Values are determined relative to the time of testosterone administration to the subject being treated.
24 hour C to calculate serum testosterone, DHT and estradiol concentrationsavgPercentage of objects whose values are above, within and below the respective reference range.
If desired, additional PK parameter exploratory analyses may have been performed.
Security data analysis
Polycythemia, anemia, and infection were monitored by measuring whole blood cell counts at screening and cessation of visits. The otorhinolaryngologist examines the subject and determines any clinically significant changes in the nasal mucosa following comparison to baseline.
Clinical chemistry and urinalysis tests, low or high blood glucose, renal function, liver function (hepatocellular or obstructive liver disease), skeletal/myocardial injury and changes in calcium homeostasis at screening visit 1 and at rest were evaluated.
Serum PSA was measured as a warning measure to measure possible changes in the prostate, although changes in prostate and serum PSA were not expected within a short treatment time frame.
Serum testosterone, dihydrotestosterone, and estradiol measurements at screening visit 1 and visit 3 allowed arbitrary overriding of two observed testosterone physiological products: deviation of the upper limits of the reference ranges for DHT and estradiol.
Security analysis was performed on all subjects receiving TBS-1. The occurrence of adverse events was indicated by treatment group, severity and relationship to study drug. All adverse events were described and evaluated with respect to causality and severity. Adverse events were classified using MedDRA. But it is rare and not drug related except for two.
Object security
Monitoring objects and emergency procedures: emergency medications, equipment and subject gurneys are available at the research center. In the "at home" phase, the subject has an emergency telephone number to be able to contact the clinical researcher.
Adverse events are defined as any adverse medical phenomenon in a subject or clinical trial subject to whom a drug product has been administered, and which may or may not have a causal relationship to this treatment. An adverse event may thus be any adverse or unintended sign, laboratory finding, symptom, or disease temporally associated with the use of the study drug product, whether or not it is considered in connection therewith. Any pre-existing condition in a clinical trial that worsens during clinical studies will be considered an adverse event.
Adverse reactions were defined as any adverse or unintended response to the study product associated with any administered dose. All adverse reactions judged by clinical researchers or sponsors to have a reasonable causal relationship with drug products were identified as adverse reactions. This is meant to generally indicate the presence of evidence or argument indicating a causal relationship.
Unexpected adverse reactions are defined as adverse reactions whose nature or severity does not fit the applicable product information.
A severe adverse event or severe adverse reaction is defined as any adverse medical phenomenon or outcome that at any dose results in death, life-threatening, requires hospitalization or prolongs existing subject hospitalization, results in persistent or overt disability or incapacity, or is congenital anomaly or congenital defect.
The observation period for the hypogonadal subjects ranged from when the subjects started studying the drug to when visit 3 ended. AEs continued at the end of the study period were followed until the investigator deemed the AE to reach a stable clinical endpoint or be resolved.
The percentage of subjects whose serum DHT and estradiol for each analyte are greater than the upper limit of the reference range.
Comparing day 8 arrest results to screening results and determining the following clinically significant changes:
o vital signs and adverse events: blood pressure, body temperature, respiration rate, heart rate.
o ear, nose, throat examination.
Complete blood cell count to assess changes in white blood cell count, hemoglobin, and hematocrit.
o clinical chemistry profile; Na/K, glucose, urea, creatine, calcium, phosphate, uric acid, total bilirubin, albumin, AST, ALT, ALP, GGT, CK, and PSA.
Classification:
o Severe Adverse Events (SAE) or severe adverse reactions: defined as any adverse medical phenomenon or outcome that, at any dose, results in death, is life-threatening, requires hospitalization of the subject or prolongs the existing subject's hospitalization, results in permanent or overt disability or incapacitation, is a congenital anomaly or congenital defect, is medically important, i.e., an AE endangers the subject or requires intervention to prevent one of the above consequences.
o non-severe AE: any AE that does not meet SAE standards.
o strength: adverse events/reactions were classified as mild, moderate or severe.
o causality: an adverse event can be considered an adverse reaction to the drug product under study-when a "reasonable causal relationship" exists between the event and the product under study. The following degrees of causality may be considered:
■ are absolutely in a reasonable time relationship with drug administration and withdrawal and reappear after drug restart.
■ are likely to be reasonably time-related to drug administration.
■ may be reasonably time-related to drug administration, but may be reasonably related to other factors.
■, it is not possible to have a reasonable time relationship with drug administration.
■ is unknown that there are insufficient components to correlate with drug intake.
■, failure to correlate with drug administration.
Procedure carried out in case of adverse events: all adverse events detected by clinical researchers were recorded in a special section of the event report. Any event classified as severe-regardless of cause and effect-will be reported to the CRO and initiator within 24 hours. No serious adverse events.
9.5.2 measurement appropriateness
All measurements used in this study are standard indicators of potency, PK and safety, and are generally considered reliable, accurate and relevant.
9.5.3. Primary efficacy variable(s)
The pharmacokinetic profile of serum testosterone in subjects dosed in treatments A, B and C had:
1.24 hours CavgValue of>300ng/dL and<1050ng/dL。
2.24 hours CavgThe percentage of subjects in each treatment group that are below, within, and above the reference range of serum testosterone from 300ng/dL to 1050ng/dL.
9.6 data quality assurance
The CRF project is verified by a monitor against the source data. All entries into the database include CRF and diary card object data, PK results and laboratory values. All data was reviewed 100% after entering the database of this report.
9.7 protocol planned statistical methods and sample size determination
9.7.1 statistical and analytical planning
The PK analysis plan is described above. After PK analysis, the vital signs and laboratory results analysis plan compares the baseline results with the final visit results. Other data, including demographic data, are descriptive. No statistical analysis was performed as the group size was not selected on a statistically significant basis.
9.7.2 sample sizing
These are sufficient for an acceptable description of the pharmacokinetic parameters for this population, based on the results obtained by conducting several pharmacokinetic studies in a panel of 10 subjects per group. Since this is a relatively modest phase II PK study aimed at examining two higher concentrations of TBS-1 gel, no actual sample size calculations were performed.
9.8 changes in study or planning analysis implementation
The protocol was modified at 7 months and 27 days 2010. The requested change is the blood draw timing. The number of blood draws remained the same. This change is needed to enable a full capture of the peak testosterone absorption after the third TID dose that occurred 1600 times after drug administration at 1300 th day 1300 or the first 2100 th day before (day 7).
10. Study object
10.1 object deployment
The study was performed in three centers located in Miami, FL, shriveport, LA and Tucson, AZ.
The three treatment groups were divided fairly at three sites. 8 subjects received treatment a and 7 subjects received treatments B and C, respectively. There were 22 subjects under study. Furthermore, 5 subjects participating in the previous clinical study were dropped and therefore not randomly selected to the study.
TABLE 10.1 deployment of Subjects by site and treatment
10.2 protocol bias
There were no significant pharmacokinetic deviations.
11. Pharmacokinetics and statistics
11.1 data set of analysis
The PK population was defined as subjects receiving treatment A, B or C and completing the study without major protocol violations or whose PK profile could be adequately characterized. The PK population was used to analyze PK data.
Based on the above criteria, the PK population included twenty-two (22) subjects. The number of subjects passing through the site and treated is shown below.
Table 11.1.1: subject deployment in PK population:
11.2 population and other baseline characteristics
Demographic data and characteristics are shown in table 11.2 by dose group for all subjects treated. No meaningful differences in any of the characteristics were observed in the three groups.
Table 11.2: demographic characteristics-overview of all objects
The mean age of the treated population for group a was 52.38, 53.86 for group B, and 51.57 for group C. The standard deviations were 12.55, 11.04, and 9.90, respectively. The ethnic and racial distributions of the groups are substantially the same.
11.3 treatment compliance measurement
Compliance with medication intake during the at-home portion of the study was determined by reviewing the diary and the returned pouches and syringes. Although the method is not absolute, it is sufficient to establish reasonable compliance. An object cannot find its diary.
11.4 pharmacokinetic and statistical results
11.4.1 method
Blood concentrations were received from ABL and electronically transferred from Trimel Biopharma SRL to the statistical unit of PharmaNet. Testosterone and dihydrotestosterone serum concentrations are provided in ng/mL. However, serum concentrations were converted to ng/dL for PK calculations to match literature reference range units.
During the course of the trial, clinical site 1 prescribes PK sampling on, say, the day after, starting on day 8 rather than day 7. This variation is not planned. Thus, the actual time for subjects at clinical site 1 was calculated relative to 2100 drug administrations on day 8, while subjects at clinical sites 2 and 3 were calculated relative to 2100h drug administrations on day 7.
For subject numbers 02-003, no medication time was recorded on day 7. Thus, the scheduled sampling time is used for PK calculations instead of the actual sampling time. 16.33h and 16.67h samples of subjects 01-001 were extracted at the same time for technical reasons. The planned sampling time was for sample 16.33h, while the actual sampling time was for sample 16.67 h.
Excluding the above exceptions, the time offset during sampling is handled as follows: for all sampling times, the difference between the planned and actual sampling times is considered acceptable if it is less than 1 minute. When the difference exceeds this time limit, the actual sampling time (rounded to three decimal places) is used to calculate the pharmacokinetic parameters, and the pre-dose samples are always reported as zero (0.000) apart from the pre-dose samples, regardless of time bias. The planned sampling times are shown in the concentration tables and graphs in the statistical report.
PK calculation Using WinNonlinTMVersion 5.2 (or higher) according to industry expectations and regulatory requirements. Descriptive statistical calculations also make use ofOffice Excel 2003.Office Excel2003 andoffice Word 2003 is used for reporting data tabulation.
Descriptive statistics (N, mean, Standard Deviation (SD), Coefficient of Variation (CV), median, minimum (Min.), and maximum (Min.)) of the serum concentration versus time for each treatment at each dose level and all pharmacokinetic parameters were provided using an evaluable population. All figures are shown using linear (a) and half-log (b) scales.
With respect to calculation of PK parameters from the last three drug administrations (treatments a and C: 0 hours to 10 hours, 10 hours and 16 hours and 24 hours; treatment B: 0 hours to 10 hours and 24 hours), serum concentration values of testosterone, dihydrotestosterone, and estradiol at time points 10 hours (before the second drug administration) and 16 hours (before the third drug administration under treatments a and C) were obtained by inputting serum concentration values observed at time points 9.75 hours and 15.75 hours, respectively.
The following pharmacokinetic parameters for testosterone, dihydrotestosterone and estradiol were determined for all subjects:
for treatments a and C (t.i.d.): AUC0-τ、AUC0-10、AUC10-16、AUC16-24、Cmax、Cmax 0-10、Cmax 10-16、Cmax 16-24、Cmin、Cmin 0-10、Cmin 10-16、Cmin 16-24、Cavg、Cavg 0-10、Cavg 10-16、Cavg16-24、tmax、tmax 0-10、tmax 10-16、tmax 16-24、tmax 10-24、PTF、PTS。
For treatment B (b.i.d.): AUC0-τ、AUC0-10、AUC10-24、Cmax、Cmax 0-10、Cmax 10-24、Cmin、Cmin 0-10、Cmin 10-24、Cavg、Cavg 0-10、Cavg 10-24、tmax、tmax 0-10、tmax 10-24、PTF、PTS。
In addition, C was calculated for serum testosterone, dihydrotestosterone and estradiol in each treatmentavgThe percentage of objects whose values are above, within and below their respective reference ranges. Furthermore, serum testosterone, dihydrotestosterone and estradiol values in the respective treatments are provided in respective referencesAverage time percentages above (within%) range, within (within%) range, and below (within%) range. The calculation of all these pharmacokinetic parameters is explained below.
11.4.1.1 maximum and minimum observed concentrations and time to peak concentration observed
For each subject and each treatment, C was determined as followsmaxMaximum observed concentration, and TmaxTime to peak concentration, and CminThe minimum observed concentration was:
Cmax: maximum observed concentration during the dosing interval. This parameter was calculated for treatments A, B and C.
Cmax 0-10: time zero to 10 hours of maximum observed concentration. This parameter was calculated for treatments A, B and C.
Cmax 10-16: time 10 hours to 16 hours of maximum observed concentration. This parameter was calculated for treatments a and C.
Cmax 16-24: time 16 hours to 24 hours of maximum observed concentration. This parameter was calculated for treatments a and C.
Cmax 10-24: time 10 hours to 24 hours of maximum observed concentration. This parameter was calculated only for treatment B.
Cmin: minimum observed concentration during dosing interval. This parameter was calculated for treatments A, B and C.
Cmin 0-10: time zero to 10 hours minimum observed concentration. This parameter was calculated for treatments A, B and C.
Cmin 10-16: time 10 hours to 16 hours minimum observed concentration. This parameter was calculated for treatments a and C.
Cmin 16-24: time 16 hours to 24 hours minimum observed concentration. This parameter was calculated for treatments a and C.
Cmin 10-24: time 10 hours to 24 hours minimum observed concentration. This parameter was calculated only for treatment B.
tmax: c observed during dosing intervalsmaxTime of (d). This parameter was calculated for treatments A, B and C.
tmax 0-10: c was observed from time zero to 10 hoursmaxTime of (d). This parameter was calculated for treatments A, B and C.
tmaxC was observed over a period of 10 to 16 hoursmaxTime of (d). This parameter was calculated for treatments a and C.
10-16:
tmaxC was observed over a period of 16 hours to 24 hoursmaxTime of (d). This parameter was calculated for treatments a and C.
16-24:
tmaxC was observed over a period of 10 hours to 24 hoursmaxTime of (d). This parameter was calculated only for treatment B.
10-24:
11.4.1.2 area under concentration-time curve
AUC calculation was performed using a linear trapezoidal method.
AUC0-τDose time (0) to dosing time □ was calculated (□ ═ 24 h). However, in the case of 24-h sample collection with time bias, the AUC was estimated based on the concentration estimated at 24 hours using the regression line calculated from the elimination phase instead of the concentration at the actual observation time0-τ。
In the case where the final concentration values (Y) are missing or do not correspond to the planned sampling times (i.e. 10 hours and 16 hours), as can be calculated, the AUC is inferred using the elimination phase of the corresponding subjectX-Y。
The following AUC was calculated:
AUC0-τ: area under the concentration-time curve for one dosing interval. This parameter was calculated for treatments A, B and C.
AUC0-10: area under the concentration-time curve from time zero to 10 hours. This parameter was calculated for treatments A, B and C.
AUC10-16: area under the concentration-time curve from time 10 hours to 16 hours. This parameter was calculated for treatments a and C.
AUC16-24: area under the concentration-time curve from time 16 hours to 24 hours. This parameter was calculated for treatments a and C.
AUC10-24: area under the concentration-time curve from time 10 hours to 24 hours. This parameter was calculated only for treatment B. Calculate C as followsavg:
Cavg: the mean concentration during the dosing interval was calculated as AUC0- τ/τ (τ: 24 hours). This parameter was calculated for treatments A, B and C.
Cavg 0-10: mean concentrations from time zero to 10 hours, calculated as AUC 0-10/10. This parameter was calculated for treatments A, B and C.
Cavg 10-16: mean concentrations over time from 10 hours to 16 hours, calculated as AUC 10-16/6. This parameter was calculated for treatments a and C.
Cavg 16-24: mean concentrations over time 16 hours to 24 hours, calculated as AUC 16-24/8. This parameter was calculated for treatments a and C.
Cavg 10-24: mean concentrations over time from 10 hours to 24 hours, calculated as AUC 10-24/14. This parameter was calculated only for treatment B.
11.4.1.3 mean drug concentration
Calculate C as followsavg:
Cavg: during intervals between dosesThe mean concentration was calculated as AUC0- τ/τ (τ.24 hours). This parameter was calculated for treatments A, B and C.
Cavg 0-10: mean concentrations from time zero to 10 hours, calculated as AUC 0-10/10. This parameter was calculated for treatments A, B and C.
Cavg 10-16: mean concentrations over time from 10 hours to 16 hours, calculated as AUC 10-16/6. This parameter was calculated for treatments a and C.
Cavg 16-24: mean concentrations over time 16 hours to 24 hours, calculated as AUC 16-24/8. This parameter was calculated for treatments a and C.
Cavg 10-24: mean concentrations over time from 10 hours to 24 hours, calculated as AUC 10-24/14. This parameter was calculated only for treatment B.
11.4.1.4 peak-to-valley fluctuation and peak-to-valley steering
Peak-to-valley fluctuation (PTF) and peak-to-valley steering are calculated as follows:
PTF: fluctuating peak and valley, press (C)max-Cmin)/CavgAnd (4) calculating. This parameter was calculated for treatments A, B and C.
PTS: turning direction from peak to valley, press (C)max-Cmin)/CminAnd (4) calculating. This parameter was calculated for treatments A, B and C.
11.4.1.5 percent of time above, within, and below a reference range and CavgPercentage of objects above, within and below the reference range
The percentage of time over which the observed results for serum testosterone, dihydrotestosterone and estradiol for each subject and treatment fell above (above% time), within (within% time) and below (below% time) the reference range was calculated. For each treatment, C was calculated for serum testosterone, dihydrotestosterone and estradiolavgThe percentage of objects whose values are above, within and below their respective reference ranges. The reference range is testosterone 300ng/dL to 1050ng/dL, dihydrotestosteroneKetones 25.5ng/dL to 97.8ng/dL and estradiol 3pg/mL to 81pg/mL.
PTS: turning direction from peak to valley, press (C)max-Cmin)/CminAnd (4) calculating. This parameter was calculated for treatments A, B and C.
11.4.1.6 statistical analysis
Only descriptive statistics (N, mean, SD, CV, median, min, and Max.) were calculated for the serum concentration and PK parameters for each treatment. Inferential statistical analysis was not performed.
11.4.2 analysis and statistical problems of pharmacokinetics
11.4.2.2 handling of missed data
The sample not analyzed due to insufficient volume (see bioassay report) was recorded in the concentration table as INV (analysis volume insufficient).
These samples were set as missed for pharmacokinetic and statistical analysis. Since PK parameters can be estimated using the remaining data points, subjects missing data are retained in the pharmacokinetic analysis.
11.4.2.3 pharmacokinetic analysis
The following pharmacokinetic parameters of testosterone, dihydrotestosterone and estradiol were determined for all subjects:
for treatments a and C (t.i.d.): AUC0-τ、AUC0-10、AUC10-16、AUC16-24、Cmax、Cmax 0-10、Cmax 10-16、Cmax 16-24、Cmin、Cmin 0-10、Cmin 10-16、Cmin 16-24、Cavg、Cavg 0-10、Cavg 10-16、Cavg16-24、tmax、tmax 0-10、tmax 10-16、tmax 16-24、tmax 10-24、PTF、PTS。
For treatment B (b.i.d.): AUC0-τ、AUC0-10、AUC10-24、Cmax、Cmax 0-10、Cmax 10-24、Cmin、Cmin 0-10、Cmin 10-24、Cavg、Cavg 0-10、Cavg 10-24、tmax、tmax 0-10、tmax 10-24、PTF、PTS。
In addition, the C of serum testosterone, dihydrotestosterone and estradiol was calculated for each treatmentavgThe percentage of objects whose values are above, within and below their respective reference ranges. Furthermore, mean time percentages are provided for each treatment for serum testosterone, dihydrotestosterone and estradiol values above (within% time), within (within% time) and below (below% time) the respective reference ranges. The calculation of all these pharmacokinetic parameters is explained below.
All tables and figures referred to in this section are shown in sections 14.2.1 and 14.2.2, respectively, with the exception of the text tables (numbered 11.4.2.3-1 through 11.4.2.3-3) and text figures (numbered 11.4.2.3-1 through 11.4.2.3-3). For simplicity, TBS-1 treatment was identified in the text of the statistical report by its treatment code: a (125. mu.L of 4% gel, t.i.d. administration, total dose 30 mg/day), B (150. mu.L of 4.5% gel, b.i.d. administration, total dose 27.0 mg/day), and C (125. mu.L of 4.5% gel, t.i.d. administration, total dose 33.75 mg/day).
Pharmacokinetic analyzed blood samples for treatments a and C were collected before and after drug administration at 2100 d 7 at hours 0.333, 0.667, 1.00, 1.50, 2.00, 3.00, 6.00, 9.00, 9.75, 10.33, 10.66, 11.0, 11.5, 12.0, 13.0, 14.0, 15.75, 16.33, 16.66, 17.0, 17.5, 18.0, 20.0, 22.0, and 24.0. The pharmacokinetic analyzed blood samples for treatment B were collected at 0.333, 0.667, 1.00, 1.50, 2.00, 3.00, 6.00, 9.00, 9.75, 10.33, 10.66, 11.0, 11.5, 12.0, 13.0, 16.0, 19.0, 22.0, and 24.0 hours before and after drug administration at day 2100. The actual sampling times for PK calculations for treatments A, B and C are shown in tables 14.2.1.22, 14.2.1.23 and 14.2.1.24, respectively.
Testosterone
The serum concentration of testosterone measured at each sampling time for each subject is shown in tables 14.2.1.1, 14.2.1.2, and 14.2.1.3 according to treatment. Plots of individual serum levels over the sampling period are shown in figures 14.2.2.1 to 14.2.2.22 using linear (a) and half log (b) scales. Lines of minimum (300ng/dL) and maximum (1050ng/dL) limits of the reference range of testosterone serum concentrations are also shown for informational purposes. Furthermore, the mean drug concentration (C) during the dosing interval (τ 24 hours) is also shown on each profileavg) The line of (2).
The plot of mean serum levels over the sampling period is also shown in plots 14.2.2.23, 14.2.2.24 and 14.2.2.25 for treatments A, B and C, respectively, using linear (a) and half log (b) scales. The error bars on these mean profiles correspond to one standard deviation. The mean graph also shows the lines of the minimum and maximum limits of the reference range.
A plot of the mean values of the linear scale for each treatment is also shown in the lower panel 11.4.2.3-1.
FIG. 11.4.2.3-1: mean testosterone serum concentration (ng/dL) -time curves for each treatment
The calculated pharmacokinetic parameters for each subject for treatment A, B and C, based on treatment, are shown in tables 14.2.1.4, 14.2.1.5 and 14.2.1.6, respectively. This is summarized in text table 11.4.2.3-1.
TABLE 11.4.2.3-1: summary of testosterone pharmacokinetic parameters for each treatment
TABLE 11.4.2.3-1: summary of testosterone pharmacokinetic parameters for each treatment
Reference range 300 + 1050ng/dL.
TBS-1, 125 μ L4.0% gel, t.i.d. (total dose 30 mg/day)
TBS-1, 150 μ L4.5% gel, b.i.d. (total dose 27.0 mg/day)
TBS-1, 125 μ L4.5% gel, t.i.d. administration (total dose 33.75 mg/day)
The percentage of time over which the observation falls above (within%) the reference range, within (within%) the reference range, and below (within%) the reference range was calculated for each subject and is shown in tables 14.2.1.4, 14.2.1.5, and 14.2.1.6 for treatments A, B and C, respectively. These results are also summarized in text table 11.4.2.3.1.
Serum testosterone C was calculated for each treatmentavgThe percentages of objects having values above, within, and below the reference range are shown in table 14.2.1.7. These results are also summarized in text table 11.4.2.3.1.
Dihydrotestosterone
The serum concentrations of dihydrotestosterone measured at each sampling time for each subject according to treatment are shown in tables 14.2.1.8, 14.2.1.9 and 14.2.1.10. Plots of individual serum levels over the sampling period are shown in figures 14.2.2.26 to 14.2.2.47 using linear (a) and half log (b) scales. Lines with reference ranges for dihydrotestosterone serum concentrations at minimum (25.5ng/dL) and maximum (97.8ng/dL) limits are also shown for informational purposes. Furthermore, the mean drug concentration (C) during the dosing interval (τ 24 hours) is also shown on each profileavg) The line of (2).
Plots of mean serum levels against treatments A, B and C versus sampling period are also shown in figures 14.2.2.48, 14.2.2.49 and 14.2.2.50, respectively, using linear (a) and half log (b) scales. The error bars on these mean profiles correspond to one standard deviation. The mean graph also shows the lines of the minimum and maximum limits of the reference range.
Plots of the linear scale mean values for each treatment are also shown in figure 11.4.2.3-2 below.
FIG. 11.4.2.3-2: mean dihydrotestosterone serum concentration (ng/dL) -time profile for each treatment
According to SAP, when the final concentration (Y) does not correspond to the planned sampling time, AUC is calculated based on the estimated concentration (Y) using a regression line calculated by eliminating the phase dataX-Y. For subject numbers 01-002 and 02-007, the elimination phase was not clearly characterized due to fluctuations in dihydrotestosterone serum concentrations at 10 to 16 hour and 0 to 10 hour intervals, respectively. Thus, AUC for subject numbers 01-002 for treatment A could not be calculated10-16And Cavg 10-16(derived from AUC)10-16) (these parameters N ═ 7). Furthermore, AUC for subject numbers 02-007 of treatment A could not be calculated0-10And Cavg 0-10(derived from AUC)0-10) (these parameters N ═ 7).
Pharmacokinetic parameters calculated from treatment for each subject for treatments A, B and C are shown in tables 14.2.1.11, 14.2.1.12, and 14.2.1.13, respectively. Which is summarized in text table 11.4.2.3-2.
TABLE 11.4.2.3-2: summary of dihydrotestosterone pharmacokinetic parameters for each treatment
TABLE 11.4.2.3-2: summary of dihydrotestosterone pharmacokinetic parameters for each treatment
The percentage of time at which the observation falls above (within%) the reference range, within (within%) the reference range, and below (within%) the reference range was calculated for each subject, and treatments A, B and C are shown in tables 14.2.1.11, 14.2.1.12, and 14.2.1.13, respectively. These results are also summarized in text table 11.4.2.3.2.
Calculation of serum dihydrotestosterone C for each treatmentavgThe percentages of objects having values above, within, and below the reference range are shown in table 14.2.1.14. These results are also summarized in text table 11.4.2.3.2.
Estradiol
Serum concentrations of estradiol measured at each sampling time according to treatment of each subject are shown in tables 14.2.1.15, 14.2.1.16, and 14.2.1.17. Plots of individual serum levels over the sampling period are shown in figures 14.2.2.51 to 14.2.2.72 using linear (a) and half log (b) scales. Lines of minimum (3pg/mL) and maximum (81pg/mL) limits of the reference range for estradiol serum concentrations are also shown for informational purposes. Also, the average drug concentration (C) during the dosing interval (τ 24 hours) is shown in each graphavg) The line of (2).
Plots of mean serum levels of treatments A, B and C versus sampling period are also shown in figures 14.2.2.73, 14.2.2.74 and 14.2.2.75, respectively, using linear (a) and half log (b) scales. The error bars on these averaged plots correspond to one standard deviation. The mean graph also shows the lines of the minimum and maximum limits of the reference range.
Plots of the linear scale average values for each treatment are also shown in figure 11.4.2.3-3 below.
FIG. 11.4.2.3-3: mean estradiol serum concentration (pg/mL) versus time curve for each treatment
According to SAP (section 8.3), when the final concentration (Y) does not correspond to the planned sampling time, AUC is calculated based on the concentration (Y) estimated using the regression line calculated by eliminating the phase dataX-Y. However, for some subjects, the elimination phase was not clearly characterized due to fluctuations in estradiol serum concentration, as follows:
object number: 02-007, treatment A at 0 to 10 hour and 0 to 24 hour intervals. The following PK parameters could not be calculated for this subject for treatment a: AUC0-10、Cavg 0-10、AUC0-τ、CavgAnd PTF (these parameters N ═ 7).
Object number: 01-002 and 01-007, 10 to 16 hour intervals for treatment A. AUC of these subjects for treatment A could not be calculated10-16And Cavg 10-16(these parameters N ═ 6).
Object number: 02-004 and 02-007, 16 to 24 hour intervals for treatment A. AUC of this subject unable to calculate treatment A16-24And Cavg 16-24(these parameters N ═ 6).
Object number: 02-003 and 02-005, at 0 to 10 hour intervals for treatment of C. AUC of these subjects unable to calculate treatment C0-10And Cavg 0-10(these parameters N ═ 5).
Pharmacokinetic parameters calculated from treatment for each subject for treatments A, B and C are shown in tables 14.2.1.18, 14.2.1.19, and 14.2.1.20, respectively. Which is summarized in text table 11.4.2.3-3.
TABLE 11.4.2.3-3: summary of estradiol pharmacokinetic parameters for each treatment
TABLE 11.4.2.3-3: summary of estradiol pharmacokinetic parameters for each treatment
TABLE 11.4.2.3-3: summary of estradiol pharmacokinetic parameters for each treatment
Reference range 3-81pg/mL.
TBS-1, 125 μ L4.0% gel, t.i.d. (total dose 30 mg/day)
TBS-1, 150 μ L4.5% gel, b.i.d. (total dose 27.0 mg/day)
TBS-1, 125 μ L4.5% gel, t.i.d. administration (total dose 33.75 mg/day)
With respect to these parameters, treatment a is N-7.
With respect to these parameters, treatment a is N-6.
With respect to these parameters, treatment C is N-5.
The percentage of time at which the observation falls above (within%) the reference range, within (within%) the reference range, and below (within%) the reference range was calculated for each subject, and treatments A, B and C are shown in tables 14.2.1.18, 14.2.1.19, and 14.2.1.20, respectively. These results are also summarized in text table 11.4.2.3.3.
Calculation of serum estradiol C for each treatmentavgThe percentages of objects having values above, within, and below the reference range are shown in table 14.2.1.21. These results are also summarized in text table 11.4.2.3.3.
11.4.2.4 pharmacokinetic analysis
Pharmacokinetic analyses were not planned or performed during this study.
11.4.7 pharmacokinetic and statistical conclusions
In this phase II study, subjects were randomly selected into three treatment groups (4.0% TBS-1 given t.i.d., and 4.5% TBS-1 given bid and t.i.d.). Treatment was given in a parallel design by the intranasal route for one week. At the end of one week, the three treatments were compared by performing a 24 hour pharmacokinetic study of systemic absorption of the drug product testosterone and its two physiological metabolites dihydrotestosterone and estradiol.
Testosterone
TBS-1 pharmacokinetic profiles after single and repeated doses were examined in 2 prior studies (TST-PKP-01-MAT/04 and TST-DF-02-MAT/05). These studies demonstrated that testosterone is well absorbed following intranasal administration. The maximum serum concentration is reached after 1-2 hours post-administration. In the current study, testosterone formulations (4.0% TBS-1 given t.i.d. and 4.5% TBS-1 given bid and t.i.d.) were rapidly absorbed and the peak concentrations ranged from 36 minutes to 1 hour 6 minutes (mean t.i.d.) following intranasal administrationmax) Internal reaching. The maximum testosterone concentration during the 24 hour interval was observed in about 57% to 71% of hypogonadal men during the first dose (0-10 hours), while about 29% to 43% of subjects had their maximum 24-h testosterone concentration during the subsequent dose.
When comparing t.i.d. treated TBS-1 dosing alone, although the mean AUC between formulations was similar, a larger AUC was observed after the first dose compared to the two subsequent doses (AUC for treatments a and C, respectively)0-10: 4178.68 and 4355.19h ng/dL>AUC10-16: 2635.05 and 2301.51h ng/dL<AUC16-24: 3016.52 and 2766.97h ng/dL). A greater AUC (AUC) is observed for the second dose of treatment B compared to the first dose0-10:4451.64h*ng/dL~AUC10-24: 5264.19h ng/dL). the differences in AUC between the t.i.d. and b.i.d. formulations administration may be due to the passage of different time periods between each administration. Mean AUC calculated over a 24 hour dosing interval0-τEquivalence between all treatments (AUC for treatments A, B and C, respectively)0-τ: 9920.07, 9781.39 and 9505.03h ng/dL).
Although mean C between treatments A and CmaxSimilar, but C was observedmaxTrend decreasing with subsequent dosing (treatments A and C are C, respectively)max 0-10: 786 and 857ng/dL>Cmax 10-16: 698 and 675ng/dL>Cmax 16-24: 556 and 595 ng/dL). Comparable mean testosterone C was observed for two doses of treatment Bmax(Cmax 0-10:894ng/dL~Cmax 10-24: 846 ng/dL). C between administration of t.i.d. formulationsmaxThe difference may be due to the different time periods that elapse between each administration. Treatment B (150 μ L4.5% gel (b.i.d.)) (C)max: 1050ng/dL) of the drug in the drug delivery solution during the 24 hour dosing intervalmaxAnd treatment A and C (C, respectively)max: 830 and 883 ng/dL). 1 of 8 subjects treated A and 3 of 7 subjects treated B and C exceeded the upper limit of the physiological reference range (1050 ng/dL).
When comparing the administration of t.i.d. and b.i.d. treatments separately, C was observedavgSlightly reduced trend (treatments A and C are C, respectively)avg 0-10: 418 and 436ng/dL>Cavg 10-16: 439 and 384ng/dL>Cavg 16-24: 377 and 346ng/dL, and treatment B is Cavg 0-10:445ng/dL>Cavg 10-24: 376 ng/dL). C between administrationsavgThe difference may be due to the different time periods that elapse between each administration. Mean C calculated during the 24 hour dosing interval for all treatmentsavgComparable (treatment A, B and C are C, respectively)avg:413,408,396ng/dL)。
These results indicate the exposure between each dose (AUC, C) of t.i.d. dosing (treatment a and C)avgAnd Cmax) Decrease, but not b.i.d. administration (treatment B). this reduction in exposure to t.i.d. administration can be explained in part by the negative feedback of endogenous testosterone production from the HPG axis. In other words, since the time interval between each administration of the t.i.d. group is small,the recovery of the HPG system from negative feedback will be less than the b.i.d. group.
Regardless of formulation, the mean drug concentration (C) in about 86% -88% of subjectsavg) C in 13% -14% of subjects within a physiological reference range (300 to 1050ng/dL)avgC below the reference range and without objectavgAbove the reference range.
The time periods during one day (24 hours) where serum testosterone concentrations were below, within and above the physiological reference range for all formulations covered 30 to 35%, 59% to 68% and 0% of the 24 hour period, respectively. That is, testosterone levels are within the normal range from about 14 to 16 hours a day.
Dihydrotestosterone
TBS-1 dosing was followed by 1 hour 24 minutes and 2 hours 23 minutes (mean T)max) The peak concentration of dihydrotestosterone is reached.
When comparing t.i.d. treated TBS-1 dosing alone, although the mean AUC between formulations was similar, a trend was observed for AUC to decrease with subsequent dosing (AUC for treatments A and C, respectively)0-10: 345.77 and 411.10h ng/dL>AUC10-16: 186.33 and 222.62h ng/dL>AUC16-24: 269.16 and 275.21h ng/dL). Comparable AUC (AUC) was observed for both doses of treatment B0-10:402.77h*ng/dL~AUC10-24: 543.29h ng/dL). the differences in AUC between t.i.d. formulation dosing may be due to the passage of different time periods between each dosing. Mean AUC calculated during the 24 hour dosing interval between all treatments0-τAre comparable (AUC for treatments A, B and C, respectively0-τ: 818.95, 946.89 and 909.68h ng/dL).
Although the average C between t.i.d. formulationsmaxSimilarly, C was observedmaxTrend decreasing with subsequent dosing (treatments A and C are C, respectively)max 0-10: 51.4 and 59.0ng/dL>Cmax 10-16: 44.2 and 48.9ng/dL>Cmax 16-24: 41.3 and 42.6 ng/dL). Equivalent mean testosterone C was observed for two doses of treatment Bmax(Cmax 0-10:56.8ng/dL~Cmax 10-24: 54.6 ng/dL). C between administration of t.i.d. formulationsmaxThe difference may be due to the different time periods that elapse between each administration. Mean C calculated during the 24 hour dosing interval for all treatmentsmaxAre comparable (treatments A, B and C are C, respectivelymax: 52.2, 61.0 and 60.3 ng/dL). Any subject treated arbitrarily did not exceed the physiological reference range upper limit (97.8 ng/dL).
C between treatments and dosing by dosingavgAre comparable (treatments A and C are C, respectively)avg 0-10: 34.6 and 41.1ng/dL>Cavg 10-16: 31.1 and 37.1ng/dL>Cavg 16-24: 33.6 and 34.4ng/dL, and treatment B is Cavg 0-10:40.3ng/dL>Cavg 10-24: 38.8 ng/dL). Mean C calculated during the 24 hour dosing interval for all treatmentsavgAre comparable (treatments A, B and C are C, respectivelyavg:34.1,39.5,37.9ng/dL)。
Treatment of C in about 63% of subjects after A administrationavgIncluded in the DHT physiological reference range (25.5 to 97.8ng/dL), and this number rose to about 86% when treatments B and C were administered. Object-free CavgC in subjects above the normal range, while treatment A and treatments B and C are 38% and 14%, respectivelyavgBelow the normal range.
The time periods during the day (24 hours) in which serum DHT concentrations were below, within and above the physiological reference range covered 32.64%, 67.36% and 0% treatment a, 26.22%, 73.78% and 0% treatment B, and 13.87%, 86.13% and 0% treatment C, respectively. That is, DHT levels for treatments A, B and C were within the normal range at approximately 16, 18, and 21 hours a day, respectively.
Estradiol
TBS-1 dosing was followed by 1 hour 12 minutes and 2 hours 41 minutes (mean T)max) The peak concentration of the estradiol is reached.
When comparing t.i.d. treated TBS-1 dosing alone, although mean AUC between formulations was similar, it was observedTrend of AUC decreasing with subsequent dosing (AUC for treatments A and C, respectively)0-10: 234.96 and 267.78h pg/mL>AUC10-16: 144.76 and 144.30h pg/mL<AUC16-24: 153.02 and 177.97h pg/mL). Comparable AUC (AUC) was observed for both doses of treatment B0-10:242.02h*pg/mL~AUC10-24: 295.12h pg/mL). the differences in AUC between t.i.d. formulation dosing may be due to the passage of different time periods between each dosing. Mean AUC calculated during the 24 hour dosing interval between all treatments0-τAre comparable (AUC for treatments A, B and C, respectively0-τ: 530.27, 537.16 and 601.91h pg/mL).
Although the average C between t.i.d. formulationsmaxSimilar, but C was observedmaxTrend decreasing with subsequent dosing (treatments A and C are C, respectively)max 0-10: 36.8 and 35.5pg/mL>Cmax 10-16: 28.9 and 31.5pg/mL>Cmax 16-24: 27.2 and 26.9 pg/mL). Equivalent mean testosterone C was observed for two doses of treatment Bmax(Cmax 0-10:35.8pg/mL~Cmax 10-24: 30.6 pg/mL). C between administration of t.i.d. formulationsmaxThe difference may be due to the different time periods that elapse between each administration. Mean C calculated during the 24 hour dosing interval for all treatmentsmaxAre comparable (treatments A, B and C are C, respectivelymax: 37.9, 36.2, and 36.4 pg/mL). Any subject treated arbitrarily did not exceed the physiological reference range upper limit (81 pg/mL).
C between treatments and dosing by dosingavgAre comparable (treatments A and C are C, respectively)avg 0-10: 23.5 and 26.8pg/mL>Cavg 10-16: 24.1 and 24.0pg/mL>Cavg 16-24: 19.1 and 22.2pg/mL, and treatment B is Cavg 0-10:24.2pg/mL>Cavg 10-24: 21.1 pg/mL). Mean C calculated during the 24 hour dosing interval for all treatmentsavgAre comparable (treatments A, B and C are C, respectivelyavg:22.1、22.4、25.1pg/mL)。
Total treatment of C in all subjects after dosingavgIs included in E2Physiology of human bodyWithin the reference range (3 to 81 pg/mL). E of all objects2The concentration was within the normal range over a 24 hour period. Without object E2Levels were below or above the normal range at any time of the day.
12. Evaluation of safety
12.1 degree of Exposure
Two sites of subjects were dosed with drug for 7 days and the other site of subjects were dosed with drug for 8 days.
12.2 adverse events
12.2.1 brief summary of adverse events
6 subjects had 8 adverse events. Of which 6 events occurred during treatment a and 2 occurred during treatment B. Subjects 01-002 and 01-007 all felt dizziness and were both indicated to be likely related to study medication. Subjects 01-002 were of moderate severity, which resolved after 5 days. 7 of the 8 adverse events were mild. 6 of the 8 events were unrelated to study drug.
Subject 02-004 was classified by the investigator as having anemia. Hemoglobin is at the lowest normal level and is considered drug independent. Table 12.2.2 summarizes the events.
12.2.2 adverse event display
Table 12.2.2: adverse events
12.2.4 adverse event enumeration of subjects
Table 12.2.2 lists adverse events for the subjects.
12.3 mortality, other serious adverse events and other significant adverse events
There were no deaths, other serious adverse events, or other significant adverse events during this study.
12.4.2 evaluation of laboratory parameters
The primary investigator determined that laboratory values did not vary clinically significantly from the beginning to the end of the study. All objects do not have some outliers at the initial visit and/or the third visit. There is no consistent change for all visits.
Uric acid levels of subjects 01-007 at visit three were 539U/L, with 289 being the upper limit of normal. About half of the subjects had elevated glucose values compared to the normal first visit values. This extends over all three doses and is only slightly elevated. Has no clinical significance.
12.5 Vital signs, body results and other safety-related observations
There was no meaningful or significant change in vital signs after test drug administration.
12.6 safety conclusions
In this and other studies, TBS-1 gel demonstrated that it was safe to use. There were no serious adverse events or any consequential events during this PK study or during the 7 days of self-administration. Tables 14.3.2.1 through 14.3.2.8 show all laboratory values for visit 1 and visit 3.
13. Discussion and general conclusions
The first objective of this study was to determine the bioavailability of 4.0% TBS-1 gel (t.i.d. administration) and 4.5% TBS-1 gel (b.i.d. and t.i.d. administration) in hypogonadal men.
In previous studies, Nasobol-01-2009, 3.2% testosterone gel, was applied, delivering 4.0mg, 5.5mg and 7.0mg testosterone intranasally with gel volumes of 125. mu.L, 172. mu.L and 219. mu.L, respectively. In this study, 5.0mg, 5.65mg and 6.75mg testosterone were administered in gel volumes of 125 μ L, 125 μ L and 150 μ L, respectively. This study allowed the investigation of similar testosterone amounts delivered in much smaller volumes.
The second objective of this study was to establish a security profile for TBS-1. In this phase II study, subjects were randomly selected into three treatment groups (4.0% TBS-1, given by t.i.d., and 4.5% TBS-1, given by bid, and t.i.d.). Treatment was given in a parallel design by the intranasal route for one week. At the end of one week, the three treatments were compared by performing a 24-hour pharmacokinetic study of systemic absorption of the drug product testosterone and its two physiological metabolites dihydrotestosterone and estradiol.
6 subjects described 8 adverse events. Of which 6 events occurred during treatment a and 2 occurred during treatment B. Subjects 01-002 and 01-007 all felt dizziness and were both indicated to be likely related to study medication. The remainder was independent of study drug.
No significant or meaningful vital signs or laboratory changes. No polycythemia, anemia, or infection was observed after the complete blood count measurements at screening and cessation. Clinical chemistry and urinalysis showed no change at rest as follows: low or high blood glucose, renal function, liver function, skeletal/cardiac muscle damage, or changes in calcium homeostasis. The nasal mucosa has no clinically significant changes.
The PK population was defined as subjects receiving treatment A, B or C and completing the study without major protocol violations or with a PK profile that could be adequately characterized. The PK population was used for PK data analysis. Based on these criteria, the PK population included twenty-two (22) subjects.
Testosterone
The pharmacokinetic profile of TBS-1 after single and repeated administrations was examined in 2 prior studies (TST-PKP-01-MAT/04 and TST-DF-02-MAT/05). These studies demonstrated that testosterone is well absorbed following intranasal administration. The highest serum concentration was reached after 1-2 hours post-dose. In the current study, testosterone formulations (4.0% TBS-1, administered by t.i.d., and 4.5% TBS-1, administered by bid and t.i.d.) were rapidly absorbed with peak concentrations ranging from 36 minutes to 1 hour 6 minutes after intranasal administration (mean t.i.d.)max) And (4) achieving the purpose. Approximately 57% to 71% of hypogonadal men observed in men during the first dose (0-10 hours) were observed over a 24 hour periodMaximum testosterone concentration during the interval, while maximum 24-h testosterone concentration in about 29% to 43% of subjects is during subsequent dosing.
When comparing t.i.d. treated TBS-1 dosing alone, although the mean AUC between formulations was similar, a greater AUC was observed after the first dose compared to the two subsequent doses (AUC for treatments A and C, respectively)0-10: 4178.68 and 4355.19h ng/dL>AUC10-16: 2635.05 and 2301.51h ng/dL<AUC16-24: 3016.52 and 2766.97h ng/dL). With respect to treatment B, a greater AUC (AUC) was observed for the second dose compared to the first dose0-10:4451.64h*ng/dL~AUC10-24: 5264.19h ng/dL). the differences in AUC between the administration of the t.i.d. and b.i.d. formulations may be due to the passage of different time periods between each administration. Mean AUC calculated during the 24 hour dosing interval between all treatments0-tAre comparable (AUC for treatments A, B and C, respectively0-t: 9920.07, 9781.39 and 9505.03h ng/dL).
When comparing t.i.d. treated TBS-1 dosing alone, although mean C between formulationsmaxSimilar, but C was observedmaxTrend decreasing with subsequent dosing (treatments A and C are C, respectively)max 0-10: 786 and 857ng/dL>Cmax 10-16: 698 and 675ng/dL>Cmax 16-24: 556 and 595 ng/dL). Equivalent mean testosterone C was observed for two doses of treatment Bmax(Cmax 0-10:894ng/dL~Cmax 10-24: 846 ng/dL). C between administration of t.i.d. formulationsmaxThe difference may be due to the different time periods that elapse between each administration. And treatment A and C (C, respectively)max: 830 and 883ng/dL), treatment B (150 μ L4.5% gel (b.i.d.)) (Cmax: 1050ng/dL) of the drug in the drug delivery solution during the 24 hour dosing intervalmaxSlightly larger. 1 of 8 subjects in treatment A and 3 of 7 subjects in treatments B and C exceeded the upper physiological reference range limit (1050 ng/dL).
When comparing the administration of t.i.d. and b.i.d. treatments separately, C was observedavgSlightly reduced trend (treatments A and C are C, respectively)avg 0-10: 418 and 436ng/dL>Cavg 10-16: 439 and 384ng/dL>Cavg 16-24: 377, and treatment B is Cavg 0-10:445ng/dL>Cavg 10-24: 376 ng/dL). C between administrationsavgThe difference may be due to the different time periods that elapse between each administration. Mean C calculated during the 24 hour dosing interval for all treatmentsavgAre comparable (treatments A, B and C are C, respectivelyavg:413,408,396ng/dL)。
These results indicate the exposure between each dose (AUC, C) of t.i.d. dosing (treatment a and C)avgAnd Cmax) Decrease, but not b.i.d. administration (treatment B). this reduction in exposure to t.i.d. administration can be explained in part by the negative feedback of endogenous testosterone production from the HPG axis. In other words, since the time interval between each administration of the t.i.d. group is small, the recovery of the HPG system from negative feedback will be smaller than that of the b.i.d. group.
Regardless of formulation, the mean drug concentration (C) in about 86% -88% of subjectsavg) C in 13% -14% of subjects within a physiological reference range (300 to 1050ng/dL)avgC below the reference range and without objectavgAbove the reference range.
The time periods during one day (24 hours) where serum testosterone concentrations were below, within and above the physiological reference range for all formulations covered 30 to 35%, 59% to 68% and 0% of the 24 hour period, respectively. That is, testosterone levels are within the normal range from about 14 to 16 hours a day.
Dihydrotestosterone
TBS-1 dosing was followed by 1 hour 24 minutes and 2 hours 23 minutes (mean T)max) The peak concentration of dihydrotestosterone is reached.
When comparing t.i.d. treated TBS-1 dosing alone, although the mean AUC between formulations was similar, a trend was observed for AUC to decrease with subsequent dosing (AUC for treatments A and C, respectively)0-10: 345.77 and 411.10h ng/dL>AUC10-16: 186.33 and 222.62h ng/dL>AUC16-24: 269.16 and 275.21h ng/dL). Treatment of twice BComparable AUC (AUC) was observed with dosing0-10:402.77h*ng/dL~AUC10-24: 543.29h ng/dL). the differences in AUC between t.i.d. formulation dosing may be due to the passage of different time periods between each dosing. Mean AUC calculated during the 24 hour dosing interval between all treatments0-τAre comparable (AUC for treatments A, B and C, respectively0-t: 818.95, 946.89 and 909.68h ng/dL).
Although the average C between t.i.d. formulationsmaxSimilarly, C was observedmaxTrend decreasing with subsequent dosing (treatments A and C are C, respectively)max 0-10: 51.4 and 59.0ng/dL>Cmax 10-16: 44.2 and 48.9ng/dL>Cmax 16-24: 41.3 and 42.6 ng/dL). Equivalent mean testosterone C was observed for two doses of treatment Bmax(Cmax 0-10:56.8ng/dL~Cmax 10-24: 54.6 ng/dL). C between administration of t.i.d. formulationsmaxThe difference may be due to the different time periods that elapse between each administration. Mean C calculated during the 24 hour dosing interval for all treatmentsmaxAre comparable (treatments A, B and C are C, respectivelymax: 52.2, 61.0 and 60.3 ng/dL). Any subject treated arbitrarily did not exceed the physiological reference range upper limit (97.8 ng/dL).
C between treatments and dosing by dosingavgAre comparable (treatments A and C are C, respectively)avg 0-10: 34.6 and 41.1ng/dL>Cavg 10-16: 31.1 and 37.1ng/dL>Cavg 16-24: 33.6 and 34.4ng/dL, and treatment B is Cavg 0-10:40.3ng/dL>Cavg 10-24: 38.8 ng/dL). Mean C calculated during the 24 hour dosing interval for all treatmentsavgAre comparable (treatments A, B and C are C, respectivelyavg:34.1,39.5,37.9ng/dL)。
Treatment of C in about 63% of subjects after A administrationavgIncluded in the DHT physiological reference range (25.5 to 97.8ng/dL), and this number rose to about 86% when treatments B and C were administered. Object-free CavgC in subjects above the normal range, while treatment A and treatments B and C are 38% and 14%, respectivelyavgIn the normal rangeThe following.
The time periods during the day (24 hours) in which serum DHT concentrations were below, within and above the physiological reference range covered 32.64%, 67.36% and 0% treatment a, 26.22%, 73.78% and 0% treatment B, and 13.87%, 86.13% and 0% treatment C, respectively. That is, DHT levels for treatments A, B and C were within the normal range at approximately 16, 18, and 21 hours a day, respectively.
Estradiol
TBS-1 dosing was followed by 1 hour 12 minutes and 2 hours 41 minutes (mean T)max) The peak concentration of the estradiol is reached.
When comparing t.i.d. treated TBS-1 dosing alone, although the mean AUC between formulations was similar, a trend was observed for AUC to decrease with subsequent dosing (AUC for treatments A and C, respectively)0-10: 234.96 and 267.78h pg/mL>AUC10-16: 144.76 and 144.30h pg/mL<AUC16-24: 153.02 and 177.97h pg/mL). Comparable AUC (AUC) was observed for both doses of treatment B0-10:242.02h*pg/mL~AUC10-24: 295.12h pg/mL). the differences in AUC between t.i.d. formulation dosing may be due to the passage of different time periods between each dosing. Mean AUC calculated during the 24 hour dosing interval between all treatments0-τAre comparable (AUC for treatments A, B and C, respectively0-t: 530.27, 537.16, and 601.91h pg/mL).
Although the average C between t.i.d. formulationsmaxSimilar, but C was observedmaxTrend decreasing with subsequent dosing (treatments A and C are C, respectively)max 0-10: 36.8 and 35.5pg/mL>Cmax 10-16: 28.9 and 31.5pg/mL>Cmax 16-24: 27.2 and 26.9 pg/mL). Equivalent mean testosterone C was observed for two doses of treatment Bmax(Cmax 0-10:35.8pg/mL~Cmax 10-24: 30.6 pg/mL). C between administration of t.i.d. formulationsmaxThe difference may be due to the different time periods that elapse between each administration. Mean C calculated during the 24 hour dosing interval for all treatmentsmaxAre equivalent (treatment A, B andc is respectively Cmax: 37.9, 36.2, and 36.4 pg/mL). Any subject treated arbitrarily did not exceed the physiological reference range upper limit (81 pg/mL).
C between treatments and dosing by dosingavgAre comparable (treatments A and C are C, respectively)avg 0-10: 23.5 and 26.8pg/mL>Cavg 10-16: 24.1 and 24.0pg/mL>Cavg 16-24: 19.1 and 22.2pg/mL, and treatment B is Cavg 0-10:24.2pg/mL>Cavg 10-24: 21.1 pg/mL). Mean C calculated during the 24 hour dosing interval for all treatmentsavgAre comparable (treatments A, B and C are C, respectivelyavg:22.1、22.4、25.1pg/mL)。
Total treatment of C in all subjects after dosingavgIs included in E2Physiological reference range (3 to 81 pg/mL). E of all objects2The concentration was within the normal range over a 24 hour period. Without object E2Levels were below or above the normal range at any time of the day.
Conclusion
TBS-1 formulations (4.0% TBS-1 gel (applied by t.i.d.) and 4.5% TBS-1 gel (applied by b.i.d. and t.i.d.)) were rapidly absorbed, with an average testosterone peak observed over 1 hour.
Overall, testosterone exposure at steady state (AUC) between all treatments0-tAnd Cmax) Are comparable.
Regardless of formulation, the mean testosterone drug concentration (C) of approximately 86% -88% of subjectsavg) Within a physiological reference range (300 to 1050 ng/dL).
Testosterone levels are within the normal range from about 14 to 16 hours a day.
TBS-1 was safe for intranasal administration at the doses and frequencies indicated. There were no meaningful adverse events, changes in vital signs, or changes in laboratory results when compared to baseline.
Based on these results, no clear evidence was found to demonstrate that one of the formulations performed better.
Example 9
TBS1A report for 4% and 8% batch gels
The purpose is as follows:
and then IMP-clinical batch preparation. The points relate to the process flow and batch appearance with respect to stability.
Improvement of the process
Viscosity of the batch gel
Stability (recrystallization)
Evaluation of alternative Material sources and grades
In vivo results, formulation changes affecting release onset
Test selection Using Franz cell
Determine the list of raw materials used for the experiment:
the equipment used was:
in addition to the Silverson high shear mixer, which was used only during the preparation of the TBS1A IMP clinical batches, a propeller type mixing unit was included for testing on several pre-mixing operations. The only application of high shear is to disperse the active agent in a co-solvent.
For more uniform mixing and temperature control, jacketed vessels with wiper blades have been proposed to remove material from the inner bowl wall (inner bowl wall) (especially important for uniform batch temperatures during heating and cooling cycles).
Preparation of IMP bathBackground information of
Observations during the preparation of IMP clinical batches include high viscosity during the preparation of a premix of the DMI/Transcutol cosolvent mixture consisting of PVP K17/S640, Klucel HF and micronized testosterone. The mixture resulted in a viscous quality when added to castor oil using a high shear mixer setup. Addition of Cab-O-Sil (referred to hereinafter as SiO) with the same high shear mixer setup2) No vortex of the incorporated material is obtained and additional manual mixing is required in the addition stage, thus suggesting a propeller type mixing unit). Even though the material had a viscosity in this addition stage, the viscosity of the final batch gel dropped to about 1,500-2,000cps upon further mixing. The mixing time and speed must be controlled to not exceed the target gel temperature (no cooling system).
The test key points are as follows:
initial trials (Placebo) focused on changing the order of addition to determine the effect on viscosity. Previous methods included the addition of SiO in the final stage2(see above) to SiO before addition of the alternative activator mixture2Dispersion in castor oil. The obtained castor oil/SiO2Mixture viscosity-with different percentages, increases with the addition of a small percentage of arlasolve (dmi).
The next step is to repeat these results with the active agent mixture (cosolvent/PVP/HPC/active agent) and add the mixture to castor oil and SiO2A premix of (1). However, this resulted in a low viscosity solution, indicating the effect of the active agent mixture on viscous gel formation.
Since the co-solvent mixture without other materials resulted in an increase in viscosity, the solvent amount was divided into 2 portions, only a portion of the solvent mixture was added to the oil mixture and the solvent mixture for dispersing PVP, HPC and the active agent was retained. The reduced co-solvent active agent mixture eliminates more stickiness and has a similarly low viscosity when added to the castor oil premix. Additional experiments included preparation of the Activity in DMI only (without PVP)And good viscosity is obtained. HPC was prepared in Transcutol P alone, creating a problem of stringing (similar to IMP observations) when added to the mixture. SiO is added at a level of 0.1-0.3%2The problem is solved.
The above method of dissolving the active agent in the co-solvent is sufficient to increase the solubility of the 4% formulation without the need for PVP, but the co-solvent in the formulation is not sufficient to achieve a solubility of 8% strength. The test for 8% included the following successful alternatives: by mixing SiO2Comprising preparing an active dispersion comprising PVP in the mixture. As for evaluation of SiO added to DMI and Transcutol P2The evaluation test of the effect of (a) proved to result in good viscosity formation of the DMI, but Transcutol did not. By dissolving PVP only in DMI, then adding the active agent and part of the available SiO at 55 deg.C (50-60 deg.C)2And preparing an active agent dispersion from the id.
Note that this method builds only at 8% during experimental work, so it can scale down to 4% strength if the PVP shows additional functionality (Franz cell test).
The comments on the addition of pure water (shown in table xxx) indicate that the viscosity increased in the tests containing HPC, but not in the tests using only PVP. These tests were included only to investigate the water intake and the effect on viscosity after administration into the nasal cavity.
A key step in the HPC setup is to provide at least 24 hours of solvation to obtain a clear solution.
As noted for experimental purposes, the formulation ratios were also performed for alternative material grades and sources and were determined in the formulation table.
To determine the effect of process changes (e.g., viscosity increase reactions with added co-solvent), experiments were conducted to investigate whether the effect was associated with DMI or Transcutol P. The test starts with SiO2Dispersion (same ratio as used for castor oil mixture) in DMI only and in Transcutol P only. DMI mixture conduitA viscous mixture, whereas the Transcutol P mixture is very fluid.
A similar experiment began with the study of the polymer and the potential for a decrease in solubility of the active agent in Transcutol P using co-solvents alone. There was no significant difference in solubility at 4% strength using either the mixture or the solvent alone. However, if PVP and HPC were prepared in DMI alone, separation of the two materials was observed upon storage overnight (not evident upon mixing in the co-solvent mixture).
To eliminate the stickiness of the dispersion when the activator/polymer mixture was added, HPC was removed from the formulation and PVP alone (grades K17-K29/32-K90, respectively, without mixture) was used. This results in varying degrees of tackiness, depending on the grade used.
The material also included the application of Labrafil M1944 CS and was summarized in the batch description and selected for Franz cell testing.
The following are commented:
the 4% strength and the 8% different tests are described below.
Test batches of both intensities have been selected for testing on Franz cells. The selected batch is determined.
The physical appearance of recrystallization and the apparent change (separation) were monitored throughout the experiment and the viscosity change was examined. The viscosity values of the tests were recorded and updated.
Ongoing Franz cell result evaluation, formulation and method optimization can be performed. This is critical to the identification, as the experimental point does not include all process parameter related effects on viscosity (including analytical testing and stability data is required).
Observations during the viscosity test-with a Brookfield Viscometer Model DV-II +, with Spindle #6, at 50rpm for 30 seconds-did show that the viscosity values of samples prepared with higher viscosity grades of HPC increased with test time. This may be attributed to the gel viscosity leading to agglomeration of the mandrel and disc, resulting in drag (not the actual viscosity value for the reported results). The batch gels for several experiments were not thixotropic. The viscosity at 37 ℃ was also examined in some tests.
Several tests were tested using the new Haupt method with mandrel 4 at 6 rpm.
The various attached tables show the number of trials of active gel, premix and placebo.
Subsequent experimental discussion and consideration of the two intensities
Even though 'viscosity increase' is not the main objective of the tests carried out, it is undoubtedly the high percentage of SiO present in the formulation considered2The design effort to investigate the cause of low viscosity is as follows. For SiO2Cross-examination of the alternative source comparisons did not show significant differences nor did it show different ratios of co-solvents, limited adjustment due to the percentage of dissolved testosterone required. Changes in the PVP grade show an effect on viscosity when used in an active agent dispersion, but do not show an effect on viscosity when added to the rest of the mixture. The change in HPC grade (using an alternative source of fine material) shows an effect on the final gel, but the higher the HPC molecular weight, the effect of viscosity and stringiness of the final gel. Testing the viscosity after several weeks showed that the gel separated and the viscose settled at the bottom of the container.
In SiO2More SiO was added as instructed by the fixed testosterone2Increasing the viscosity is not an option, the goal being to reduce the% used. In particular for TBS1A 4% strength, it showed a significantly higher percentage of T fixation compared to 8% TBS 1A. The goal is to obtain at least the same SiO as 8% strength at 4% strength2The ratio to T (hence, the goal is to scale down to 3%). SiO As tests were completed and showed viscosity effects associated with process and formulation changes2The reduction is clearly possible for a 4% strength, which would also include the use of PVP in the formulation — by using the method for 8% strength.
The above is based on viscosity only; the effect of formulation changes on slowing the initial rate of absorption in vivo can be assessed only from data obtained from experiments for testing using Franz cell analysis. These results will be examined and evaluated, with possible recommendations for further experiments to repeat previous experiments or based on DOE.
The viscosity chart shows the date of preparation and the latest test results (helpful for test selection on Franz cells). In the review section, the raw data will be referenced or referred to in the test method description.
Further alternative material source evaluations were suggested after the main formulation and method for each strength had been established for direct comparison.
TBS 1A-4% formulation/composition
Table 1A (see the above examples and including the formulation in example 10)
Lot#RD11037
The IMP batch (4%) process was repeated without HPC. K17 and S630 were dissolved in the DMI/Transcutol mixture, followed by addition of the active agent. And (4) a transparent solution. Castor oil preheat and add the active mixture described above. A clear solution was observed. Cabosil was then added with low shear. The viscosity at the time of preparation was 500cps, and then the result was checked to be 620cps after 48 hours.
The lower viscosity is mainly due to the absence of HPC (note that IMP 4% is about 1,500 cps).
Lot#RD11038
The order of addition was changed, the same formulation was used-with DMI/Transcutol reduced and adjusted with castor oil. Cabosil was mixed into castor oil to obtain a clear viscous solution. The active agent mixture was prepared according to RD 11037.
The viscosity of the castor oil/Cabosil mixture became 1180cps (higher viscosity was expected in the placebo run based on the addition of co-solvent). PVP and active agents may have an effect on the solvent mixture.
Lot#RD11039
This was repeated based on a placebo mixture also containing Labrafil in castor oil and Cabosil (for IP). The same viscosity reduction reaction occurs upon addition of the active agent mixture.
Lot#RD11040
The placebo process was repeated, adding a portion of the DMI/Transcutol P cosolvent mixture to the castor oil/Cabosil mixture. The viscosity of the oil mixture increases. The active agent mixture was prepared with the remaining co-solvent without PVP and added to the oil mixture. The final viscosity of the batch gel was 10,400 cps. F/C can be carried out.
Lot#RD11041
The process was repeated according to RD11040, the active agent mixture comprised PVP K17 and S630, and the viscosity dropped to 500cps (to 1,500cps after 3 weeks). The effect of PVP on viscosity reduction was clearly shown using K17 and S630.
Lot#RD11042
The experiment was repeated with castor oil/Labrafil addition according to RD11037 and Cabosil was reduced with the active cosolvent mixture but no PVP. The viscosity was 1,750 cps.
The following tests were designed to determine the impact of changing to a higher PVP grade and replacing the HPC source (grade 2). Premixing was performed with natural castor oil and Aerosil 200 as shown in table 3 for the mixture without Labrafil.
Lot#RD11050
A dispersion of castor oil and Aerosil 200 (premix I) was prepared and the viscosity was increased by adding part of DMI (4%). Preparation of active agent mixture a premix of RD11047A (PVP K17-3%) in DMI only was used, 0.3% of HPC Nisso H was added, followed by the active agent. Add the active agent mixture to premix I.
Lot#RD11050A
The same base formulation as RD11050 with the change that another 1% Aerosil 200 was added to one part.
Lot#RD11051
A dispersion of castor oil and Aerosil 200 (premix I) was prepared and the viscosity was increased by adding part of DMI (4%). Preparation of active agent mixture a premix of RD11047B (PVP K30-3%) in DMI only was used, 0.3% of HPC Nisso M was added, followed by the active agent. The active agent mixture was added to premix I.
Lot#RD11051A
The same base formulation as RD11051, with the change being the addition of another 1% Aerosil 200 to a portion.
Lot#RD11053
A dispersion of castor oil and Aerosil 200 (premix I) was prepared and the viscosity increased by adding part of DMI and Transcutol P. Preparation of the active agent mixture A premix of RD11048A (PVP K17-3%) was used, 0.3% HPC Nisso H was added, and then the active agent was added. The active agent mixture was added to premix I.
Lot#RD11054
A dispersion of castor oil and Aerosil 200 (premix I) was prepared and the viscosity increased by adding part of DMI and Transcutol P. Preparation of active agent mixture premix RD11048B (PVP K30-3%) was used, 0.3% of HPC Nisso H was added, followed by the active agent. The active agent mixture was added to premix I.
Lot#RD11055
A dispersion of castor oil and Aerosil 200 (premix I) was prepared and the viscosity increased by adding part of DMI and Transcutol P. The preparation of the active agent mixture used a premix of RD11048C (PVP K90-3%). No HPC addition. The active agent mixture was added to premix I.
Lot#RD11056
A dispersion of castor oil and Aerosil 200 (premix I) was prepared and the viscosity increased by adding part of the DMI. The preparation of the active agent mixture used a premix of RD11047C (PVP K90-3%). No HPC addition. The active agent mixture was added to premix I.
Lot#RD11059
A mixture of castor oil and Cabosil (2.5%) was prepared. The active agent is dissolved in DMI and Transcutol P. Resulting in a milky appearance. The mixture was added to the castor oil premix and the mixture did not become clear. A PVP (K30) solution was prepared with DMI and added to the mixture with no change in appearance but with a decrease in viscosity.
Note that there was no apparent change in the evaluation of the mixture with 0.1% HPC added, and the viscosity increased slightly. The test is not reported as test batch number.
Lot#RD11060
Castor oil was prepared with 3.5% Cabosil added, followed by the addition of a DMI/Transcutol P mixture for thickening. A dispersion of the active agent in PVP (K30) was prepared with DMI as co-solvent. (without HPC)
Lot#RD11061
Castor oil was prepared with 3% Cabosil added followed by Labrafil (2%) to thicken. Dispersions of active agents in DMI mixtures containing PVPK17 (2%) were prepared. Mixing results in low viscosity, but F/C testing is contemplated.
Lot#RD11062
Natural castor oil was mixed with Aerosil 200 (3%) and a DMI/Transcutol P (6+2) mixture was added to thicken. The PVP mixture of K17 and K30 was dissolved in DMI/Transcutol P, then HPC H and solvate for 4 days. The mixture is then heated before the active agent is added. The castor oil premix is heated prior to addition of the active dispersion. F/C is recommended.
Lot#RD11063
Natural castor oil was mixed with Aerosil 200 (4%) and DMI (6%) was added, resulting in a highly viscous gum mixture. The mixture of PVPK17 and L29/32 was dissolved in DMI and HPC Nisso H (0.2) was added. At the overnight setting, separation was observed, requiring remixing. The active was added to a high viscosity castor oil premix. Followed by a compositional change.
F/C or application RD11065 can be carried out.
Lot#RD11064
Add 0.3% to partial batch RD 11062.
Lot#RD11065
Add 0.3% to partial batch RD 11063.
Lot#RD11066
Add 0.3% to partial batch RD 11041.
Lot#RD11070
Add 0.3% to partial batch RD 11037.
Lot#RD11071
Add 0.3% to partial batch RD 11042.
Lot#RD11072
Add 0.3% to partial batch RD 11040.
Lot#RD11073
A castor oil/Aerosil 200 premix was prepared. Testosterone was dissolved in DMI without PVP (6%) and added to the castor oil premix. A viscosity of 6,300cps was obtained. HPC M (prepared with 0.25% only) was dispersed in the Transcutol P and DMI mixture and added to the main mixture. F/C is recommended.
Lot#RD11074
Add 0.3% to partial batch RD 11072.
Lot#RD11075
Stock mixtures were prepared to complete a 3x 500g trial consisting of castor oil (68%) Aerosil 200 (3%) DMI (6%). To this mixture was added PVP K29-32 (1%) in DMI (10) and an active agent. The batch was divided into 3 portions to complete 3 trials: different mixtures and HPC Nisso grades contained in Transcutol (reference batch RD11067/68/69)
Lot#RD11076
Batches from RD11075 were used and HPC mixture RD11067(Transcutol P with Nisso H (0.15%)
Lot#RD11077
Batches from RD11075 were used and HPC mixture RD11068(Transcutol P with Nisso H (0.2%)
Lot#RD11078
Batches from RD11075 were used and HPC mixture RD11069(Transcutol P with Nisso H (0.1) and M (0.1) was added
Lot#RD11079
Add 0.3% to partial batch RD 11076.
Lot#RD11080
Add 0.3% to partial batch RD 11077.
Lot#RD11081
Add 0.3% to partial batch RD 11078.
Lot#RD11082
The test attempted to prepare batches without SiO2 and failed.
Lot#RD11085
A castor oil premix was prepared and 2.5% Aerosil 200 was added followed by a mixture of DMI (10) and testosterone. A viscosity of 3,100cps was obtained. HPC Nisso L (0.2%) and Nisso M (0.3%) were then added mixed in DMI and Transcutol and 0.3% Aerosil 200 was added to reduce tack. The material addition did not have any stringing to the main mix and achieved a viscosity of 4,800cps on the day of preparation and a viscosity of 4,900cps after 3 weeks. F/C is recommended.
Lot#RD11086
Add 0.3% to a portion of batch RD 11085.
TABLE 2TBS1A 4% strength
The test was repeated using the viscosity value of the mandrel 6, 20rpm, with reference to the Franz cell: F/C
TBS1A
8% formulation/composition
TABLE 3
The method for activity test comprises the following steps:
Lot#RD11087
the test was started without PVP to determine the effect on T solubility. The active dispersion in the% DMI used did not appear as a clear solution and did not become clear when added to the castor oil/SiO 2 mixture. The clear bulk gel does not appear even if a co-solvent is present in the HPC mixture. 0.1% SiO2 was added to the HPV mixture to reduce stringiness and stickiness.
The viscosity is 4,400
However, this test will be selected for Franz cell testing to determine the diffusion rate excluding PVP.
Lot#RD11088
0.3% water was added to a portion of batch RD11087 to determine the effect on viscosity. As observed in the 4% trial, the viscosity increase of the batch mixed with SiO2 in HPC was not significant. This test does not take into account F/C.
Lot#RD11089
This trial used the same quantitative formulation as IMP in clinical 8% but used an alternative HPC source (original HPC source Klucel HF). A minor process change was also made to dissolve PVP in DMI only and add the active agent. HPC was prepared in Transcutol and added separately to the main batch.
A clear solution was obtained upon addition of the active co-solvent mixture to castor oil and no significant stringiness was observed upon addition of HPC after addition of SiO 2.
The gel viscosity was 1,800cps on the day of preparation, 3,700 on review after 24 hours, and up to 4,300 after 48 hours. A review of 10 months and 3 days (see Table) recorded 4,500 cps.
The test selects to carry out F/C test
Lot#RD11089A
0.3% water was added to a portion of batch RD11089 to determine the effect on viscosity.
The viscosity change over time was similar to the test described above, being 2,700cps on the day of preparation, 3,920 on review after 24 hours and up to 4,600 after 48 hours. A review of 10 months and 3 days (see Table) recorded 5,040 cps.
Selection for study on Water effects
Lot#RD11090
A higher percentage of DMI and Transcutol was used, which would be divided into multiple premixes, similar to SiO2 would be added to HPC. A premix of castor oil and SiO2 was prepared, but due to the lower ratio between the 2 excipients, the mixture became quite viscous and thickened further upon addition of part of the DMI.
The test was completed, ending at a low viscosity, 900cps on the day of preparation, testing-1,260 cps 3 days 10 months. Lower levels of SiO2 were considered for research effects, but for processing issues (see RD11100)
Is not suitable for F/C test
Lot#RD11100
Using part of the above test RD11090, an additional 2% SiO2 (total 5.5%) was added to investigate the effect on viscosity. Increasing to 1,900cps on the day of preparation, a 10 month 3 day review (see table) resulted in a value of 3.060.
Lot#RD11101
To possibly reduce the effect of PVP, it was necessary to dissolve the active agent, adding 2% SiO2 to the DMI-PVP-testosterone mixture during the addition to the castor oil/SiO 2 mixture, obtaining a viscous mixture. After adding the mixture to a castor oil dispersion containing 1% SiO2, the viscous mixture was kept at a temperature of 50% (which would further thicken on cooling). The viscosity further increased with the addition of the HPC mixture and the final amount of SiO 2.
The viscosity was 3,800cps after cooling the gel to 21 ℃. (Note that will require review over time, batches were prepared on day 3/10)
This test was selected for F/C
Lot#RD11102
In the case where TBS1A projected target was 5,000cps viscosity, RD11101 described above was the best candidate to evaluate the effect of further adding SiO2, so an additional 1% SiO2 was added to a portion of the batch. 6% is to obtain the same ratio of active agent to SiO2 as the target level of 3% SiO2 in 4% strength.
The viscosity increased to 8,000cps and the batch was selected for F/C studies to determine the effect of viscosity on diffusion rate compared to RD11101 of the same composition except for the addition of 1% SiO2, which may need to be considered based on the analysis obtained.
Lot#RD11103
Addition of water to obtain an effect on viscosity, irrespective of the subsequent tests (see viscosity results table, RD11101 increasing from 3,800 to 4,500cps)
Lot#RD11104
This test was included to evaluate the addition of Labrafil. Labrafil was added to castor oil mixed with 1% SiO 2. As previously observed, adding Labrafil to castor oil containing SiO2 increased viscosity. All other mixtures were prepared and added according to test RD11101, wherein 2% SiO2 was added to complete the mixture. The mixture contained a large percentage of bubbles, common to formulations containing Labrafil.
A viscosity of 3,300cps was obtained and would be tracked and verified at various time points.
The F/C test was selected.
Lot#RD11105
An additional 0.5% SiO2 (adjusted% to avoid the height increase observed on RD11102) was added to RD 11104.
From 3,300 to 4,100cps
F/C test was not selected
Note: a placebo trial was set up to determine the effect on viscosity using castor oil from 2 different sources and SiO 2. These trials will also answer potential questions about TBS1 and TBS 2.
TABLE 4TBS1A 8% strength
Viscosity values using spindle #6, 20rpm, Franz cell ═ F/C
Premix RD test (for addition in the active agent test)
TABLE 5
Placebo TBS1A test
TABLE 6
Example 10
Franz cell study-Testosterone diffusion Rate
In general, the membrane was soaked in the diffusion solution for 30 minutes. After placing the membrane on the Franz cell. The ring and donor chamber were placed on the membrane and clamped. About 1 g of gel (TBS 1A 4% or 8%) was added. The level of diffusion solution in the Franz cell was examined. It should be on the label. A "Parafilm" was placed on the sampling port to avoid evaporation. 0.3mL of sample was taken with a syringe at 60, 120, 180, 240, 300 and 360 minutes. The diffusion solution was added to allow it to rise to Franz cell label. Each sample should be collected into the insert.
A general Fanz cell used according to this example 9 and the invention is shown in fig. 13. The material comprises:
diffusion solution: ethanol/water 50: 50
Film formation: millipore 0.45 μm.
Temperature: 37 ℃ plus or minus 0.5 ℃.
Stirring speed: 600 rpm.
Volume of medium: 20 mL.
Surface area: 1.7671cm2
Number of Franz pools: 6.
sampling time (min): 60. 120, 180, 240, 300 and 360.
Volume of aliquot: 0.3 mL.
Inserting: 0.4 mL.
TBS1A formulation was as follows and is reported above and in the examples herein. The resulting rate of testosterone diffusion through the Franz cell membrane, normalized for each gel concentration tested, measured as slope/mgT%, is reported in the Franz cell table below.
4% TBS1A test formulation for Franz cell
Test batch # RD11063Batch size 500g
Test batch # RD11085Batch size 500g
Test batch # RD11038Batch size 500g
Test batch # RD11039Batch size 500g
Test batch # RD11040Batch size 500g
Test batch # RD11042Batch size 500g
Test batch # RD11051Batch size 500g
Test batch # RD11055Batch size 500g
Test batch # RD11078Batch size 500g
Test batch # RD11054Batch size 500g
Test batch # RD11061Batch size 500g
In vitro release rate validation of TBS-1A gels summarized with respect to TBS-1A gel 4.0% and TBS-1A gel 4.5% release rate studies is shown in appendices A and B of the accompanying filing.
These summaries summarize the release rate experimental data for an exemplary TBS-1A gel. Four Nasobol gels (0.15%, 0.6%, 4.0% and 4.5%) were used for method validation. The purpose of the day 1 and day 2 tests was to determine the characteristics of the slope (release rate) and day-to-day/day accuracy, and the purpose of the day 3 and day 4 tests was to assess the sensitivity of the slope to changes in sample intensity.
See appendix A (4.0%) and appendix B (4.5%) filed herewith, both of which are incorporated herein by reference in their entirety.
List of references
Tietz Textbook of Clinical Chemistry and Molecular Diagnostics,4th edition,2006. Editors; burtis CA, Ashwood ER, andBurns DE.
Wang C, Swerdloff rs. android replacement therapy (androgen replacement therapy). Ann Med 1997; 29:365-370.
(iii) clinical indications of the deciline hormone levels with the formation of men's menopause: clinical implications of men's concomitant decline in serum Testosterone levels with aging.) J Gerontol A Med Sci 2002; 57: M76-M99.
Haren MT, Kim MJ, Tariq SH, Wittert GA, Morley JE.Andropause: a quality-of-life issue in emulsions male (andropause: quality of life problem in elderly males). Med Clin NorthAm 2006; 90:1005-1023.
Nieschlag E.Testosterone treatment communities of age: new options for hypogonadal men (New choice of Testosterone treatment epoch: hypogonadal Male.) Clin Endocrinol (Oxf)2006:65: 275-.
Tenover JL, the android-specific imaging male current treatment options Rev Urol 2003; 5(Suppl) S22-S28.
Jockenhevel f.testosterone therapy-what, when and to whom? Aging Male 2004; 7:319-324.
Kunz GH, Klein KO, Clemons RD, Gottschalk ME, Jones KL. viriliation of youung childern topical androgen use by the third party (feminization of young children after topical androgen application by parents.) Pediatrics 2004; 114:282-284.
Brachhet C, Vermeulen J, Heinrichs C, children's viriliation and the use of a Testosterone gel by the hair disorders (Testosterone gel for feminization and father of children), EurJ Pediatr 2005; 164:646-647.
Bagchus WM, hurt R, Maris F, Schnabel PG, Houwing ns.inportant effect food on the bioavailability of oral Testosterone monocanate (a significant effect of food on the bioavailability of oral Testosterone undecanoate.) Pharmacotherapy 2003; 23:319-325.
Haren M, Chapman IM, Haren MT, MacKintosh S, coats P, Morley JE. Oral Testosterone supplementation creates muscles and inventions fat in fatty aldehydes polypeptides with low normal gonadal status J Gerontol A Biol Sci Med Sci 2003; 58:618-625.
Haren M, Chapman I, Coates P, Morley JE, Wittert G.Effect of 12mon temporal ketone on Testosterone deficience systems in symptomatic eldiales with low-normal gonadal symptoms (the effect of Testosterone deficiency symptoms by oral administration for 12 months in symptomatic elderly men with low normal gonadal status.) Age Ageing 2005; 34:123-130.
Mattern C, Hoffmann C, Morley JE, Badiu C.the Aging Male 2008; 11:171-178.
Nasobol gel 4.0% in vitro release rate method verification
NASBOBOL gel in vitro release rate verification update
Summary of Release Rate Studies
Section 3: nasobol gel 4.0%
Purpose(s) to
This summary summarizes all release rate experimental data for the Nasobol gel.
Four Nasobol gels (0.15%, 0.6%, 4.0% and 4.5%) were used for method validation.
The purpose of the day 1 and day 2 tests was to determine the specificity and intra/inter-day accuracy of the slope (release rate), and the purpose of the day 3 and day 4 tests was to assess the sensitivity of the slope to changes in sample intensity.
Appendix A
Nasobol gel 4.0% in vitro release rate method verification
Nasobol gel 4.0% release Rate
Nasobol gel 4.0% in vitro release rate method verification
4% day 1 Release Rate
Testosterone 4% gel
Actual amount of active agent released (. mu.g/cm)2) And time0.5
Nasobol gel 4.0% in vitro release rate method verification
Systemic Adaptation of 4% gel, day 1
1 medium (diluent)
| Results | No interference peak at testosterone RT from diluent injection |
| Standard of merit | There should be no significant interfering peaks at testosterone from diluent RT |
| Review the following | By passing |
Injection reproducibility RT, tailing factor and theoretical plate number for six repeated injections of 2: STD-4
3 calibration curve Y-16016.225515 x-601.467936
Check STD Rec. (%)
Nasobol gel 4.0% in vitro release rate method verification
4% day 2 Release Rate
Testosterone 4% gel
Active agent concentration (μ g/mL) and time
Actual amount of active agent released (μ g/cm2) versus time0.5
Nasobol gel 4.0% in vitro release rate method verification
4% systemic Adaptation of Nasobol gel, day 2
1 medium (diluent)
| Results | No interference peak at testosterone RT from diluent injection |
| Standard of merit | There should be no significant interfering peaks at testosterone from diluent RT |
| Review the following | By passing |
Injection reproducibility RT, tailing factor and theoretical plate number for six repeated injections of 2: STD-4
3 calibration curve Y-16191.343821 x-559.963706
Check STD Rec. (%)
Nasobol gel 4.0% in vitro release rate method verification
4% day 3 Release Rate
Testosterone gel 2.0%
Actual amount of active agent released (μ g/cm2) versus time0.5
Nasobol gel 4.0% in vitro release rate method verification
4.0% day 3 Release Rate
Testosterone gel 4.0%
Actual amount of active agent released (μ g/cm2) versus time0.5
Nasobol gel 4.0% in vitro release rate method verification
4.0% day 3 Release Rate
Testosterone gel 8.0%
Actual amount of active agent released (μ g/cm2) versus time0.5
Nasobol gel 4.0% in vitro release rate method verification
4% systemic Adaptation of Nasobol gel, day 3
1 medium (diluent)
| Results | No interference peak at testosterone RT from diluent injection |
| Standard of merit | There should be no significant interfering peaks at testosterone from diluent RT |
| Review the following | By passing |
Injection reproducibility RT, tailing factor and theoretical plate number for six repeated injections of 2: STD-4
3 calibration curve Y-16400.350881 x-586.919769
Check STD Rec. (%)
Nasobol gel 4.0% in vitro release rate method verification
Testosterone gel 2% day 4 Release Rate
Active agent concentration (μ g/mL) and time
Actual amount of active agent released (μ g/cm2) versus time0.5
Nasobol gel 4.0% in vitro release rate method verification
Testosterone gel 4% day 4 Release Rate
Active agent concentration (μ g/mL) and time
Actual amount of active agent released (. mu.g/cm)2) And time0.5
Nasobol gel 4.0% in vitro release rate method verification
Testosterone gel 8% day 4 Release Rate
Testosterone gel 8%
Actual amount of active agent released (μ g/cm2) versus time0.5
Nasobol gel 4.0% in vitro release rate method verification
System adaptability
1 medium (diluent)
| Results | No interference peak at testosterone RT from diluent injection |
| Standard of merit | There should be no significant interfering peaks at testosterone from diluent RT |
| Review the following | By passing |
Injection reproducibility RT, tailing factor and theoretical plate number for six repeated injections of 2: STD-4
3 calibration curve Y-16123.2297 x-239.5651
Check STD Rec. (%)
Nasobol gel 4.5% in vitro release rate method verification
NASBOBOL gel in vitro release rate verification update
Summary of Release Rate Studies
Part 4 Nasobol gel 4.5%
Purpose(s) to
This summary summarizes all release rate experimental data for the Nasobol gel. Four Nasobol gels (0.15%, 0.6%, 4.0% and 4.5%) were used for method validation. The purpose of the day 1 and day 2 tests was to determine the specificity and intra/inter-day accuracy of the slope (release rate), and the purpose of the day 3 and day 4 tests was to assess the sensitivity of the slope to changes in sample intensity.
Appendix B
Nasobol gel 4.5% in vitro release rate method verification
Nasobol gel 4.5% release Rate
Nasobol gel 4.5% in vitro release rate method verification
4.5% day 1 Release Rate
Testosterone 4.5% gel
Active agent concentration (μ g/mL) and time
Actual amount of active agent released (μ g/cm2) versus time0.5
Nasobol gel 4.5% in vitro release rate method verification
Systemic Adaptation of 4% gel, day 1
1 medium (diluent)
| Results | No interference peak at testosterone RT from diluent injection |
| Standard of merit | There should be no significant interfering peaks at testosterone from diluent RT |
| Review the following | By passing |
Injection reproducibility RT, tailing factor and theoretical plate number for six repeated injections of 2: STD-4
3 calibration curve Y-16214.013222 x-404.968835
Check STD Rec. (%)
Nasobol gel 4.5% in vitro release rate method verification
4.5% day 2 Release Rate
Testosterone 4.5% gel
Active agent concentration (μ g/mL) and time
Actual amount of active agent released (μ g/cm2) versus time0.5
Nasobol gel 4.5% in vitro release rate method verification
Nasobol gel 4.5% systemic adaptability, day 2
1 medium (diluent)
| Results | No interference peak at testosterone RT from diluent injection |
| Standard of merit | There should be no significant interfering peaks at testosterone from diluent RT |
| Review the following | By passing |
Injection reproducibility RT, tailing factor and theoretical plate number for six repeated injections of 2: STD-4
3 calibration curve Y-16354.946833 x-532.850889
Check STD Rec. (%)
Nasobol gel 4.5% in vitro release rate method verification
4.5% day 3 Release Rate
Testosterone gel 2.25%
Active agent concentration (μ g/mL) and time
Actual amount of active agent released (μ g/cm2) versus time0.5
Nasobol gel 4.5% in vitro release rate method verification
4.5% day 3 Release Rate
Testosterone gel 4.5% gel
Active agent concentration (μ g/mL) and time
Actual amount of active agent released (μ g/cm2) versus time0.5
Nasobol gel 4.5% in vitro release rate method verification
4.5% day 3 Release Rate
Testosterone gel 9.0%
Active agent concentration (μ g/mL) and time
Actual amount of active agent released (μ g/cm2) versus time0.5
Nasobol gel 4.5% in vitro release rate method verification
Nasobol gel 4.5% systemic adaptability, day 3
1 medium (diluent)
| Results | No interference peak at testosterone RT from diluent injection |
| Standard of merit | There should be no significant interfering peaks at testosterone from diluent RT |
| Review the following | By passing |
Injection reproducibility RT, tailing factor and theoretical plate number for six repeated injections of 2: STD-4
3 calibration curve Y-16446.511438 x-909.542212
Check STD Rec. (%)
Nasobol gel 4.5% in vitro release rate method verification
4.5% day 4 Release Rate
Testosterone gel 2.25%
Active agent concentration (μ g/mL) and time
Actual amount of active agent released (μ g/cm2) versus time0.5
Nasobol gel 4.5% in vitro release rate method verification
4.5% day 4 Release Rate
Testosterone gel 4.5%
Active agent concentration (μ g/mL) and time
Actual amount of active agent released (μ g/cm2) versus time0.5
Nasobol gel 4.5% in vitro release rate method verification
4.5% day 4 Release Rate
Testosterone gel 9.0%
Active agent concentration (μ g/mL) and time
Actual amount of active agent released (μ g/cm2) versus time0.5
Nasobol gel 4.5% in vitro release rate method verification
Nasobol gel 4.5% systemic adaptability, day 4
1 medium (diluent)
| Results | No interference peak at testosterone RT from diluent injection |
| Standard of merit | There should be no significant interfering peaks at testosterone from diluent RT |
| Review the following | By passing |
Injection reproducibility RT, tailing factor and theoretical plate number for six repeated injections of 2: STD-4
3 calibration curve Y-16050.748753 x-354.26124
Check STD Rec. (%)
Claims (28)
1. A testosterone gel formulation for nasal administration, said testosterone gel formulation comprising:
a. (ii) about 4.0% testosterone by weight of the gel formulation; and
b. a pharmaceutically acceptable carrier.
2. A testosterone gel formulation for nasal administration, said testosterone gel formulation comprising:
a. (ii) about 4.5% testosterone by weight of the gel formulation; and
b. a pharmaceutically acceptable carrier.
3. A testosterone gel formulation for nasal administration, said testosterone gel formulation comprising:
a. (ii) about 8.0% testosterone by weight of the gel formulation; and
b. a pharmaceutically acceptable carrier.
4. The testosterone gel formulation of claims 1-3, wherein said gel formulation comprises a solvent, a wetting agent, and a viscosity increasing agent.
5. The testosterone gel formulation of claim 4, wherein said solvent is castor oil.
6. The testosterone gel formulation of claim 4, wherein said wetting agent is oleoyl polyoxylglyceride.
7. The testosterone gel formulation of claim 4, wherein said viscosity increasing agent is colloidal silicon dioxide.
8. The testosterone gel formulation of claims 1-3, wherein said gel formulation comprises castor oil, oleoyl polyoxylglycerides, and colloidal silicon dioxide.
9. The testosterone gel formulation of any one of claims 1-8, wherein said gel formulation is a bioequivalent formulation.
10. The testosterone gel formulation of any one of claims 1-8, wherein said gel formulation is a pharmaceutically equivalent formulation.
11. The testosterone gel formulation of any one of claims 1-8, wherein said gel formulation is a therapeutically equivalent formulation.
12. An encapsulated drug comprising:
(a) a testosterone gel formulation for nasal administration or a pharmaceutically acceptable salt or prodrug thereof, wherein said gel formulation comprises about 4.0% testosterone by weight; and (b) instructions for using the testosterone gel formulation in testosterone replacement therapy or in the treatment of hypogonadism or testosterone deficiency.
13. An encapsulated drug comprising:
(a) a testosterone gel formulation for nasal administration or a pharmaceutically acceptable salt or prodrug thereof, wherein said gel formulation comprises about 4.5% testosterone by weight; and (b) instructions for using the testosterone gel formulation in testosterone replacement therapy or in the treatment of hypogonadism or testosterone deficiency.
14. An encapsulated drug comprising:
(a) a testosterone gel formulation for nasal administration or a pharmaceutically acceptable salt or prodrug thereof, wherein said gel formulation comprises about 8.0% testosterone by weight; and (b) instructions for using the testosterone gel formulation in testosterone replacement therapy or in the treatment of hypogonadism or testosterone deficiency.
15. The packaged pharmaceutical of claims 12-14, wherein said testosterone is present in a pharmaceutical composition comprising a therapeutically effective amount of testosterone or a pharmaceutically acceptable salt or prodrug thereof and a pharmaceutically acceptable carrier.
16. The packaged pharmaceutical of claims 12-14, further comprising the step of identifying a subject in need of said pharmaceutical.
17. A method of treating hypogonadism in a male subject, the method comprising intranasally administering the gel formulation of any one of claims 1-16 to a male subject to deliver a therapeutically effective amount of testosterone to effectively treat hypogonadism.
18. A method of treating testosterone deficiency in a male subject, said method comprising intranasally administering the gel formulation of any one of claims 1-16 to a male subject to deliver a therapeutically effective amount of testosterone to effectively treat testosterone deficiency.
19. A method of providing testosterone replacement therapy in a male subject, said method comprising administering intranasally to a male subject said gel formulation of any one of claims 1-16 to deliver a therapeutically effective amount of testosterone to effectively provide testosterone replacement therapy.
20. A method of preparing a testosterone gel formulation for nasal administration, said testosterone gel formulation comprising about 4.0% testosterone by weight of said gel formulation; and a pharmaceutically acceptable carrier, the method comprising:
a. mixing the micronized testosterone to contact a solvent to form a first mixture;
b. mixing oleoyl polyoxylglyceride with the first mixture to form a second mixture; and
c. mixing colloidal silicon dioxide with the second mixture to provide a testosterone gel formulation for nasal administration.
21. A method of preparing a testosterone gel formulation for nasal administration, said testosterone gel formulation comprising about 4.5% testosterone by weight of said gel formulation; and a pharmaceutically acceptable carrier, the method comprising:
a. mixing the micronized testosterone to contact a solvent to form a first mixture;
b. mixing oleoyl polyoxylglyceride with the first mixture to form a second mixture; and
c. mixing colloidal silicon dioxide with the second mixture to provide a testosterone gel formulation for nasal administration.
22. A method of preparing a testosterone gel formulation for nasal administration, said testosterone gel formulation comprising about 8.0% testosterone by weight of said gel formulation; and a pharmaceutically acceptable carrier, the method comprising:
a. mixing the micronized testosterone to contact a solvent to form a first mixture;
b. mixing oleoyl polyoxylglyceride with the first mixture to form a second mixture; and
c. mixing colloidal silicon dioxide with the second mixture to provide a testosterone gel formulation for nasal administration.
23. A gel formulation of claims 1-22, wherein the testosterone gel formulation has a testosterone rate of diffusion of between about 28 to about 100 slope/mgT%.
24. A gel formulation of claims 1-22, wherein the testosterone gel formulation has a testosterone rate of diffusion of between about 30 to about 95 slope/mgT%.
25. A gel formulation of claims 1-22, wherein the testosterone gel formulation has a testosterone rate of diffusion of between about 28 to about 35 slope/mgT%.
26. A testosterone gel formulation for nasal administration, said testosterone gel formulation comprising:
a. testosterone; and
b. a pharmaceutically acceptable carrier, wherein the testosterone gel formulation has a testosterone rate of diffusion of between about 28 to about 100 slope/mgT%.
27. A testosterone gel formulation for nasal administration, said testosterone gel formulation comprising:
a. testosterone; and
b. a pharmaceutically acceptable carrier, wherein the testosterone gel formulation has a testosterone diffusion rate of between about 30 to about 95 slope/mgT%.
28. A testosterone gel formulation for nasal administration, said testosterone gel formulation comprising:
a. testosterone; and
b. a pharmaceutically acceptable carrier, wherein the testosterone gel formulation has a testosterone rate of diffusion of between about 28 to about 35 slope/mgT%.
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US61/486,324 | 2011-05-15 | ||
| US61/486,634 | 2011-05-16 |
Publications (2)
| Publication Number | Publication Date |
|---|---|
| HK1263036A1 true HK1263036A1 (en) | 2020-01-24 |
| HK1263036B HK1263036B (en) | 2023-11-17 |
Family
ID=
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| CN109481394B (en) | Intranasal testosterone bioadhesive gel formulations and their use for treating male hypogonadism | |
| US20250064831A1 (en) | Controlled release topical testosterone formulations and methods | |
| US20250009756A1 (en) | Male Testosterone Titration Methods, Male Intranasal Testosterone Bio-Adhesive Gel Formulations and Use Thereof for Treating Hypogonadism and TRT | |
| US20240316063A1 (en) | Methods of treating hypogonadism with transnasal testosterone bio-adhesive gel formulations in male with allergic rhinitis, and methods for preventing an allergic rhinitis event | |
| HK1263036A1 (en) | Intranasal testosterone bio-adhesive gel formulations and use thereof for treating male hypogonadism | |
| HK1263036B (en) | Intranasal testosterone bio-adhesive gel formulations and use thereof for treating male hypogonadism | |
| HK1196287B (en) | Intranasal testosterone bio-adhesive gel formulations and use thereof for treating male hypogonadism | |
| HK1196287A (en) | Intranasal testosterone bio-adhesive gel formulations and use thereof for treating male hypogonadism | |
| EA039080B1 (en) | Intranasal testosterone bio-adhesive gel formulations and use thereof for treating male hypogonadism | |
| HK1196261B (en) | Controlled release nasal testosterone gels, methods and pre-filled multi-dose applicator systems for pernasal administration |