Vaginal inserted estradiol pharmaceutical compositions and methods

Information

  • Patent Grant
  • 11351182
  • Patent Number
    11,351,182
  • Date Filed
    Friday, March 27, 2020
    4 years ago
  • Date Issued
    Tuesday, June 7, 2022
    a year ago
Abstract
According to various embodiments of this disclosure, pharmaceutical compositions comprising solubilized estradiol are provided. In various embodiments, such compositions are encapsulated in soft capsules which may be vaginally inserted for the treatment of vulvovaginal atrophy.
Description
BACKGROUND

This application is directed to pharmaceutical compositions, methods, and devices related to hormone replacement therapy.


Postmenopausal women frequently suffer from atrophic vaginitis or vulvar and vaginal atrophy (hereinafter “vulvovaginal atrophy” or “VVA”) with symptoms including, for example, vaginal dryness, vaginal odor, vaginal or vulvar irritation or itching, dysuria (pain, burning, or stinging when urinating), dysparuenia (vaginal pain associated with sexual activity), or vaginal bleeding associated with sexual activity. Other symptoms include soreness; with urinary frequency and urgency; urinary discomfort and incontinence also occurring (“estrogen-deficient urinary state(s)”). One symptom of vaginal atrophy is an increased vaginal pH, which creates an environment more susceptible to infections. The mucosal epithelium of the VVA patients also reported to show signs of severe atrophy and upon cytological examination accompanied by an increased number of the parabasal cells and a reduced number of superficial cells.


Each of these VVA-related states manifest symptoms associated with decreased estrogenization of the vulvovaginal tissue, and can even occur in women treated with oral administration of an estrogen-based pharmaceutical drug product. Although VVA is most common with menopausal women, it can occur at any time in a woman's life cycle.


Estrogen treatment has proven to be very successful in controlling menopausal symptoms, including vaginal atrophy (VVA). Several studies have shown that the symptoms connected with vaginal atrophy are often relieved by estrogen treatment given either systemically or topically. The existing treatments have numerous problems, for example compliance issues with patients not completing or continuing treatment due to the problems associated with the form of treatment.


Accordingly, disclosed herein is, among other things, a new soft gel vaginal pharmaceutical composition and dosage form containing solubilized estradiol for the treatment of VVA. The soft gel vaginal pharmaceutical composition has been designed to mitigate common limitations found with other vaginal forms of estradiol. The soft gel vaginal pharmaceutical composition is expected to ease vaginal administration, provide improved safety of insertion, minimize vaginal discharge following administration, and provide a more effective dosage form with improved efficacy, safety and patient compliance.


SUMMARY

According to various aspects and embodiments of this disclosure, a soft gel vaginal pharmaceutical composition as a potential treatment for post-menopausal women suffering with moderate to severe symptoms of VVA is provided.


Provided herein is a pessary comprising: a) a therapeutically effective amount of estradiol; and b) a solubilizing agent comprising a medium chain oil.


In some embodiments, the pessary comprises about 1 μg to about 25 μg of estradiol. For example, the pessary can include about 1 μg to about 10 μg of estradiol; and about 10 μg to about 25 μg of estradiol.


In some embodiments, the estradiol is solubilized.


In some embodiments, the medium chain oil comprises at least one C6-C12 fatty acid or a glycol, monoglyceride, diglyceride, or triglyceride ester thereof.


In some embodiments, the solubilizing agent comprises at least one ester selected from the group consisting of: an ester of caproic fatty acid, an ester of caprylic fatty acid, an ester of capric fatty acid, and combinations thereof. For example, the solubilizing agent can include a caprylic/capric triglyceride.


In some embodiments, the pessary further comprises a capsule. For example, the capsule can be a soft gelatin capsule.


Also provided herein is a pessary comprising: a) a therapeutically effective amount of estradiol; b) a caprylic/capric triglyceride; c) a non-ionic surfactant comprising PEG-6 palmitostearate and ethylene glycol palmitostearate; and d) a soft gelatin capsule.


In some embodiments, a pessary provided herein comprises about 25 μg of estradiol, wherein administration of the pessary to a patient provides, in a plasma sample from the patient: 1) a corrected geometric mean peak plasma concentration (Cmax) of estradiol of about 19 pg*hr/ml to about 29 pg*hr/ml; and 2) a corrected geometric mean area under the curve (AUC)0-24 of estradiol of about 75 pg*hr/ml to about 112 pg*hr/ml.


In some embodiments, a pessary provided herein comprises about 25 μg of estradiol, wherein administration of the pessary to a patient provides, in a plasma sample from the patient: 1) a corrected geometric mean peak plasma concentration (Cmax) of estrone of about 9 pg*hr/ml to about 14 pg*hr/ml; and 2) a corrected geometric mean area under the curve (AUC)0-24 of estrone of about 43 pg*hr/ml to about 65 pg*hr/ml.


In some embodiments, a pessary provided herein comprises about 25 μg of estradiol, wherein administration of the pessary to a patient provides, in a plasma sample from the patient: 1) a corrected geometric mean peak plasma concentration (Cmax) of estrone sulfate of about 416 pg*hr/ml to about 613 pg*hr/ml; and 2) a corrected geometric mean area under the curve (AUC)0-24 of estrone sulfate of about 3598 pg*hr/ml to about 5291 pg*hr/ml.


In some embodiments, a pessary provided herein comprises about to g of estradiol, wherein administration of the pessary to a patient provides, in a plasma sample from the patient: 1) a corrected geometric mean peak plasma concentration (Cmax) of estradiol of about 12 pg*hr/ml to about 18 pg*hr/ml; and 2) a corrected geometric mean area under the curve (AUC)0-24 of estradiol of about 42 pg*hr/ml to about 63 pg*hr/ml. In some embodiments, the pessary further provides a corrected geometric mean time to peak plasma concentration (Tmax) of estradiol of about 1 hrs to about 3 hrs.


In some embodiments, a pessary provided herein comprises about 10 μg of estradiol, wherein administration of the pessary to a patient provides, in a plasma sample from the patient: 1) a corrected geometric mean peak plasma concentration (Cmax) of estrone of about 4 pg*hr/ml to about 7 pg*hr/ml; and 2) a corrected geometric mean area under the curve (AUC)0-24 of estrone of about 20 pg*hr/ml to about 31 pg*hr/ml. In some embodiments, the pessary further provides a corrected geometric mean time to peak plasma concentration (Tmax) of estrone of about 4 hrs to about 8 hrs.


In some embodiments, a pessary provided herein comprises about 10 μg of estradiol, wherein administration of the pessary to a patient provides, in a plasma sample from the patient: 1) a corrected geometric mean peak plasma concentration (Cmax) of estrone sulfate of about 10 pg*hr/ml to about 16 pg*hr/ml; and 2) a corrected geometric mean area under the curve (AUC)0-24 of estrone sulfate of about 56 pg*hr/ml to about 84 pg*hr/ml. In some embodiments, the pessary further provides a corrected geometric mean time to peak plasma concentration (Tmax) of estrone sulfate of about 4 hrs to about 7 hrs.


In some embodiments, a pessary provided herein comprises about 4 μg of estradiol, wherein administration of the pessary to a patient provides, in a plasma sample from the patient: 1) a corrected geometric mean peak plasma concentration (Cmax) of estradiol of about 4 pg*hr/ml to about 8 pg*hr/ml; and 2) a corrected geometric mean area under the curve (AUC)0-24 of estradiol of about 16 pg*hr/ml to about 26 pg*hr/ml. In some embodiments, the pessary further provides a corrected geometric mean time to peak plasma concentration (Tmax) of estradiol of about 0.25 hrs to about 2 hrs.


In some embodiments, a pessary provided herein comprises about 4 μg of estradiol, wherein administration of the pessary to a patient provides, in a plasma sample from the patient: 1) a corrected geometric mean peak plasma concentration (Cmax) of estrone of about 1 pg*hr/ml to about 3 pg*hr/ml; and 2) a corrected geometric mean area under the curve (AUC)0-24 of estrone of about 8 pg*hr/ml to about 13 pg*hr/ml. In some embodiments, the pessary further provides a corrected geometric mean time to peak plasma concentration (Tmax) of estrone of about 1 hrs to about 4 hrs.


In some embodiments, a pessary provided herein comprises about 4 μg of estradiol, wherein administration of the pessary to a patient provides, in a plasma sample from the patient: 1) a corrected geometric mean peak plasma concentration (Cmax) of estrone sulfate of about 4 pg*hr/ml to about 7 pg*hr/ml; and 2) a corrected geometric mean area under the curve (AUC)0-24 of estrone sulfate of about 22 pg*hr/ml to about 34 pg*hr/ml. In some embodiments, the pessary further provides a corrected geometric mean time to peak plasma concentration (Tmax) of estrone sulfate of about 1 hrs to about 3 hrs.


Also provided herein is a pessary comprising about 1 μg to about 25 μg of estradiol, wherein administration of the pessary to a patient provides a corrected geometric mean peak plasma concentration (Cmax) of estradiol that is less than about 30 pg*hr/ml. For example, administration of the pessary to a patient provides a corrected geometric mean peak plasma concentration (Cmax) of estradiol that is less than about 18 pg*hr/ml.


In some embodiments, a pessary comprising about 1 μg to about 25 μg of estradiol is provided, wherein administration of the pessary to a patient provides a corrected geometric mean area under the curve (AUC)0-24 of estradiol that is less than about 112 pg*hr/ml. For example, administration of the pessary to a patient provides a corrected geometric mean area under the curve (AUC)0-24 of estradiol that is less than about 63 pg*hr/ml.


In some embodiments, a pessary comprising about 1 μg to about 25 μg of estradiol is provided, wherein administration of the pessary to a patient provides a corrected geometric mean peak plasma concentration (Cmax) of estrone that is less than about 14 pg*hr/ml. For example, administration of the pessary to a patient provides a corrected geometric mean peak plasma concentration (Cmax) of estrone that is less than about 7 pg*hr/ml.


In some embodiments, a pessary comprising about 1 μg to about 25 μg of estradiol is provided, wherein administration of the pessary to a patient provides a corrected geometric mean area under the curve (AUC)0-24 of estrone that is less than about 65 pg*hr/ml. For example, administration of the pessary to a patient provides a corrected geometric mean area under the curve (AUC)0-24 of estrone that is less than about 31 pg*hr/ml.


In some embodiments, a pessary comprising about 1 μg to about 25 μg of estradiol is provided, wherein administration of the pessary to a patient provides a corrected geometric mean peak plasma concentration (Cmax) of estrone sulfate that is less than about 613 pg*hr/ml. For example, administration of the pessary to a patient provides a corrected geometric mean peak plasma concentration (Cmax) of estrone sulfate that is less than about 16 pg*hr/ml.


In some embodiments, a pessary comprising about 1 μg to about 25 μg of estradiol is provided, wherein administration of the pessary to a patient provides a corrected geometric mean area under the curve (AUC)0-24 of estrone sulfate that is less than about 5291 pg*hr/ml. For example, administration of the pessary to a patient provides a corrected geometric mean area under the curve (AUC)0-24 of estrone sulfate that is less than about 84 pg*hr/ml.


Further provided herein is a pessary comprising about 1 μg to about 25 μg of estradiol, wherein administration of the pessary to the proximal region of the vagina of a patient provides a therapeutically effective concentration of estradiol over 24 hours in the proximal region of the vagina.


This disclosure also provides a method of treating an estrogen-deficient state, the method comprising administering to a patient in need thereof, a pessary as provided herein. In some embodiments, a method of treating vulvovaginal atrophy is provided, the method comprising administering to a patient in need thereof, a pessary as provided herein.


In some embodiments of the methods provided herein, treatment comprises reducing the severity of one or more symptoms selected from the group consisting of: vaginal dryness, dyspareunia, vaginal or vulvar irritation, vaginal or vulvar burning, vaginal or vulvar itching, dysuria, and vaginal bleeding associated with sexual activity.


In some embodiments of the methods provided herein treatment comprises reducing the vaginal pH of the patient. For example, treatment comprises reducing the vaginal pH of the patient to a pH of less than about 5.0.


In some embodiments of the methods provided herein treatment comprises a change in cell composition of the patient. For example, the change in cell composition comprises reducing the number of parabasal vaginal cells or increasing the number of superficial vaginal cells. In some embodiments, the number of parabasal vaginal cells in the patient are reduced by at least about 35% (e.g., at least about 50%). In some embodiments, the number of superficial vaginal cells are increased by at least about 5% (e.g., at least about 35%).


Further provided herein is a method for reducing vaginal discharge following administration of a pessary, the method comprising administering to a patient in need thereof, a pessary provided herein, wherein the vaginal discharge following administration of the pessary is compared to the vaginal discharge following administration of a reference drug.





DRAWINGS

The above-mentioned features and objects of the this disclosure will become more apparent with reference to the following description taken in conjunction with the accompanying drawings wherein like reference numerals denote like elements and in which:



FIG. 1 is a flow diagram illustrating a process in accordance with various embodiments of the invention;



FIG. 2 illustrates a suppository in accordance with various embodiments of the invention;



FIG. 3 is a linear plot of mean plasma estradiol—baseline adjusted concentrations versus time (N=36);



FIG. 4 is a semi-logarithmic plot of mean plasma estradiol—baseline adjusted concentrations versus time (N=36);



FIG. 5 is a linear plot of mean plasma estrone—baseline adjusted concentrations versus time (N=36);



FIG. 6 is a semi-logarithmic plot of mean plasma estrone—baseline adjusted concentrations versus time (N=36);



FIG. 7 is a linear plot of mean plasma estrone sulfate—baseline adjusted concentrations versus time (N=36);



FIG. 8 is a semi-logarithmic plot of mean plasma estrone sulfate—baseline adjusted concentrations versus time (N=36);



FIG. 9 is a linear plot of mean plasma estradiol—baseline adjusted concentrations versus time (N=34);



FIG. 10 is a semi-logarithmic plot of mean plasma estradiol—baseline adjusted concentrations versus time (N=34);



FIG. 11 is a linear plot of mean plasma estrone—baseline adjusted concentrations versus time (N=33);



FIG. 12 is a semi-logarithmic plot of mean plasma estrone—baseline adjusted concentrations versus time (N=33);



FIG. 13 is a linear plot of mean plasma estrone sulfate—baseline adjusted concentrations versus time (N=24); and



FIG. 14 is a semi-logarithmic plot of mean plasma estrone sulfate—baseline adjusted concentrations versus time (N=24).





DETAILED DESCRIPTION

In the following detailed description of embodiments of this disclosure, reference is made to the accompanying drawings in which like references indicate similar elements, and in which is shown by way of illustration specific embodiments in which the this disclosure may be practiced. These embodiments are described in sufficient detail to enable those skilled in the art to practice the this disclosure, and it is to be understood that other embodiments may be utilized and that other changes may be made without departing from the scope of the this disclosure. The following detailed description is, therefore, not to be taken in a limiting sense, and the scope of this disclosure is defined only by the appended claims. As used in this disclosure, the term “or” shall be understood to be defined as a logical disjunction (i.e., and/or) and shall not indicate an exclusive disjunction unless expressly indicated as such with the terms “either,” “unless,” “alternatively,” and words of similar effect.


Definitions

The term “active pharmaceutical ingredient” (“API”) as used herein, means the active compound(s) used in formulating a drug product.


The term “co-administered” as used herein, means that two or more drug products are administered simultaneously or sequentially on the same or different days.


The term “drug product” as used herein means at least one active pharmaceutical ingredient in combination with at least one excipient and provided in unit dosage form.


The term “area under the curve” (“AUC”) refers to the area under the curve defined by changes in the blood concentration of an active pharmaceutical ingredient (e.g., estradiol or progesterone), or a metabolite of the active pharmaceutical ingredient, over time following the administration of a dose of the active pharmaceutical ingredient. “AUC0-∞” is the area under the concentration-time curve extrapolated to infinity following the administration of a dose. “AUC0-t” is the area under the concentration-time curve from time zero to time t following the administration of a dose, wherein t is the last time point with a measurable concentration.


The term “Cmax” refers to the maximum value of blood concentration shown on the curve that represents changes in blood concentrations of an active pharmaceutical ingredient (e.g., progesterone or estradiol), or a metabolite of the active pharmaceutical ingredient, over time.


The term “Tmax” refers to the time that it takes for the blood concentration an active pharmaceutical ingredient (e.g., estradiol or progesterone), or a metabolite of the active pharmaceutical ingredient, to reach the maximum value.


The term “bioavailability,” which has the meaning defined in 21 C.F.R. § 320.1(a), refers to the rate and extent to which an API or active ingredient or active moiety is absorbed from a drug product and becomes available at the site of action. For example, bioavailability can be measured as the amount of API in the blood (serum or plasma) as a function of time. Pharmacokinetic (PK) parameters such as AUC, Cmax, or Tmax may be used to measure and assess bioavailability. For drug products that are not intended to be absorbed into the bloodstream, bioavailability may be assessed by measurements intended to reflect the rate and extent to which the API or active ingredient or active moiety becomes available at the site of action.


The term “bioequivalent,” which has the meaning defined in 21 C.F.R. § 320.1(e), refers to the absence of a significant difference in the rate and extent to which the API or active ingredient or active moiety in pharmaceutical equivalents or pharmaceutical alternatives becomes available at the site of drug action when administered at the same molar dose under similar conditions in an appropriately designed study. Where there is an intentional difference in rate (e.g., in certain extended release dosage forms), certain pharmaceutical equivalents or alternatives may be considered bioequivalent if there is no significant difference in the extent to which the active ingredient or moiety from each product becomes available at the site of drug action. This applies only if the difference in the rate at which the active ingredient or moiety becomes available at the site of drug action is intentional and is reflected in the proposed labeling, is not essential to the attainment of effective body drug concentrations on chronic use, and is considered medically insignificant for the drug. In practice, two products are considered bioequivalent if the 90% confidence interval of the AUC, Cmax, or optionally Tmax is within 80.00% to 125.00%.


The term “bio-identical,” “body-identical,” or “natural” used in conjunction with the hormones disclosed herein, means hormones that match the chemical structure and effect of those that occur naturally or endogenously in the human body. An exemplary natural estrogen is estradiol.


The term “bio-identical hormone” or “body-identical hormone” refers to an active pharmaceutical ingredient that is structurally identical to a hormone naturally or endogenously found in the human body (e.g., estradiol and progesterone).


The term “estradiol” refers to (17β)-estra-1,3,5(10)-triene-3,17-diol. Estradiol is also interchangeably called 17β-estradiol, oestradiol, or E2, and is found endogenously in the human body. As used herein, estradiol refers to the bio-identical or body-identical form of estradiol found in the human body having the structure:




embedded image


Estradiol is supplied in an anhydrous or hemi-hydrate form. For the purposes of this disclosure, the anhydrous form or the hemihydrate form can be substituted for the other by accounting for the water or lack of water according to well-known and understood techniques.


The term “solubilized estradiol” means that the estradiol or a portion thereof is solubilized or dissolved in the solubilizing agent(s) or the formulations disclosed herein. Solubilized estradiol may include estradiol that is about 80% solubilized, about 85% solubilized, about 90% solubilized, about 95% solubilized, about 96% solubilized, about 97% solubilized, about 98% solubilized, about 99% solubilized or about 100% solubilized. In some embodiments, the estradiol is “fully solubilized” with all or substantially all of the estradiol being solubilized or dissolved in the solubilizing agent. Fully solubilized estradiol may include estradiol that is about 97% solubilized, about 98% solubilized, about 99% solubilized or about 100% solubilized. Solubility can be expressed as a mass fraction (% w/w, which is also referred to as wt %).


The term “progesterone” refers to pregn-4-ene-3,20-dione. Progesterone is also interchangeably called P4 and is found endogenously in the human body. As used herein, progesterone refers to the bio-identical or body-identical form of progesterone found in the human body having the structure:




embedded image


The term “solubilized progesterone” means that the progesterone or a portion thereof is solubilized or dissolved in the solubilizing agent(s) or the formulations disclosed herein. In some embodiments, the progesterone is “partially solubilized” with a portion of the progesterone being solubilized or dissolved in the solubilizing agent and a portion of the progesterone being suspended in the solubilizing agent. Partially solubilized progesterone may include progesterone that is about 1% solubilized, about 5% solubilized, about 10% solubilized, about 15% solubilized, about 20% solubilized, about 30% solubilized, about 40% solubilized, about 50% solubilized, about 60% solubilized, about 70% solubilized, about 80% solubilized, about 85% solubilized, about 90% solubilized or about 95% solubilized. In other embodiments, the progesterone is “fully solubilized” with all or substantially all of the progesterone being solubilized or dissolved in the solubilizing agent. Fully solubilized progesterone may include progesterone that is about 97% solubilized, about 98% solubilized, about 99% solubilized or about 100% solubilized. Solubility can be expressed as a mass fraction (% w/w, which is also referred to as wt %).


The terms “micronized progesterone” and “micronized estradiol,” as used herein, include micronized progesterone and micronized estradiol having an X50 particle size value below about 15 microns or having an X90 particle size value below about 25 microns. The term “X50” means that one-half of the particles in a sample are smaller in diameter than a given number. For example, micronized progesterone having an X50 of 5 microns means that, for a given sample of micronized progesterone, one-half of the particles have a diameter of less than 5 microns. Similarly, the term “X90” means that ninety percent (90%) of the particles in a sample are smaller in diameter than a given number.


The term “glyceride” is an ester of glycerol (1,2,3-propanetriol) with acyl radicals of fatty acids and is also known as an acylglycerol. If only one position of the glycerol molecule is esterified with a fatty acid, a “monoglyceride” or “monoacylglycerol” is produced; if two positions are esterified, a “diglyceride” or “diacylglycerol” is produced; and if all three positions of the glycerol are esterified with fatty acids, a “triglyceride” or “triacylglycerol” is produced. A glyceride is “simple” if all esterified positions contain the same fatty acid; whereas a glyceride is “mixed” if the esterified positions contained different fatty acids. The carbons of the glycerol backbone are designated sn-1, sn-2 and sn-3, with sn-2 being in the middle carbon and sn-1 and sn-3 being the end carbons of the glycerol backbone.


The term “solubilizing agent” refers to an agent or combination of agents that solubilize an active pharmaceutical ingredient (e.g., estradiol or progesterone). For example and without limitation, suitable solubilizing agents include medium chain oils and other solvents and co-solvents that solubilize or dissolve an active pharmaceutical ingredient to a desirable extent. Solubilizing agents suitable for use in the formulations disclosed herein are pharmaceutical grade solubilizing agents (e.g., pharmaceutical grade medium chain oils). It will be understood by those of skill in the art that other excipients or components can be added to or mixed with the solubilizing agent to enhance the properties or performance of the solubilizing agent or resulting formulation. Examples of such excipients include, but are not limited to, surfactants, emulsifiers, thickeners, colorants, flavoring agents, etc. In some embodiments, the solubilizing agent is a medium chain oil and, in some other embodiments, the medium chain oil is combined with a co-solvent(s) or other excipient(s).


The term “medium chain” is used to describe the aliphatic chain length of fatty acid containing molecules. “Medium chain” specifically refers to fatty acids, fatty acid esters, or fatty acid derivatives that contain fatty acid aliphatic tails or carbon chains that contain 6 (C6) to 14 (C14) carbon atoms, 8 (C8) to 12 (C12) carbon atoms, or 8 (C8) to 10 (C10) carbon atoms.


The terms “medium chain fatty acid” and “medium chain fatty acid derivative” are used to describe fatty acids or fatty acid derivatives with aliphatic tails (i.e., carbon chains) having 6 to 14 carbon atoms. Fatty acids consist of an unbranched or branched aliphatic tail attached to a carboxylic acid functional group. Fatty acid derivatives include, for example, fatty acid esters and fatty acid containing molecules, including, without limitation, mono-, di- and triglycerides that include components derived from fatty acids. Fatty acid derivatives also include fatty acid esters of ethylene or propylene glycol. The aliphatic tails can be saturated or unsaturated (i.e., having one or more double bonds between carbon atoms). In some embodiments, the aliphatic tails are saturated (i.e., no double bonds between carbon atoms). Medium chain fatty acids or medium chain fatty acid derivatives include those with aliphatic tails having 6-14 carbons, including those that are C6-C14, C6-C12, C8-C14, C8-C12, C6-C10, C8-C10, or others. Examples of medium chain fatty acids include, without limitation, caproic acid, caprylic acid, capric acid, lauric acid, myristic acid, and derivatives thereof.


The term “oil,” as used herein, refers to any pharmaceutically acceptable oil, especially medium chain oils, and specifically excluding peanut oil, that can suspend or solubilize bioidentical progesterone or estradiol, including starting materials or precursors thereof, including micronized progesterone or micronized estradiol as described herein.


The term “medium chain oil” refers to an oil wherein the composition of the fatty acid fraction of the oil is substantially medium chain (i.e., C6 to C14) fatty acids, i.e., the composition profile of fatty acids in the oil is substantially medium chain. As used herein, “substantially” means that between 20% and 100% (inclusive of the upper and lower limits) of the fatty acid fraction of the oil is made up of medium chain fatty acids, i.e., fatty acids with aliphatic tails (i.e., carbon chains) having 6 to 14 carbons. In some embodiments, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 85%, about 90% or about 95% of the fatty acid fraction of the oil is made up of medium chain fatty acids. Those of skill in the art that will readily appreciate that the terms “alkyl content” or “alkyl distribution” of an oil can be used in place of the term “fatty acid fraction” of an oil in characterizing a given oil or solubilizing agent, and these terms are used interchangeable herein. As such, medium chain oils suitable for use in the formulations disclosed herein include medium chain oils wherein the fatty acid fraction of the oil is substantially medium chain fatty acids, or medium chain oils wherein the alkyl content or alkyl distribution of the oil is substantially medium chain alkyls (C6-C12 alkyls). It will be understood by those of skill in the art that the medium chain oils suitable for use in the formulations disclosed herein are pharmaceutical grade (e.g., pharmaceutical grade medium chain oils). Examples of medium chain oils include, for example and without limitation, medium chain fatty acids, medium chain fatty acid esters of glycerol (e.g., for example, mono-, di-, and triglycerides), medium chain fatty acid esters of propylene glycol, medium chain fatty acid derivatives of polyethylene glycol, and combinations thereof.


The term “ECN” or “equivalent carbon number” means the sum of the number of carbon atoms in the fatty acid chains of an oil, and can be used to characterize an oil as, for example, a medium chain oil or a long-chain oil. For example, tripalmitin (tripalmitic glycerol), which is a simple triglyceride containing three fatty acid chains of 16 carbon atoms, has an ECN of 3×16=48. Conversely, a triglyceride with an ECN=40 may have “mixed” fatty acid chain lengths of 8, 16 and 16; 10, 14 and 16; 8, 14 and 18; etc. Naturally occurring oils are frequently “mixed” with respect to specific fatty acids, but tend not to contain both long chain fatty acids and medium chain fatty acids in the same glycerol backbone. Thus, triglycerides with ECN's of 21-42 typically contain predominately medium chain fatty acids; while triglycerides with ECN's of greater than 43 typically contain predominantly long chain fatty acids. For example, the ECN of corn oil triglyceride in the USP would be in the range of 51-54. Medium chain diglycerides with ECN's of 12-28 will often contain predominately medium chain fatty chains, while diglycerides with ECN's of 32 or greater will typically contain predominately long chain fatty acid tails. Monoglycerides will have an ECN that matches the chain length of the sole fatty acid chain. Thus, monoglyceride ECN's in the range of 6-14 contain mainly medium chain fatty acids, and monoglycerides with ECN's 16 or greater will contain mainly long chain fatty acids.


The average ECN of a medium chain triglyceride oil is typically 21-42. For example, as listed in the US Pharmacopeia (USP), medium chain triglycerides have the following composition as the exemplary oil set forth in the table below:














Fatty-acid Tail Length
% of oil
Exemplary Oil

















6
≤2.0
2.0


8
50.0-80.0
70.0


10
20.0-50.0
25.0


12
≤3.0
2.0


14
≤1.0
1.0










and would have an average ECN of 3*[(6*0.02)+(8*0.70)+(10*0.25)+(12*0.02)+(14*0.01)]=25.8. The ECN of the exemplary medium chain triglycerides oil can also be expressed as a range (per the ranges set forth in the USP) of 24.9-27.0. For oils that have mixed mono-, di-, and trigylcerides, or single and double fatty acid glycols, the ECN of the entire oil can be determined by calculating the ECN of each individual component (e.g., C8 monoglycerics, C8 diglycerides, C10 monoglycerides, and C10 monoglycerides) and taking the sum of the relative percentage of the component multiplied by the ECN normalized to a monoglyceride for each component. For example, the oil having C8 and C10 mono- and diglycerides shown in the table below has an ECN of 8.3, and is thus a medium chain oil.

















ECN as % of oil
ECN as % of oil


Fatty-acid
% of
(chain length) ×
normalized to


ChainLength
oil
(% in oil)
monoglyceride


















C8 monoglyceride
47
    8 × 0.47 = 3.76
 3.76


C10 monoglyceride
8
  10 × 0.08 = 0.8
0.8


C8 diglyceride
38
2 × (8 × 0.38) = 6.08
6.08/2 = 3.04


C10 diglyceride
7
2 × (10 × 0.07) = 1.4 
1.4/2 = 0.7


OIL ECN


8.3


(normalized to





monoglycerides)










Expressed differently, ECN can be calculated as each chain length in the composition multiplied by its relative percentage in the oil: (8*0.85)+(10*0.15)=8.3.


The term “excipients,” as used herein, refers to non-API ingredients such as solubilizing agents, anti-oxidants, oils, lubricants, and others used in formulating pharmaceutical products.


The term “patient” or “subject” refers to an individual to whom the pharmaceutical composition is administered.


The term “pharmaceutical composition” refers to a pharmaceutical composition comprising at least a solubilizing agent and estradiol. As used herein, pharmaceutical compositions are delivered, for example via pessary (i.e., vaginal suppository), or absorbed vaginally.


The term “progestin” means any natural or man-made substance that has pharmacological properties similar to progesterone.


The term “reference listed drug product” (“RLD”) means VAGIFEM® (estradiol vaginal tablets) or ESTRACE® vaginal cream.


The terms “treat,” “treating,” and “treatment” refer to any indicia of success in the treatment or amelioration of an injury, disease, or condition, including any objective or subjective parameter such as abatement; remission; diminishing of symptoms or making the injury, disease, or condition more tolerable to the patient; slowing in the rate of degeneration or decline; or improving a patient's physical or mental well-being. The treatment or amelioration of symptoms can be based on objective or subject parameters, including the results of a physical examination, neuropsychiatric examinations, or psychiatric evaluation.


The terms “atrophic vaginitis,” “vulvovaginal atrophy,” “vaginal atrophy,” and “VVA” are used herein interchangeably. The molecular morphology of VVA is well known in the medical field.


INTRODUCTION

Provided herein are pharmaceutical compositions comprising solubilized estradiol designed to be absorbed vaginally. The pharmaceutical compositions disclosed herein are designed to be absorbed and have their therapeutic effect locally, e.g., in vaginal or surrounding tissue. Further disclosed herein are data demonstrating efficacy of the pharmaceutical compositions disclosed, as well as methods relating to the pharmaceutical compositions. Generally, the pharmaceutical compositions disclosed herein are useful in VVA, dysparuenia, and other indications caused by decrease or lack of estrogen.


Additional aspects and embodiments of this disclosure include: providing increased patient ease of use while potentially minimizing certain side effects from inappropriate insertion, minimizing incidence of vulvovaginal mycotic infection compared to incidence of vulvovaginal mycotic infection due to usage of other vaginally applied estradiol products; and, improved side effect profile (e.g., pruritus) compared to the reference drug: VAGIFEM® (estradiol vaginal tablets, Novo Nordisk; Princeton, N.J.).


Pharmaceutical Composition

Functionality


According to embodiments, the pharmaceutical compositions disclosed herein are alcohol-free or substantially alcohol-free. The pharmaceutical compositions offer provide for improved patient compliance because of improvements over the prior offering. According to embodiments, the pharmaceutical compositions disclosed herein are encapsulated in soft gelatin capsules, which improve comfort during use. According to embodiments, the pharmaceutical compositions are substantially liquid, which are more readily absorbed in the vaginal tissue, and also are dispersed over a larger surface area of the vaginal tissue.


Estradiol


According to embodiments, the pharmaceutical compositions disclosed herein are for vaginal insertion in a single or multiple unit dosage form. According to embodiments, the estradiol in the pharmaceutical compositions is at least about: 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% solubilized. According to embodiments and where the estradiol is not 100% solubilized, the remaining estradiol is present in a micronized (crystalline) form that is absorbable by the body and retains biological functionality, either in its micronized form or in another form which the micronized form is converted to after administration.


According to embodiments, all or some of the estradiol is solubilized in a solubilizing agent during manufacturing process. According to embodiments, all or some of the estradiol is solubilized following administration (e.g., the micronized portion where the estradiol is not 100% solubilized is solubilized in a body fluid after administration). According to embodiments, because the estradiol is solubilized, the solubilizing agents taught herein, with or without additional excipients other than the solubilizing agents, are liquid or semi-solid. To the extent the estradiol is not fully solubilized at the time of administration/insertion, the estradiol should be substantially solubilized at a body temperature (average of 37° C.) and, generally, at the pH of the vagina (ranges from 3.8 to 4.5 in healthy patients; and 4.6 to 6.5 in VVA patients).


According to embodiments, the estradiol can be added to the pharmaceutical compositions disclosed herein as estradiol, estradiol hemihydrate, or other grade estradiol forms used in pharmaceutical compositions or formulations.


According to embodiments, estradiol dosage strengths vary. Estradiol (or estradiol hemihydrate, for example, to the extent the water content of the estradiol hemihydrate is accounted for) dosage strength of is from at least about 1 microgram (μg or μg) to at least about 50 μg. Specific dosage embodiments contain at least about: 1 μg, 2 μg, 3 μg, 4 μg, 5 μg, 6 μg, 7 μg, 8 μg, 9 μg, 10 μg, 11 μg, 12 μg, 13 μg, 14 μg, 15 μg, 16 μg, 17 μg, 18 μg, 19 μg, 20 μg, 21 μg, 22 μg, 23 μg, 24 μg, 25 μg, 26 μg, 27 μg, 28 μg, 29 μg, 30 μg, 31 μg, 32 μg, 33 μg, 34 μg, 35 μg, 36 μg, 37 μg, 38 μg, 39 μg, 40 μg, 41 μg, 42 μg, 43 μg, 44 μg, 45 μg, 46 μg, 47 μg, 48 μg, 49 μg, or 50 μg estradiol. According to embodiments, the pharmaceutical compositions contain at least about 2.5 μg; 4 μg 6.25 μg, 7.5 μg, 12.5 μg, 18.75 μg of estradiol. According to embodiments, the pharmaceutical compositions contain from about 1 μg to about 10 μg, from 3 μg to 7 μg, from about 7.5 μg to 12.5 μg, from about 10 μg to about 25 μg, about 1 μg, about 2.5 μg, from about 23.5 μg to 27.5 μg, from about 7.5 μg to 22.5 μg, from 10 μg to 25 μg of estradiol. The lowest clinically effective dose of estradiol is used for treatment of VVA and other indications set forth herein. In some embodiments, the estradiol dosage is about 4 μg. In one embodiment, the estradiol dosage is about 10 μg. In another embodiment, the estradiol dosage is about 25 μg.


Solvent System


According to embodiments, the solvent system that solubilizes the estradiol are medium chain fatty acid based solvents, together with other excipients. According to embodiments, the solvent system comprises non-toxic, pharmaceutically acceptable solvents, co-solvents, surfactants, and other excipients suitable for vaginal delivery or absorption.


According to embodiments, oils having medium chain fatty acids as a majority component are used as solubilizing agents to solubilize estradiol. According to embodiments, the solubilizing agents comprise medium chain fatty acid esters (e.g., esters of glycerol, ethylene glycol, or propylene glycol) or mixtures thereof. According to embodiments, the medium chain fatty acids comprise chain lengths from C6 to C14. According to embodiments the medium chain fatty acids comprise chain lengths from C6 to C12. According to embodiments the medium chain fatty acids substantially comprise chain lengths from C8-C10. ECN's for medium chain oils will be in the range of 21-42 for triglycerides, 12-28 for diglycerides, and 6-14 for monoglycerides.


According to embodiments, the medium chain fatty acids are saturated. According to embodiments, the medium chain fatty acids are predominantly saturated, i.e., greater than about 60% or greater than about 75% saturated.


According to embodiments, estradiol is soluble in the solubilizing agent at room temperature, although it may be desirable to warm certain solubilizing agents during manufacture to improve viscosity. According to embodiments, the solubilizing agent is liquid at between room temperature and about 50° C., at or below 50° C., at or below 40° C., or at or below 30° C.


According to embodiments, the solubility of estradiol in the medium chain oil, medium chain fatty acid, or solubilizing agent (or oil/surfactant) is at least about 0.01 wt %, 0.02 wt %, 0.05 wt %, 0.06 wt %, 0.08 wt %, 0.1 wt %, 0.2 wt %, 0.3 wt %, 0.4 wt %, 0.5 wt %, 0.6 wt %, 0.7 wt %, 0.8 wt %, 0.9 wt %, 1.0 wt %, or higher.


According to embodiments, medium chain solubilizing agents include, for example and without limitation saturated medium chain fatty acids: caproic acid (C6), enanthic acid (C7), caprylic acid (C8), pelargonic acid (C9), capric acid (C10), undecylic acid (C11), lauric acid (C12), tridecylic acid (C13), or myristic acid (C14). According to embodiments, the solubilizing agent comprises oils made of these free medium chain fatty acids, oils of medium chain fatty acid esters of glycerin, propylene glycol, or ethylene glycol, or combinations thereof. These examples comprise predominantly saturated medium chain fatty acids (i.e., greater than 50% of the fatty acids are medium chain saturated fatty acids). According to embodiments, predominantly C6 to C12 saturated fatty acids are contemplated. According to embodiments, the solubilizing agent is selected from at least one of a solvent or co-solvent.


According to embodiments, glycerin based solubilizing agents include: mono-, di-, or triglycerides and combinations and derivatives thereof. Exemplary glycerin based solubilizing agents include MIGLYOLs®, which are caprylic/capric triglycerides (SASOL Germany GMBH, Hamburg). MIGLYOLs includes MIGLYOL 810 (caprylic/capric triglyceride), MIGLYOL 812 (caprylic/capric triglyceride), MIGLYOL 816 (caprylic/capric triglyceride), and MIGLYOL 829 (caprylic/capric/succinic triglyceride). Other caprylic/capric triglyceride solubilizing agents are likewise contemplated, including, for example: caproic/caprylic/capric/lauric triglycerides; caprylic/capric/linoleic triglycerides; caprylic/capric/succinic triglycerides. According to embodiments, CAPMUL MCM, medium chain mono- and di-glycerides, is the solubilizing agent. Other and triglycerides of fractionated vegetable fatty acids, and combinations or derivatives thereof can be the solubilizing agent, according to embodiments. For example, the solubilizing agent can be 1,2,3-propanetriol (glycerol, glycerin, glycerine) esters of saturated coconut and palm kernel oil and derivatives thereof.


Ethylene and propylene glycols (which include polyethylene and polypropylene glycols) solubilizing agents include: glyceryl mono- and di-caprylates; propylene glycol monocaprylate (e.g., CAPMUL® PG-8 (the CAPMUL brands are owned by ABITEC, Columbus, Ohio)); propylene glycol monocaprate (e.g., CAPMUL PG-10); propylene glycol mono- and dicaprylates; propylene glycol mono- and dicaprate; diethylene glycol mono ester (e.g., TRANSCUTOL®, 2-(2-Ethoxyethoxy)ethanol, GATIEFOSSÉ SAS); and diethylene glycol monoethyl ether. Other combinations of mono- and di-esters of propylene glycol or ethylene glycol are expressly contemplated are the solubilizing agent.


According to embodiments, the solubilizing agent comprises combinations of mono- and di-propylene and ethylene glycols and mono-, di-, and triglyceride combinations. According to embodiments, polyethylene glycol glyceride (GELUCIRE®, GATIEFOSSÉ SAS, Saint-Priest, France) can be used herein as the solubilizing agent or as a surfactant. For example, GELUCIRE 44/14 (PEG-32 glyceryl laurate EP), a medium chain fatty acid esters of polyethylene glycol, is a polyethylene glycol glyceride composed of mono-, di- and triglycerides and mono- and diesters of polyethylene glycol.


According to embodiments, commercially available fatty acid glycerol and glycol ester solubilizing agents are often prepared from natural oils and therefore may comprise components in addition to the fatty acid esters that predominantly comprise and characterize the solubilizing agent. Such other components may be, e.g., other fatty acid mono-, di-, and triglycerides; fatty acid mono- and diester ethylene or propylene glycols, free glycerols or glycols, or free fatty acids, for example. In some embodiments, when an oil/solubilizing agent is described herein as a saturated C8 fatty acid mono- or diester of glycerol, the predominant component of the oil, i.e., >50 wt % (e.g., >75 wt %, >85 wt % or >90 wt %) is caprylic monoglycerides and caprylic diglycerides. For example, the Technical Data Sheet by ABITEC for CAPMUL MCM C8 describes CAPMUL MCM C8 as being composed of mono and diglycerides of medium chain fatty acids (mainly caprylic) and describes the alkyl content as ≤1% C6, ≥95% C8, ≤5% C10, and ≤1.5% C12 and higher.


For example, MIGLYOL 812 is a solubilizing agent that is generally described as a C8-C10 triglyceride because the fatty acid composition is at least about 80% triglyceride esters of caprylic acid (C8) and capric acid (C10). However, it also comprises small amounts of other fatty acids, e.g., less than about 5% of caproic acid (C6), lauric acid (C12), and myristic acid (C14). The product information sheet for various MIGLYOLs illustrate the various fatty acid components as follows:

















Tests
810
812
818
829
840







Caproic acid
max. 2.0
max. 2.0
max. 2
max. 2
max. 2


(C6:0)







Caprylic acid
65.0-80.0
50.0-65.0
45-65
45-55
65-80


(C8:0)







Capric acid
20.0-35.0
30.0-45.0
30-45
30-40
20-35


(C10:0)







Lauric acid
max. 2  
max. 2  
max. 3
max. 3
max. 2


(C12:0)







Myristic acid
max. 1.0
max. 1.0
max. 1
max. 1
max. 1


(C14:0)







Linoleic acid


2-5




(C18:2)







Succinic acid



15-20



ECN
25.5-26.4
26.1-27  
26.52-28.56
26-27.6
25.5-26.4









According to embodiments, anionic or non-ionic surfactants may be used in pharmaceutical compositions containing solubilized estradiol. Ratios of solubilizing agent(s) to surfactant(s) vary depending upon the respective solubilizing agent(s) and the respective surfactant(s) and the desired physical characteristics of the resultant pharmaceutical composition. For example and without limitation, CAPMUL MCM and a non-ionic surfactant may be used at ratios including 65:35, 70:30, 75:25, 80:20, 85:15 and 90:10. Other non-limiting examples include: CAPMUL MCM and GELUCIRE 39/01 used in ratios including, for example and without limitation, 6:4, 7:3, and 8:2; CAPMUL MCM and GELUCIRE 43/01 used in ratios including, for example and without limitation, 7:3, and 8:2; CAPMUL MCM and GELUCIRE 50/13 used in ratios including, for example and without limitation, 7:3, and 8:2, and 9:1.


Other Excipients


According to embodiments, the pharmaceutical composition further comprises a surfactant. The surfactant can be a nonionic surfactant, cationic surfactant, anionic surfactant, or mixtures thereof. Suitable surfactants include, for example, water-insoluble surfactants having a hydrophilic-lipophilic balance (HLB) value less than 12 and water-soluble surfactants having a HLB value greater than 12. Surfactants that have a high HLB and hydrophilicity, aid the formation of oil-water droplets. The surfactants are amphiphilic in nature and are capable of dissolving or solubilizing relatively high amounts of hydrophobic drug compounds.


Non-limiting examples, include, Tween, Dimethylacetamide (DMA), Dimethyl sulfoxide (DMSO), Ethanol, Glycerin, N-methyl-2-pyrrolidone (NMP), PEG 300, PEG 400, Poloxamer 407, Propylene glycol, Phospholipids, Hydrogenated soy phosphatidylcholine (HSPC), Distearoylphosphatidylglycerol (DSPG), L-α-dimyristoylphosphatidylcholine (DMPC), L-α-dimyristoylphosphatidylglycerol (DMPG), Polyoxyl 35 castor oil (CREMOPHOR EL, CREMOPHOR ELP), Polyoxyl 40 hydrogenated castor oil (Cremophor RH 40), Polyoxyl 60 hydrogenated castor oil (CREMOPHOR RH 60), Polysorbate 20 (TWEEN 20), Polysorbate 80 (TWEEN 80), d-α-tocopheryl polyethylene glycol 1000 succinate (TPGS), Solutol HS-15, Sorbitan monooleate (SPAN 20), PEG 300 caprylic/capric glycerides (SOFTIGEN 767), PEG 400 caprylic/capric glycerides (LABRASOL), PEG 300 oleic glycerides (LABRAFIL M-1944CS), Polyoxyl 35 Castor oil (ETOCAS 35), Glyceryl Caprylate (Mono- and Diglycerides) (IMWITOR), PEG 300 linoleic glycerides (LABRAFIL M-2125CS), Polyoxyl 8 stearate (PEG 400 monosterate), Polyoxyl 40 stearate (PEG 1750 monosterate), and combinations thereof. Additionally, suitable surfactants include, for example, polyoxyethylene derivative of sorbitan monolaurate such as polysorbate, caprylcaproyl macrogol glycerides, polyglycolyzed glycerides, and the like.


According to embodiments, the non-ionic surfactant is selected from one or more of glycerol and polyethylene glycol esters of long chain fatty acids, for example, lauroyl macrogol-32 glycerides or lauroyl polyoxyl-32 glycerides, commercially available as GELUCIRE, including, for example, GELUCIRE 39/01 (glycerol esters of saturated C12-C18 fatty acids), GELUCIRE 43/01 (hard fat NF/JPE) and GELUCIRE 50/13 (stearoyl macrogol-32 glycerides EP, stearoyl polyoxyl-32 glycerides NF, stearoyl polyoxylglycerides (USA FDA IIG)). These surfactants may be used at concentrations greater than about 0.01%, and typically in various amounts of about 0.01%-10.0%, 10.1%-20%, and 20.1%-30%. In some embodiments, surfactants may be used at concentrations of about 1% to about 10% (e.g., about 1% to about 5%, about 2% to about 4%, about 3% to about 8%).


According to embodiments, non-ionic surfactants include, for example and without limitation: one or more of oleic acid, linoleic acid, palmitic acid, and stearic acid. According to embodiments, non-ionic surfactants comprise polyethylene sorbitol esters, including polysorbate 80, which is commercially available under the trademark TWEEN® 80 (polysorbate 80) (Sigma Aldrich, St. Louis, Mo.). Polysorbate 80 comprises approximately 60%-70% oleic acid with the remainder comprising primarily linoleic acids, palmitic acids, and stearic acids. Polysorbate 80 may be used in amounts ranging from about 5 to 50%, and according to embodiments, about 30% of the pharmaceutical composition total mass.


According to embodiments, the non-ionic surfactant includes PEG-6 palmitostearate and ethylene glycol palmitostearate, which are available commercially as TEFOSE® 63 (GATTEFOSSÉ SAS, Saint-Priest, France), which can be used with, for example, CAPMUL MCM having ratios of MCM to TEFOSE 63 of, for example, 8:2 or 9:1. According to embodiments, other solubilizing agents/non-ionic surfactants combinations include, for example, MIGLYOL 812:GELUCIRE 50/13 or MIGLYOL 812:TEFOSE 63.


According to embodiments, the surfactant can be an anionic surfactant, for example: ammonium lauryl sulfate, dioctyl sodium sulfosuccinate, perfluoro-octane sulfonic acid, potassium lauryl sulfate, or sodium stearate. Cationic surfactants are also contemplated.


According to embodiments, non-ionic or anionic surfactants can be used alone with at least one solubilizing agent or can be used in combination with other surfactants. Accordingly, such surfactants, or any other excipient as set forth herein, may be used to solubilize estradiol. The combination of solubilizing agent, surfactant, and other excipients should be designed whereby the estradiol is absorbed into the vaginal tissue. According to embodiments, the pharmaceutical composition will result in minimal vaginal discharge.


According to embodiments, the pharmaceutical composition further comprises at least one thickening agent. Generally, a thickening agent is added when the viscosity of the pharmaceutical composition results less than desirable absorption. According to embodiments, the surfactant(s) disclosed herein may also provide thickening of the pharmaceutical composition that, upon release, will aid the estradiol in being absorbed by the vaginal mucosa while minimizing vaginal discharge. Examples of thickening agents include: hard fats; propylene glycol; a mixture of hard fat EP/NF/JPE, glyceryl ricinoleate, ethoxylated fatty alcohols (ceteth-20, steareth-20) EP/NF (available as OVUCIRE® 3460, GATTEFOSSÉ, Saint-Priest, France); a mixture of hard fat EP/NF/JPE, glycerol monooleate (type 40) EP/NF (OVUCIRE WL 3264; a mixture of hard fat EP/NF/JPE, glyceryle monooleate (type 40) EP/NF (OVUCIRE WL 2944); a non-ionic surfactant comprising PEG-6 stearate, ethylene glycol palmitostearate, and PEG-32 stearate; TEFOSE 63 or a similar product; and a mixture of various hard fats (WITEPSOL®, Sasol Germany GmbH, Hamburg, Germany). Other thickening agents such as the alginates, certain gums such as xanthan gums, agar-agar, iota carrageenans, kappa carrageenans, etc. Several other compounds can act as thickening agents like gelatin, and polymers like HPMC, PVC, and CMC. According to embodiments, the viscosity of pharmaceutical compositions in accordance with various embodiments may comprise from about 50 cps to about 1000 cps at 25° C. A person of ordinary skill in the art will readily understand and select from suitable thickening agents.


According to embodiments, the thickening agent is a non-ionic surfactant. For example, polyethylene glycol saturated or unsaturated fatty acid ester or diester is the non-ionic surfactant thickening agent. In embodiments, the non-ionic surfactant comprises a polyethylene glycol long chain (C16-C20) fatty acid ester and further comprises an ethylene glycol long chain fatty acid ester, such as PEG-fatty acid esters or diesters of saturated or unsaturated C16-C18 fatty acids, e.g., oleic, lauric, palmitic, and stearic acids. In embodiments, the non-ionic surfactant comprises a polyethylene glycol long chain saturated fatty acid ester and further comprises an ethylene glycol long chain saturated fatty acid ester, such as PEG- and ethylene glycol-fatty acid esters of saturated C16-C18 fatty acids, e.g., palmitic and stearic acids. Such non-ionic surfactant can comprise PEG-6 stearate, ethylene glycol palmitostearate, and PEG-32 stearate, such as but not limited to TEFOSE 63.


According to embodiments, the non-ionic surfactant used as a thickening agent is not hydrophilic and has good emulsion properties. An illustrative example of such surfactant is TEFOSE 63, which has a hydrophilic-lipophilic balance (HLB) value of about 9-10.


According to embodiments, the pharmaceutical composition further comprises one or more mucoadherent agents to improve vaginal absorption of the estradiol. For example, a mucoadherent agent can be present to aid the pharmaceutical composition with adherence to the mucosa upon activation with water. According to embodiments, polycarbophil is the mucoadherent agent. According to embodiments, other mucoadherent agents include, for example and without limitation: poly (ethylene oxide) polymers having a molecular weight of from about 100,000 to about 900,000; chitosans carbopols including polymers of acrylic acid crosslinked with allyl sucrose or allyl pentaerythritol; polymers of acrylic acid and C10-C30 alkyl acrylate crosslinked with allyl pentaerythritol; carbomer homopolymer or copolymer that contains a block copolymer of polyethylene glycol and a long chain alkyl acid ester; and the like. According to embodiments, various hydrophilic polymers and hydrogels may be used as the mucoadherent agent. According to certain embodiments, the polymers or hydrogels can swell in response to contact with vaginal tissue or secretions, enhancing moisturizing and mucoadherent effects. The selection and amount of hydrophilic polymer may be based on the selection and amount of solubilizing agent. In some embodiments, the pharmaceutical composition includes a hydrophilic polymer but optionally excludes a gelling agent. In embodiments having a hydrogel, from about 5% to about 10% of the total mass may comprise the hydrophilic polymer. In further embodiments, hydrogels may be employed. A hydrogel may comprise chitosan, which swell in response to contact with water. In various embodiments, a cream pharmaceutical composition may comprise PEG-90M. In some embodiments, a mucoadherent agent is present in the pharmaceutical formulation, in the soft gel capsule, or both.


According to embodiments, the pharmaceutical compositions include one or more thermoreversible gels, typically of the hydrophilic nature including for example and without limitation, hydrophilic sucrose and other saccharide-based monomers (U.S. Pat. No. 6,018,033, which is incorporated by reference).


According to embodiments, the pharmaceutical composition further comprises a lubricant. In some embodiments, a lubricant can be present to aid in formulation of a dosage form. For example, a lubricant may be added to ensure that capsules or tablets do not stick to one another during processing or upon storage. Any suitable lubricant may be used. For example, lecithin, which is a mixture of phospholipids, is the lubricant.


According to embodiments, the pharmaceutical composition further comprises an antioxidant. Any suitable anti-oxidant may be used. For example, butylated hydroxytoluene, butylated hydroxyanisole, and Vitamin E TPGS.


According to embodiments, the pharmaceutical composition comprises about 20% to about 80% solubilizing agent by weight, about 0.1% to about 5% lubricant by weight, and about 0.01% to about 0.1% antioxidant by weight.


The choice of excipient will depend on factors such as, for example, the effect of the excipient on solubility and stability. Additional excipients used in various embodiments may include colorants and preservatives. Examples of colorants include FD&C colors (e.g., blue No. 1 and Red No. 40), D&C colors (e.g., Yellow No. 10), and opacifiers (e.g., Titanium dioxide). According to embodiments, colorants, comprise about 0.1% to about 2% of the pharmaceutical composition by weight. According to embodiments, preservatives in the pharmaceutical composition comprise methyl and propyl paraben, in a ratio of about 10:1, and at a proportion of about 0.005% and 0.05% by weight.


Generally, the solubilizing agents, excipients, other additives used in the pharmaceutical compositions described herein, are non-toxic, pharmaceutically acceptable, compatible with each other, and maintain stability of the pharmaceutical composition and the various components with respect to each other. Additionally, the combination of various components that comprise the pharmaceutical compositions will maintain will result in the desired therapeutic effect when administered to a subject.


Solubility of Estradiol


According to embodiments, solubilizing agents comprising mixtures of medium chain fatty acid glycerides, e.g., C6-C12, C8-C12, or C8-C10 fatty acid mono- and diglycerides or mono-, di-, and triglycerides dissolve estradiol. As illustrated in the Examples, good results were obtained with solubilizing agents that are predominantly a mixture of C8-C10 saturated fatty acid mono- and diglycerides, or medium chain triglycerides (e.g., Miglyol 810 or 812). Longer chain glycerides appear to be not as well suited for dissolution of estradiol.


A solubilizing agent comprising propylene glycol monocaprylate (e.g., CAPRYOL) and 2-(2-Ethoxyethoxy)ethanol (e.g., TRANSCUTOL) solubilized estradiol well.


Manufacture of the Pharmaceutical Composition

According to embodiments, the pharmaceutical composition is prepared via blending estradiol with a pharmaceutically acceptable solubilizing agent, including for example and without limitation, at least one medium chain fatty acid such as medium chain fatty acids consisting of at least one mono-, di-, or triglyceride, or derivatives thereof, or combinations thereof. According to embodiments, the pharmaceutical composition also comprises at least one glycol or derivatives thereof or combinations thereof or combinations of at least one glyceride and glycol. The glycol(s) may be used as solubilizing agents or to adjust viscosity and, thus, may be considered thickening agents, as discussed further herein. Optionally added are other excipients including, for example and without limitation, anti-oxidants, lubricants, and the like. According to embodiments, the pharmaceutical composition comprises sufficient solubilizing agent to fully solubilize the estradiol. It is expressly understood, however, the other volumes of solubilizing agent can be used depending on the level of estradiol solubilization desired. Persons of ordinary skill in the art will know and understand how to determine the volume of solubilizing agent and other excipients depending on the desired percent of estradiol to be solubilized in the pharmaceutical composition.


In illustrative embodiments, GELUCIRE 44/14 (lauroyl macrogol-32 glycerides EP, lauroyl polyoxyl-32 glycerides NF, lauroyl polyoxylglycerides (USA FDA IIG)) is heated to about 65° C. and CAPMUL MCM is heated to about 40° C. to facilitate mixing of the oil and non-ionic surfactant, although such heating is not necessary to dissolve the estradiol.


Specific Examples disclosed herein provide additional principles and embodiments illustrating the manufactures of the pharmaceutical compositions disclosed herein.


Delivery Vehicle

Generally, the pharmaceutical compositions described herein delivered intravaginally inside of a delivery vehicle, for example a capsule. According to embodiments, the capsules are soft capsules made of materials well known in the pharmaceutical arts, for example, gelatin. However, according to embodiments, the delivery vehicle is integral with the pharmaceutical composition (i.e., the pharmaceutical composition is the delivery vehicle). In such embodiments the pharmaceutical compositions is a gel, cream, ointment, tablet, or other preparation that is directly applied and absorbed vaginally.


According to embodiments, pharmaceutical compositions disclosed herein are contained in capsules, such as soft gelatin capsules. According to embodiments, the capsules contain one or more of the following: hydrophilic gel-forming bioadhesive (e.g., mucoadhesive) agents; a lipophilic agent; a gelling agent for the lipophilic agent, or a hydrodispersible agent. According to embodiments, the hydrophilic gel-forming bioadhesive agent is carboxyvinylic acid; hydroxypropylcellulose; carboxymethylcellulose; gelatin; xanthane gum; guar gum; aluminum silicate; or mixtures thereof. According to embodiments, the lipophilic agent is a liquid triglyceride; solid triglyceride (e.g., with a melting point of about 35° C.); carnauba wax; cocoa butter; or mixtures thereof. According to embodiments, the gelling agent is a hydrophobic colloidal silica. According to embodiments, the hydrodispersible agent is: polyoxyethylene glycol; polyoxyethylene glycol 7-glyceryl-cocoate; or mixtures thereof.


According to embodiments, the delivery vehicle is designed for ease of insertion. According to embodiments, the delivery vehicle is sized whereby it can be comfortably inserted into the vagina. According to embodiments, the delivery vehicle is prepared in a variety of geometries. For example, the delivery vehicle is shaped as a tear drop, a cone with frustoconical end, a cylinder, a cylinder with larger “cap” portion, or other shapes suitable for and that ease insertion into the vagina. According to embodiments, delivery vehicle is used in connection with an applicator. According to other embodiments, delivery vehicle is inserted digitally.


With reference to FIG. 2, delivery vehicle 200 comprises pharmaceutical composition 202 and capsule 204. Width 208 represents the thickness of capsule 204, for example about 0.108 inches. The distance from one end of delivery vehicle 200 to another is represented by distance 206, for example about 0.690 inches. The size of delivery vehicle 200 may also be described by the arc swept by a radius of a given length. For example, arc 210, which is defined by the exterior of gelatin 204, is an arc swept by a radius of about 0.189 inches. Arc 212, which is defined by the interior of capsule 204, is an arc swept by a radius of about 0.0938 inches. Arc 214, which is defined by the exterior of gelatin 204 opposite arc 210, is an arc swept by a radius of about 0.108 inches. Suitable capsules of other dimensions may be provided. According to embodiments, capsule 204 has dimensions the same as or similar to the ratios as provided above relative to each other.


According to embodiments, the delivery vehicle is designed to remaining in the vagina until the pharmaceutical compositions are released. According to embodiments, delivery vehicle dissolves intravaginally and is absorbed into the vaginal tissue with the pharmaceutical composition, which minimizes vaginal discharge. In such embodiments, delivery mechanism is made from constituents that are non-toxic, for example, gelatin.


Design Factors for Vaginally Inserted Pharmaceutical Compositions

According to embodiments, the pharmaceutical composition is designed to maximize favorable characteristics that lead to patient compliance (patients that discontinue treatment prior to completion of the prescribed course of therapy), without sacrificing efficacy. Favorable characteristics include, for example, lack of or reduction of irritation relative to other hormone replacement pessaries, lack of or reduction in vaginal discharge of the pharmaceutical composition and delivery vehicle relative to other hormone replacement pessaries, lack of or reduction of pharmaceutical composition or delivery vehicle residue inside the vagina, ease of administration compared to other hormone replacement pessaries, or improved efficacy of drug product relative to otherwise similar pharmaceutical compositions.


According to embodiments, the pharmaceutical composition is non-irritating or minimizes irritation. Patient irritation comprises pain, pruritis (itching), soreness, excessive discharge, swelling, or other similar conditions. Patient irritation results in poor compliance. Non-irritating or reduced irritation pharmaceutical compositions are measured relative to competing hormone pessaries, including tablets, creams, or other intravaginal estrogen delivery forms.


According to embodiments, the pharmaceutical compositions does not result in systemic exposure (e.g., blood circulation of estradiol), which improves safety. According to other embodiments, the pharmaceutical compositions disclosed herein result in significantly reduced systemic exposure (e.g., blood circulation of estradiol) when compared to RLDs.


According to embodiments, the pharmaceutical composition does not leave residue inside the vagina. Rather, the pharmaceutical composition and delivery vehicle are substantially absorbed or dispersed without resulting in unabsorbed residue or unpleasant sensations of non-absorbed or non-dispersed drug product. Measurement of lack of residue is relative to other vaginally inserted products or can be measured objectively with inspection of the vaginal tissues. For example, certain other vaginally inserted products contain starch which can result in greater discharge from the vagina following administration than. In some embodiments, the pharmaceutical compositions provided herein provide a lower amount, duration, or frequency of discharge following administration compared to other vaginally inserted products (e.g., compressed tablets).


According to embodiments, the pharmaceutical composition improves vaginal discharge compared to other pessaries, including pessaries that deliver hormones. Ideally, vaginal discharge is eliminated, minimized, or improved compared to competing products.


According to embodiments, the pharmaceutical compositions disclosed herein are inserted digitally. According to embodiments, the pharmaceutical compositions are digitally inserted approximately two inches into the vagina without a need for an applicator. According to embodiments, the pharmaceutical compositions are designed to be also inserted with an applicator, if desired. According to some embodiments, because the site of VVA is in the proximal region of the vagina (towards the vaginal opening), the pharmaceutical compositions disclosed herein are designed to be inserted in the proximal portion of the vagina.


Through extensive experimentation, various medium chain fatty acid esters of glycerol and propylene glycol demonstrated one or more favorable characteristics for development as a human drug product. According to embodiments, the solubilizing agent was selected from at least one of a solvent or co-solvent. Suitable solvents and co-solvents include any mono-, di- or triglyceride and glycols, and combinations thereof.


According to embodiments, the pharmaceutical composition is delivered via a gelatin capsule delivery vehicle. According to these embodiments, the pharmaceutical composition is a liquid pharmaceutical composition. According to embodiments, the delivery vehicle is a soft capsule, for example a soft gelatin capsule. Thus, the pharmaceutical composition of such embodiments is encapsulated in the soft gelatin capsule or other soft capsule.


According to embodiments, the pharmaceutical composition comprises estradiol that is at least about 80% solubilized in a solubilizing agent comprising one or more C6 to C14 medium chain fatty acid mono-, di-, or triglycerides and, optionally, a thickening agent. According to embodiments, the pharmaceutical composition comprises estradiol that is at least about 80% solubilized one or more C6 to C12 medium chain fatty acid mono-, di-, or triglycerides, e.g., one or more C6 to C14 triglycerides, e.g., one or more C6 to C12 triglycerides, such as one or more C8-C10 triglycerides. These embodiments specifically contemplate the estradiol being at least 80% solubilized. These embodiments specifically contemplate the estradiol being at least 90% solubilized. These embodiments specifically contemplate the estradiol being at least 95% solubilized. These embodiments specifically contemplate the estradiol being fully solubilized.


As noted above, liquid pharmaceutical compositions are liquid at room temperature or at body temperature. For example, in some embodiments, a pharmaceutical composition provided herein is a liquid formulation contained within a soft gel capsule. Gels, hard fats, or other solid forms that are not liquid at room or body temperature are less desirable in embodiments of the pharmaceutical composition that are liquid.


The thickening agent serves to increase viscosity, e.g., up to about 10,000 cP (10,000 mPa-s), typically to no more than about 5000 cP, and more typically to between about 50 and 1000 cP. In embodiments, the non-ionic surfactant, e.g., GELUCIRE or TEFOSE, may be solid at room temperature and require melting to effectively mix with the solubilizing agent. However, in these embodiments, the resultant pharmaceutical composition remains liquid, albeit with greater viscosity, not solid.


According to embodiments, the pharmaceutical composition comprises estradiol, the medium chain solubilizing agent, and the thickening agent as the ingredients delivered via a soft capsule delivery vehicle. Other ingredients, e.g., colorants, antioxidants, preservatives, or other ingredients may be included as well. However, the addition of other ingredients should be in amounts that do not materially change the solubility of the estradiol, the pharmacokinetics of the pharmaceutical composition, or efficacy of the pharmaceutical composition. Other factors that should be considered when adjusting the ingredients of the pharmaceutical composition include the irritation, vaginal discharge, intravaginal residue, and other relevant factors, for example those that would lead to reduced patient compliance. Other contemplated ingredients include: oils or fatty acid esters, lecithin, mucoadherent agents, gelling agents, dispersing agents, or the like.


Methods

According to embodiments, the pharmaceutical compositions disclosed herein can be used for the treatment of WA, including the treatment of at least one WA symptom including: vaginal dryness, vaginal or vulvar irritation or itching, dysuria, dysparuenia, and vaginal bleeding associated with sexual activity, among others. According to embodiments the methods of treatment are generally applicable to females.


According to embodiments, the pharmaceutical compositions disclosed herein can be used for the treatment of estrogen-deficient urinary states. According to embodiments, the pharmaceutical compositions disclosed herein can be used for the treatment of dysparuenia, or vaginal bleeding associated with sexual activity.


According to embodiments, treatment of the VVA, estrogen-deficient urinary states, and dysparuenia and vaginal bleeding associated with sexual activity occurs by administering the pharmaceutical compositions intravaginally. According to embodiments where the delivery vehicle is a capsule, the patient obtains the capsule and inserts the capsule into vagina, where the capsule dissolves and the pharmaceutical composition is releases into the vagina where it is absorbed into the vaginal tissue. In some embodiments, the pharmaceutical composition is completely absorbed into the vaginal tissue. In some embodiments, the pharmaceutical composition is substantially absorbed into the vaginal tissue (e.g., at least about 80% by weight, at least about 85% by weight, at least about 90% by weight, at least about 95% by weight, at least about 97% by weight, at least about 98% by weight, or at least about 99% by weight of the composition is absorbed). According to embodiments, the capsule is inserted about two inches into the vagina digitally, however the depth of insertion is generally any depth that allows for adsorption of substantially all of the pharmaceutical composition. According to embodiments, the capsule can also be applied using an applicator that deposits the capsule at an appropriate vaginal depth as disclosed herein.


According to embodiments where the pharmaceutical composition is a cream, gel, ointment, or other similar preparation, the pharmaceutical composition is applied digitally, as is well known and understood in the art.


Upon release of the pharmaceutical composition in the vagina, estradiol is locally absorbed. For example, following administration of the pessary to the proximal region of the vagina of a patient provides a therapeutically effective concentration of estradiol over 24 hours in the proximal region of the vagina.


According to embodiments, the timing of administration of the pharmaceutical composition of this disclosure may be conducted by any safe means as prescribed by an attending physician. According to embodiments, a patient will administer the pharmaceutical composition (e.g., a capsule) intravaginally each day for 14 days, then twice weekly thereafter.


According to embodiments, the pharmaceutical compositions are vaginally administered with co-administration of an orally administered estrogen-based (or progestin-based or progestin- and estrogen-based) pharmaceutical drug product, or patch, cream, gel, spray, transdermal delivery system or other parenterally-administered estrogen-based pharmaceutical drug product, each of which can include natural, bio-similar, or synthetic or other derived estrogens or progestins. According to embodiments, modulation of circulating estrogen levels provided via the administration of the pharmaceutical compositions disclosed herein, if any, are not intended to be additive to any co-administered estrogen product and its associated circulating blood levels. According to other embodiments, co-administrated estrogen products are intended to have an additive effect as would be determined by the patient physician.


According to embodiments, the efficacy and safety of the pharmaceutical compositions described herein in the treatment of the symptoms of VVA may be determined. According to embodiments, the size, effect, cytology, histology, and variability of the VVA may be determined using various endpoints to determine efficacy and safety of the pharmaceutical compositions described herein or as otherwise accepted in the art, at present or as further developed. On source of endpoints is with the US Food and Drug Administration's (FDA) published guidelines for treatment of VVA with estradiol.


Measurement of Efficacy

According to embodiments, administration of the pharmaceutical compositions described herein resulted in treatment of the WA, as well as improvement of one or more of the associated symptoms. Patients with WA experience shrinking of the vaginal canal in both length and diameter and the vaginal canal has fewer glycogen-rich vaginal cells to maintain moisture and suppleness. In addition, the vaginal wall can become thin, pale, dry, or sometimes inflamed (atrophic vaginitis). These changes can manifest as a variety of symptoms collectively referred to as VVA. Such symptoms include, without limitations, an increase in vaginal pH; reduction of vaginal epithelial integrity, vaginal secretions, or epithelial surface thickness; pruritis; vaginal dryness; dyspareunia (pain or bleeding during sexual intercourse); urinary tract infections; or a change in vaginal color. According to embodiments, efficacy is measured as a reduction of vulvar and vaginal atrophy in a patient back to premenopausal conditions. According to embodiments, the change is measured as a reduction in the severity of one or more atrophic effects measured at baseline (screening, Day 1) and compared to a measurement taken at Day 15 (end of treatment). Severity of the atrophic effect may be measured using a scale of 0 to 3 where, for example, none=0, mild=1, moderate=2, or severe=3. Such scoring is implemented to evaluate the pre-treatment condition of patients; to determine the appropriate course of a treatment regime; such as dosage, dosing frequency, and duration, among others; and post-treatment outcomes.


One of the symptoms of VVA is increased vaginal pH. In further aspects of this disclosure, treatment with the pharmaceutical compositions described herein resulted in a decrease in vaginal pH. A decrease in vaginal pH is measured as a decrease from the vaginal pH at baseline (screening) to the vaginal pH at Day 15, according to embodiments. In some embodiments, a pH of 5 or greater may be associated with WA. In some embodiments, pH is measured using a pH indicator strip placed against the vaginal wall. In some embodiments, a change in vaginal pH is a change in a patient's vaginal pH to a pH of less than about pH 5.0. In some embodiments, a subject's vaginal pH may be less than about pH 4.9, pH 4.8, pH 4.7, pH 4.6, pH 4.5, pH 4.4, pH 4.3, pH 4.2, pH 4.1, pH 4.0, pH 3.9, pH 3.8, pH 3.7, pH 3.6, or pH 3.5.


According to embodiments, treatment with the pharmaceutical compositions described herein resulted in improvements in the vaginal Maturation Index. The Maturation Index is measured as a change in cell composition. According to embodiments and as related to VVA, a change in cell composition is measured as the change in percent of composition or amount of parabasal vaginal cells, intermediate cells, and superficial vaginal cells, such as a change in the composition or amount of parabasal vaginal cells compared with or, relative to, a change in superficial vaginal cells. A subject having VVA symptoms often has an increased number of parabasal cells and a reduced number of superficial cells (e.g., less than about 5%) compared with women who do not suffer from VVA. Conversely, a subject having decreasing VVA symptoms, or as otherwise responding to treatment, may demonstrate an improvement in the Maturation Index, specifically a decrease in the amount of parabasal cells or an increase in the amount of superficial cells compared to baseline (screening). In embodiments, a decrease in parabasal cells is measured as a reduction in the percent of parabasal cells; the percent reduction may be at least about an 85%, 80%, 75%, 70%, 65%, 60%, 55%, 50%, 45%, 40%, 35%, 30%, 25%, 20%, 15% or 10% reduction in the number of parabasal cells. In embodiments, a percent reduction may be at least about a 54% reduction in the number of parabasal cells. In embodiments, an increase in superficial cells is measured as an increase in the percent of superficial cells; the percent increase in superficial cells may be at least about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% increase in the number of superficial cells. In further embodiments, a percent increase may be at least about a 35% increase in the number of superficial cells.


In some embodiments, an improvement in the Maturation Index is assessed as a change over time. For example, as a change in cell composition measured at a baseline (screening) at Day 1 compared to the cell composition measured at Day 15. The change in cell composition may also be assessed as a change in the amount of parabasal cells over time, optionally in addition to measuring changes in parabasal cells and superficial cells as described above. Such cells may be obtained from the vaginal mucosal epithelium through routine gynecological examination and examined by means of a vaginal smear.


In various further aspects of this disclosure, treatment with the pharmaceutical compositions described herein resulted in any of: an increase in superficial cells; a decrease in parabasal cells; and an increase in intermediate cells.


In further aspects of this disclosure, samples may be collected to determine hormone levels, in particular, estradiol levels. In some embodiments, blood samples may be taken from a subject and the level of estradiol measured (pg/ml). In some embodiments, estradiol levels may be measured at 0 hours (for example, at time of first treatment), at 1 hour (for example, post first treatment), at 3 hours, and at 6 hours. In some embodiments, samples may be taken at day 8 (for example, post first treatment) and at day 15 (for example, one day post the last treatment on day 14). In some embodiments, descriptive statistics of plasma estradiol concentrations at each sampling time and observed Cmax and Tmax values may be measured and the AUC calculated.


In some embodiments, a pessary can comprise about 25 μg of estradiol. In such cases, administration of the pessary to a patient can provide, in a plasma sample from the patient, parameters including one or more parameters selected from: 1) a corrected geometric mean peak plasma concentration (Cmax) of estradiol of about 19 pg*hr/ml to about 29 pg*hr/ml (e.g., 19.55 pg*hr/ml to about 28.75 pg*hr/ml); or 2) a corrected geometric mean area under the curve (AUC)0-24 of estradiol of about 75 pg*hr/ml to about 112 pg*hr/ml (e.g., 75.82 pg*hr/ml to about 111.50). In some embodiments, administration of the pessary to a patient provides, in a plasma sample from the patient, one or more parameters selected from: 1) a corrected geometric mean peak plasma concentration (Cmax) of estrone of about 9 pg*hr/ml to about 14 pg*hr/ml (e.g., 9.17 pg*hr/ml to about 13.49 pg*hr/ml); and 2) a corrected geometric mean area under the curve (AUC)0-24 of estrone of about 43 pg*hr/ml to about 65 pg*hr/ml (e.g., 43.56 pg*hr/ml to about 64.06 pg*hr/ml). In some embodiments, administration of the pessary to a patient provides, in a plasma sample from the patient, provides one or more parameters selected from: 1) a corrected geometric mean peak plasma concentration (Cmax) of estrone sulfate of about 416 pg*hr/ml to about 613 pg*hr/ml (e.g., 416.53 pg*hr/ml to about 612.55 pg*hr/ml); and 2) a corrected geometric mean area under the curve (AUC)0-24 of estrone sulfate of about 3598 pg*hr/ml to about 5291 pg*hr/ml (e.g., 3598.04 pg*hr/ml to about 5291.24 pg*hr/ml).


In some embodiments, a pessary can comprise about 10 μg of estradiol. In such cases, administration of the pessary to a patient can provide, in a plasma sample from the patient, one or more parameters selected from: 1) a corrected geometric mean peak plasma concentration (Cmax) of estradiol of about 12 pg*hr/ml to about 18 pg*hr/ml (e.g., 12.22 pg*hr/ml to about 17.98 pg*hr/ml); 2) a corrected geometric mean area under the curve (AUC)0-24 of estradiol of about 42 pg*hr/ml to about 63 pg*hr/ml (e.g., 42.18 pg*hr/ml to about 62.02 pg*hr/ml); and 3) a corrected geometric mean time to peak plasma concentration (Tmax) of estradiol of about 1 hrs to about 3 hrs (e.g., 1.49 hrs to about 2.19 hrs). In some embodiments, administration of the pessary to a patient provides, in a plasma sample from the patient, one or more parameters selected from: 1) a corrected geometric mean peak plasma concentration (Cmax) of estrone of about 4 pg*hr/ml to about 7 pg*hr/ml (e.g., 4.38 pg*hr/ml to about 6.44 pg*hr/ml); 2) a corrected geometric mean area under the curve (AUC)0-24 of estrone of about 20 pg*hr/ml to about 31 pg*hr/ml (e.g., 20.60 pg*hr/ml to about 30.30 pg*hr/ml); and 3) a corrected geometric mean time to peak plasma concentration (Tmax) of estrone of about 4 hrs to about 8 hrs (e.g., 4.99 hrs to about 7.34 hrs). In some embodiments, administration of the pessary to a patient provides, in a plasma sample from the patient, one or more parameters selected from: 1) a corrected geometric mean peak plasma concentration (Cmax) of estrone sulfate of about 10 pg*hr/ml to about 16 pg*hr/ml (e.g., 10.34 pg*hr/ml to about 15.20 pg*hr/ml); 2) a corrected geometric mean area under the curve (AUC)0-24 of estrone sulfate of about 56 pg*hr/ml to about 84 pg*hr/ml (e.g., 56.61 pg*hr/ml to about 83.25 pg*hr/ml); and 3) a corrected geometric mean time to peak plasma concentration (Tmax) of estrone sulfate of about 4 hrs to about 7 hrs (e.g., 4.67 hrs to about 6.86 hrs).


In some embodiments, a pessary can comprise about 4 μg of estradiol. In such cases, administration of the pessary to a patient can provide, in a plasma sample from the patient, one or more parameters selected from: 1) a corrected geometric mean peak plasma concentration (Cmax) of estradiol of about 4 pg*hr/ml to about 8 pg*hr/ml; 2) a corrected geometric mean area under the curve (AUC)0-24 of estradiol of about 16 pg*hr/ml to about 26 pg*hr/ml; and 3) a corrected geometric mean time to peak plasma concentration (Tmax) of estradiol of about 0.25 hrs to about 2 hrs. In some embodiments, administration of the pessary to a patient provides, in a plasma sample from the patient, one or more parameters selected from: 1) a corrected geometric mean peak plasma concentration (Cmax) of estrone of about 1 pg*hr/ml to about 3 pg*hr/ml; 2) a corrected geometric mean area under the curve (AUC)0-24 of estrone of about 8 pg*hr/ml to about 13 pg*hr/ml; and 3) a corrected geometric mean time to peak plasma concentration (Tmax) of estrone of about 1 hrs to about 4 hrs. In some embodiments, administration of the pessary to a patient provides, in a plasma sample from the patient, one or more parameters selected from: 1) a corrected geometric mean peak plasma concentration (Cmax) of estrone sulfate of about 4 pg*hr/ml to about 7 pg*hr/ml; 2) a corrected geometric mean area under the curve (AUC)0-24 of estrone sulfate of about 22 pg*hr/ml to about 34 pg*hr/ml; and 3) a corrected geometric mean time to peak plasma concentration (Tmax) of estrone sulfate of about 1 hrs to about 3 hrs.


A pharmaceutical composition provided herein can result in substantially local delivery of estradiol. For example, plasma concentrations of estradiol, estrone, and estrone sulfate measured in the plasma of a patient following administration of a pharmaceutical composition as provided herein be statistically similar to those measured following administration of a placebo formulation (i.e. a similar formulation lacking the estradiol). Accordingly, in some embodiments, the plasma concentrations of estradiol, estrone, or estrone sulfate measured following administration of a pharmaceutical composition provided herein may be low compared to RLD formulations.


In some embodiments, a pessary can include about 1 μg to about 25 μg of estradiol. Upon administration the pessary to a patient, a plasma sample from the patient can provide a corrected geometric mean peak plasma concentration (Cmax) of estradiol that is less than about 30 pg*hr/ml. For example, administration of the pessary to a patient provides a corrected geometric mean peak plasma concentration (Cmax) of estradiol that is less than about 18 pg*hr/ml. In some embodiments, administration of the pessary to a patient provides a corrected geometric mean area under the curve (AUC)0-24 of estradiol that is less than about 112 pg*hr/ml. For example, administration of the pessary to a patient provides a corrected geometric mean area under the curve (AUC)0-24 of estradiol that is less than about 63 pg*hr/ml.


In some embodiments, administration of the pessary to a patient provides a corrected geometric mean peak plasma concentration (Cmax) of estrone that is less than about 14 pg*hr/ml. For example, administration of the pessary to a patient provides a corrected geometric mean peak plasma concentration (Cmax) of estrone that is less than about 7 pg*hr/ml. In some embodiments, administration of the pessary to a patient provides a corrected geometric mean area under the curve (AUC)0-24 of estrone that is less than about 65 pg*hr/ml. For example, administration of the pessary to a patient provides a corrected geometric mean area under the curve (AUC)0-24 of estrone that is less than about 31 pg*hr/ml.


In some embodiments, administration of the pessary to a patient provides a corrected geometric mean peak plasma concentration (Cmax) of estrone sulfate that is less than about 613 pg*hr/ml. For example, administration of the pessary to a patient provides a corrected geometric mean peak plasma concentration (Cmax) of estrone sulfate that is less than about 16 pg*hr/ml. In some embodiments, administration of the pessary to a patient provides a corrected geometric mean area under the curve (AUC)0-24 of estrone sulfate that is less than about 5291 pg*hr/ml. For example, administration of the pessary to a patient provides a corrected geometric mean area under the curve (AUC)0-24 of estrone sulfate that is less than about 84 pg*hr/ml.


In further aspects of this disclosure, capsule disintegration may be determined. In some embodiments, delivery vehicle disintegration or absorption (presence or absence of the delivery vehicle after administration) at day 1 of treatment (for example, at 6 hours post first treatment) and at day 15 (for example, one day post the last treatment on day 14).


Statistical Measurements


According to embodiments, pharmacokinetics of the pharmaceutical composition disclosed herein are measured using statistical analysis. According to embodiments, Analysis of Variance (“ANOVA”) or Analysis of CoVariance (“ANCOVA”) are used to evaluate differences between a patient receiving treatment with a pharmaceutical composition comprising an active pharmaceutical composition (for example, a pharmaceutical composition comprising estradiol) and a patient receiving treatment with a placebo (for example, the same pharmaceutical composition but without estradiol) or a reference drug. A person of ordinary skill in the art will understand how to perform statistical analysis of the data collected.


EXAMPLES

The following examples are of pharmaceutical compositions, delivery vehicles, and combinations thereof. Methods of making are also disclosed. Data generated using the pharmaceutical compositions disclosed herein are also disclosed.


Example 1: Pharmaceutical Composition

In embodiments, estradiol is procured and combined with one or more pharmaceutically acceptable solubilizing agents. The estradiol is purchased as a pharmaceutical grade ingredient, often as micronized estradiol, although other forms can also be used. In embodiments, the pharmaceutical composition comprises estradiol in a dosage strength of from about 1 μg to about 50 μg. In embodiments, the pharmaceutical composition comprises 10 μg of estradiol. In embodiments, the pharmaceutical composition comprises 25 μg of estradiol.


In embodiments, the estradiol is combined with pharmaceutically acceptable solubilizing agents, and, optionally, other excipients, to form a pharmaceutical composition. In embodiments, the solubilizing agent is one or more of CAPMUL MCM, MIGLYOL 812, GELUCIRE 39/01, GELUCIRE 43/01, GELUCIRE 50/13, and TEFOSE 63.


GELUCIRE 39/01 and GELUCIRE 43/01 each have an HLB value of 1. GELUCIRE 50/13 has an HLB value of 13. TEFOSE 63 has an HLB value of between 9 and 10.


Various combinations of pharmaceutically acceptable solubilizing agents were combined with estradiol and examined as shown in Table 1.









TABLE 1







Capmul MCM (“MCM”), Gelucire 39/01 (“39/01”), Gelucire


43/01(“43/01”), Gelucire 50/13(“50/13”), and Tefose (“Tefose 63”)

















Physical
Physical








state @
state @ 37° C.

Melting
Dispersion



Vehicle

Room
after ~30
Viscosity
Time @
in water


#
system
Ratio
Temperature
minutes
(cps)
37° C.
37° C.

















1
MCM:39/01
8:2
Solid
Clear liquid
50 @
Start: 6 min
Small oil







37° C.
Finish: 12 min
drops on









top


2
MCM:39/01
7:3
Solid
Clear liquid

Start: 9 min








Finish: 19 min


3
MCM:39/01
6:4
Solid
Clear liquid

Start: 20 min








Finish: 32 min


4
MCM:43/01
8:2
Solid
Liquid with






solid particles


5
MCM:43/01
7:3
Solid
Liquid with






solid particles


6
MCM:50/13
9:1
Liquid/
Liquid/cloudy
140@
Clear after
Uniformly





cloudy

25° C.
20 min
cloudy









dispersion


7
MCM:50/13
8:2
Liquid/
Liquid/cloudy
190@

Uniformly





cloudy

25° C.

cloudy









dispersion


8
MCM:50/13
7:3
Semisolid
Semisolid


9
MCM:TEFOSE
9:1
Semisolid
Liquid/cloudy
150@
Start: 1 min
Uniformly



63



25° C.
Finish: 5 min
cloudy









dispersion


10
MCM:TEFOSE
8:2
Semisolid
Semisolid
240@

Uniformly



63



25° C.

cloudy









dispersion


11
MCM:TEFOSE
7:3
Semisolid
Semisolid
380@
Semisolid
Uniformly



63



25° C.
after 30 min
cloudy








at 37° C.,
dispersion








doesn't melt








at 41° C.


12
MIGLYOL
9:1
Semisolid
Semisolid
140@

2 phases,



812:50/13



25° C.

oil on top


13
MIGLYOL
9:1
Liquid/
Liquid/cloudy
90@
Start: 1 min
2 phases,



812:TEFOSE

cloudy

25° C.
Finish: 5 min
oil on top



63









Pharmaceutical compositions in Table 1 that were liquid or semisolid at room temperature were tested using a Brookfield viscometer (Brookfield Engineering Laboratories, Middleboro, Mass.) at room temperature. Pharmaceutical compositions appearing in Table 1 that were solid at ambient temperature were tested using a Brookfield viscometer at 37° C.


Pharmaceutical compositions appearing in Table 1 that were solid at room temperature were assessed at 37° C. to determine their melting characteristics. The viscosity of the gels can be important during encapsulation of the formulation. For example, in some cases, it is necessary to warm the formulation prior to filing of the gelatin capsules. In addition, the melting characteristics of the composition can have important implications following administration of the formulation into the body. For example, in some embodiments, the formulation will melt at temperatures below about 37° C. Pharmaceutical Composition 11 (Capmul MCM/Tefose 63), for example, did not melt at 37° C. or 41° C.


A dispersion assessment of the pharmaceutical compositions appearing in Table 1 was performed. The dispersion assessment was performed by transferring 300 mg of each vehicle system in 100 ml of 37° C. water, without agitation, and observing for mixing characteristics. Results varied from formation of oil drops on the top to separation of phases to uniform, but cloudy dispersions. Generally speaking, it is believed that formulations able to readily disperse in aqueous solution will have better dispersion characteristics upon administration. It was surprisingly found, however, as shown below in Examples 7-9, that formulations that did not readily disperse in aqueous solution (e.g., Formulation 13) and instead formed two phases upon introduction to the aqueous solution were found to be the most effective when administered to the human body.


Example 2: Delivery Vehicle

In embodiments, the pharmaceutical composition is delivered in a gelatin capsule delivery vehicle. The gelatin capsule delivery vehicle comprises, for example, gelatin (e.g., Gelatin, NF (150 Bloom, Type B)), hydrolyzed collagen (e.g., GELITA®, GELITA AG, Eberbach, Germany), glycerin, sorbitol special, or other excipients in proportions that are well known and understood by persons of ordinary skill in the art. Sorbitol special may be obtained commercially and may tend to act as a plasticizer and humectant.


A variety of delivery vehicles were developed, as show in Table 2, Gels A through F. In Table 2, each delivery vehicle A through F differs in the proportion of one or more components.









TABLE 2







Gelatin Capsule Delivery Vehicles














A
B
C
D
E
F


Ingredient
% w/w
% w/w
% w/w
% w/w
% w/w
% w/w
















Gelatin, NF (150 Bloom, Type B)
41.0
41.0
41.0
41.0
43.0
43.0


Glycerin 99.7%, USP
6.0
6.0
6.0
6.0
18.0
18.0


Sorbitol Special, USP
15.0
15.0
15.0
15.0


GELITA ® (hydrolyzed collagen)
3



3.0


Citric acid

0.1
0.5
1

0.1


Purified Water
35.0
37.9
37.5
37.0
36.0
38.9


Total
100.0
100.0
100.0
100.0
100.0
100.0


Dissolution gel strips, Avg of 3
48 min
50 min
75 min
70 min


(500 ml DH2O, 50 rpm @ 37° C.)
(42, 45, 58)
(50, 51, 50)
(76, 75, 74)
(70, 71, 70)


Dissolution gel strips, Avg of 3
70 min



78 min
82 min


(500 ml pH 4 buffer, 50 rpm @ 37° C.)









Each delivery vehicle A through F was prepared at a temperature range from about 45° C. to about 85° C. Each molten delivery vehicle A through F was cast into a film, dried, and cut into strips. The strips were cut into uniform pieces weighing about 0.5 g, with about 0.5 mm thickness. Strips were placed into a USP Type 2 dissolution vessel in either water or pH 4 buffer solution and the time for them to completely dissolve was recorded (see TABLE 2). Delivery vehicle A had the fastest dissolution in both water and pH 4 buffer solution.


Example 3: Pharmaceutical Compositions and Delivery Vehicle

Various combinations of the pharmaceutical compositions from TABLE 1 and from TABLE 2 were prepared. The combinations are shown in TABLE 3.













TABLE 3








Batch Size
Delivery


Trial
Pharmaceutical Composition
Ratio
g
Vehicle







1
MCM:39/01
8:2
750
A


2
MCM:50/13
8:2
750
A


3
MCM:TEFOSE 63
8:2
750
A


4
MCM:TEFOSE 63
8:2
750
B


5
MIGLYOL 812:TEFOSE 63
9:1
750
A









Each aliquot of the pharmaceutical compositions of Table 3 about 300 mg to about 310 mg. Batch size was as listed in TABLE 3. To encapsulate the vehicle system, each 300 mg to about 310 mg pharmaceutical composition aliquot was encapsulated in about 200 mg of the gelatin capsule delivery vehicle. Thus, for example, in Trial 1, the pharmaceutical composition denoted by MCM:39/01 was encapsulated in gelatin capsule delivery vehicle A for a total encapsulated weight of about 500 mg to about 510 mg. The aliquot size is arbitrary depending on the concentration of the estradiol and the desired gelatin capsule delivery vehicle size. Artisans will readily understand how to adjust the amount of estradiol in the pharmaceutical composition to accommodate a given size of delivery vehicle, when the delivery vehicle encapsulates the pharmaceutical composition.


Example 4: Estradiol Solubility

In various experiments, solubilizing agents were tested to determine whether they were able to solubilize 2 mg of estradiol for a total pharmaceutical composition weight of 100 mg. The solubilizing agents were considered suitable if estradiol solubility in the solubilizing agent was greater than or equal to about 20 mg/g. Initial solubility was measured by dissolving micronized estradiol into various solubilizing agents until the estradiol was saturated (the estradiol/solubilizing agent equilibrated for three days), filtering the undissolved estradiol, and analyzing the resulting pharmaceutical composition for estradiol concentration by HPLC.









TABLE 4







Solubility of Solubilizing Agents (*denotes literature reference)










Ingredient
Solubility (mg/g)






PEG 400
105*



Propylene Glycol
 75*



Polysorbate 80
 36*



TRANSCUTOL HP
141 



CAPMUL PG8
  31.2









Example 5: Pharmaceutical Compositions

The following pharmaceutical compositions are contemplated.


Gel Mass














Ingredient
% w/w
Qty/Batch (kg)

















Gelatin 150 Bloom Limed Bone, NF
41.00
82.00


Hydrolyzed Gelatin
3.00
6.00


Glycerin 99.7%
6.00
12.00


Sorbitol Special, NF
15.00
30.00


Opatint White G-18006
1.20
2.40


Opatine Red DG-15001
0.06
0.12


Purified Water, USP
33.74
67.48


Total
100.00
200.00 Kg









Pharmaceutical Composition 1: 10 μg Estradiol
















Qty/Capsule




Ingredients
(mg)
% w/w
Qty/Batch



















Estradiol hemihydrate
0.010
0.003
0.10
g


micronized, USP






CAPMUL ® MCM, NF (Glyceryl
240.0
79.997
2.40
kg


Caprylate/Caprate or Medium






Chain Mono- and Diglycerides)






GELUCIRE ® 50/13 (stearoyl
60.0
20.0
600.0
g


polyoxyl-32 glycerides NF)






Total
300.0
100.0
3.0
kg









Pharmaceutical Composition 2: 10 μg Estradiol
















Qty/Capsule




Ingredients
(mg)
% w/w
Qty/Batch



















Estradiol hemihydrate
0.010
0.003
0.10
g


micronized, USP






MIGLOYL ® 812 (medium
270.0
89.997
2.70
kg


chain triglyceride)






TEFOSE ® 63 (mixture of
30.0
10.0
300.0
g


PEG-6 stearate or






ethylene glycol






palmitostearate or PEG-32






stearate; polyoxyl 6 and






polyoxyl 32






palmitostearate/glycol






stearate)






Total
300.0
100.0
3.00
kg









Pharmaceutical Composition 3: 25 μg Estradiol
















Qty/Capsule




Ingredients
(mg)
% w/w
Qty/Batch



















Estradiol hemihydrate
0.026*
0.009
0.26
g


micronized, USP






MIGLOYL ® 812 (medium
270.0
89.991
2.70
kg


chain triglyceride)






TEFOSE ® 63 (mixture of
30.02
10.0
300.0
g


PEG-6 stearate or






ethylene glycol






palmitostearate or PEG-32






stearate; polyoxyl 6 and






polyoxyl 32






palmitostearate/glycol






stearate)






Total
300.0
100.0
3.00
kg





*1.0 mg estradiol is equivalent to 1.03 mg estradiol hemihydrate






Pharmaceutical Composition 4: 4 μg Estradiol
















Qty/Capsule




Ingredients
(mg)
% w/w
Qty/Batch



















Estradiol hemihydrate
0.0041*
0.001
0.041
g


micronized, USP






MIGLOYL ® 812 (medium
269.99
89.999
2700.0
g


chain triglyceride)






TEFOSE ® 63 (mixture of
30.0
10.0
300.0
g


PEG-6 stearate or






ethylene glycol






palmitostearate or PEG-32






stearate; polyoxyl 6 and






polyoxyl 32






palmitostearate/glycol






stearate)






Total
300.0
100.0
3000.0
g





*1.0 mg estradiol is equivalent to 1.03 mg estradiol hemihydrate






Example 6: Process


FIG. 1 illustrates an embodiment of a method making pharmaceutical composition comprising estradiol solubilized in CapmulMCM/Gelucire solubilizing agent encapsulated in a soft gelatin delivery vehicle 100. In operation 102, the CapmulMCM is heated to 40° C.±5° C. Heating may be accomplished through any suitable means. The heating may be performed in any suitable vessel, such as a stainless steel vessel. Other pharmaceutical compositions can be made using the same general method by substituting various excipients, including the solubilizing agent.


In operation 104, GELUCIRE is mixed with the CapmulMCM to form the finished solubilizing agent. As used herein, any form of GELUCIRE may be used in operation 104. For example, one or more of GELUCIRE 39/01, GELUCIRE 43/01, GELUCIRE 50/13 may be used in operation 104. Mixing is performed as would be known to persons of ordinary skill in the art, for example by impeller, agitator, stirrer, or other like devices used to mix pharmaceutical compositions. Operation 104 may be performed under an inert or relatively inert gas atmosphere, such as nitrogen gas. Mixing may be performed in any vessels that are known to persons of ordinary skill in the art, such as a stainless steel vessel or a steel tank.


In operation 106 estradiol is mixed into the solubilizing agent. In embodiments, the estradiol in micronized when mixed into the solubilizing agent. In other embodiments, the estradiol added is in a non-micronized form. Mixing may be facilitated by an impeller, agitator, stirrer, or other like devices used to mix pharmaceutical compositions. Operation 106 may be performed under an inert or relatively inert gas atmosphere, such as nitrogen gas.


In embodiments, however, the addition of estradiol may be performed prior to operation 104. In that regard, operations 104 and 106 are interchangeable with respect to timing or can be performed contemporaneously with each other.


In operation 110, the gelatin delivery vehicle is prepared. Any of the gelatin delivery vehicles described herein may be used in operation 110. In embodiments, gelatin, hydrolyzed collagen, glyercin, and other excipients are combined at a temperature range from about 45° C. to about 85° C. and prepared as a film. Mixing may occur in a steel tank or other container used for preparing gelatin delivery vehicles. Mixing may be facilitated by an impellor, agitator, stirrer, or other devices used to combine the contents of gelatin delivery vehicles. Operation 110 may be performed under an inert or relatively inert gas atmosphere, such as nitrogen gas. In embodiments, the gelatin delivery vehicle mixture is degassed prior to being used to encapsulate the pharmaceutical composition.


In operation 112, the gelatin delivery vehicle encapsulates the pharmaceutical composition, according to protocols well known to persons of ordinary skill in the art. In operation 112, a soft gelatin capsule delivery vehicle is prepared by combining the pharmaceutical composition made in operation 106 with the gelatin delivery vehicle made in operation 110. The gelatin may be wrapped around the material, partially or fully encapsulating it or the gelatin can also be injected or otherwise filled with the pharmaceutical composition made in operation 106.


In embodiments, operation 112 is completed in a suitable die to provide a desired shape. Vaginal soft gel capsules may be prepared in a variety of geometries. For example, vaginal soft gel capsules may be shaped as a tear drop, a cone with frustoconical end, a cylinder, a cylinder with larger “cap” portion as illustrated in FIG. 2, or other shapes suitable for insertion into the vagina. The resulting pharmaceutical composition encapsulated in the soft gelatin delivery vehicle may be inserted digitally or with an applicator.


Example 7: Study of Estradiol Pharmaceutical Composition on the Improvement of Vulvovaginal Atrophy (VVA)

The objective of this study was designed to evaluate the efficacy and safety of a pharmaceutical composition comprising 10 μg estradiol (i.e., Pharmaceutical Composition 2) in treating moderate to severe symptoms of VVA associated with menopause after 14 days of treatment, and to estimate the effect size and variability of vulvovaginal atrophy endpoints. In addition, the systemic exposure to estradiol from single and multiple doses of the pharmaceutical composition was investigated.


This study was a phase 1, randomized, double-blind, placebo-controlled trial to evaluate safety and efficacy of the pharmaceutical composition in reducing moderate to severe symptoms of vaginal atrophy associated with menopause and to investigate the systemic exposure to estradiol following once daily intravaginal administrations of a pharmaceutical composition for 14 days.


Postmenopausal subjects who met the study entry criteria were randomized to one of two treatment groups (pharmaceutical composition or placebo). During the screening period subjects were asked to self-assess the symptoms of VVA, including vaginal dryness, vaginal or vulvar irritation or itching, dysuria, vaginal pain associated with sexual activity, and vaginal bleeding associated with sexual activity. Subjects with at least one self-assessed moderate to severe symptom of VVA identified by the subject as being most bothersome to her were eligible to participate in the study.


Clinical evaluations were performed at the following time points:


Screening Period (up to 28 days);


Visit 1—Randomization/Baseline (day 1);


Visit 2—Interim (day 8); and


Visit 3—End of the treatment (day 15).


Eligible subjects were randomized in a 1:1 ratio to receive either pharmaceutical composition comprising estradiol 10 μg or a matching placebo vaginal softgel capsule, and self-administered their first dose of study medication at the clinical facility under the supervision of the study personnel. Serial blood samples for monitoring of estradiol level were collected at 0.0, 1.0, 3.0, and 6.0 hours relative to first dose administration on day 1. Subjects remained at the clinical site until completion of the 6-hour blood draw and returned to clinical facility for additional single blood draws for measurement of estradiol concentration on day 8 (before the morning dose) and day 15. Subjects were provided with enough study medication until the next scheduled visit and were instructed to self-administer their assigned study treatment once a day intravaginally at approximately the same time (±1 hour) every morning. Each subject was provided with a diary in which she was required to daily record investigational drug dosing dates and times. Subjects returned to clinical facility on day 8 for interim visit and on day 15 for end of treatment assessments and post study examinations. Capsule disintegration state was assessed by the investigator at day 1 (6 hours post-dose) and day 15.


The study involved a screening period of up to 28 days before randomization and treatment period of 14 days. Selection of dosage strength (estradiol 10 μg) and treatment regimen (once daily for two weeks) was based on the FDA findings on safety and efficacy of the RLD.


Number of Subjects (Planned and Analyzed)


Up to 50 (25 per treatment group) postmenopausal female subjects 40 to 75 years old with symptoms of moderate to severe VVA were randomized. 50 subjects were enrolled, 48 subjects completed the study, and 48 subjects were analyzed.


Diagnosis and Main Criteria for Inclusion


Fifty female subjects were enrolled in the study. Post-menopausal female subjects 40 to 75 years of age, with a mean age was 62.3 years were enrolled. Subjects' mean weight (kg) was 71.2 kg with a range of 44.5-100 kg. Subjects' mean height (cm) was 162.6 cm with a range of 149.9-175.2 cm, and the mean BMI (kg/m2) was 26.8 kg/m2 with a range of 19-33 kg/m2. Criteria of inclusion in the study included: self-identification of at least one moderate to severe symptom of VVA, for example, vaginal dryness, dysparuenia, vaginal or vulvar irritation, burning, or itching, dysuria, vaginal bleeding associated with sexual activity, that was identified by the subject as being most bothersome to her; ≤5% superficial cells on vaginal smear cytology; vaginal pH>5.0; and estradiol level ≤50 pg/ml. Subject who were judged as being in otherwise generally good health on the basis of a pre-study physical examination, clinical laboratory tests, pelvic examination, and mammography were enrolled.


Estradiol 10 μg or Placebo, Dose, and Mode of Administration


Subjects were randomly assigned (in 1:1 allocation) to self-administer one of the following treatments intravaginally once daily for 14 days:

    • Treatment A: The pharmaceutical composition of Example 5 (Pharmaceutical Composition 2: 10 μg estradiol); or
    • Treatment B: Placebo vaginal softgel capsule, containing the same formulation as Treatment A, except for the 10 μg of estradiol.


The estradiol formulation was a tear drop shaped light pink soft gel capsule. Treatment B had the same composition, appearance, and route of administration as the Treatment A, but contained no estradiol.


Duration of Treatment


The study involved a screening period of up to 28 days before randomization and a treatment period of 14 days.


Criteria for Evaluation


Efficacy Endpoints:

    • Change from baseline (screening) to day 15 in the Maturation Index (percent of parabasal vaginal cells, superficial vaginal cells, and intermediate vaginal cells) of the vaginal smear. Data for this endpoint are shown in Tables 6-8.
    • Change from baseline (screening) to day 15 in vaginal pH. Data for this endpoint are shown in Table 9.
    • Change from baseline (randomization) to day 15 in severity of the most bothersome symptoms: (1) vaginal dryness; (2) vaginal or vulvar irritation, burning, or itching; (3) dysuria; (4) dysparuenia; (5) vaginal bleeding associated with sexual activity. Data for this endpoint are shown in Tables 13 and 15.
    • Change from baseline (randomization) to day 15 in investigator's assessment of the vaginal mucosa. Data for this endpoint are shown in Tables 18-21.


Unless otherwise noted, the efficacy endpoints were measured as a change-from Visit 1—Randomization/Baseline (day 1) to Visit 3—End of the treatment (day 15), except for vaginal bleeding which was expressed as either treatment success or failure.


Other endpoints include:

    • Vital signs, weight, changes in physical exam, pelvic and breast exam, and adverse events were evaluated as part of the safety endpoints.
    • Concentration of estradiol at each sampling time.
    • Peak concentration of estradiol on day 1 and sampling time at which peak occurred.
    • Delivery vehicle disintegration to measure the amount of residual delivery vehicle remains in the vagina post treatment.


Results from the assessment of plasma concentrations of estradiol are presented in Table 5.









TABLE 5







Safety Results: The descriptive statistics for Day 1


plasma estradiol Cmax and Tmax are provided below.










Estradiol 10 μg
Placebo












Cmax
Tmax
Cmax
Tmax














N
24
24
26
26


Mean ± SD
30.7 ± 7.47
2.12 ± 1.73
27.5 ± 17.26
4.00 ± 2.68


Geometric
29.9

24.7



Mean






Median
29.8
1.00
22.1
6.00


Min, Max
19.7, 52.3
1.00, 6.00
15.1, 90.0
0.00, 6.00


CV %
24.3%
81.3%
62.9%
67.1%









Other Endpoints:


Maturation Index Results


Vaginal cytology data was collected as vaginal smears from the lateral vaginal walls according to standard procedures to evaluate vaginal cytology at screening and Visit 3—End of treatment (day 15). The change in the Maturation Index was assessed as a change in cell composition measured at Visit 1—Baseline (day 1) compared to the cell composition measured at Visit 3—End of treatment (day 15). The change in percentage of superficial, parabasal, and intermediate cells obtained from the vaginal mucosal epithelium from a vaginal smear was recorded. Results from these assessments are presented in Tables 6, 7, and 8.









TABLE 6







Primary Efficacy Analysis Results of Change from Baseline (Screening) to Day


15 in the Maturation Index of the Vaginal Smear (Percent Parabasal Cells)

















Difference

Estradiol






Between

10 μg vs.




Estradiol

Treatment
90% CI for
Placebo


Population
Statistics
10 μg
Placebo
Means
Difference
P-value
















Intent-to-
N
24 
24





Treat



Least-
−54.4
−4.80
−49.6
(−60.4, −38.8)
<0.0001



Squares



Mean



Mean ± SD
−53.8 ± 39.7
−5.4 ± 22.3






Median
−60.0
−5.0






Min, Max
−100.0, 0.0
−60.0, 60.0









1 Confidence interval for the estradiol 10 μg-Placebo from ANCOVA with treatment as a fixed effect and baseline as a covariate.




2 P-value for treatment comparison from ANCOVA with treatment as a fixed effect and baseline as a covariate.














TABLE 7







Primary Efficacy Analysis Results of Change from Baseline (Screening) to


Day 15 in the Maturation Index of the Vaginal Smear (Superficial Cells)

















Difference

Estradiol






Between

10 μg vs.




Estradiol

Treatment
90% CI for
Placebo


Population
Statistics
10 μg
Placebo
Means
Difference
P-value
















Intent-to-
N
24  
24





Treat



Least-
35.2
8.75
26.5
(15.4, 37.6)
0.0002



Squares



Mean



Mean ± SD
35.2 ± 26.4
8.8 ± 18.7






Median
40.0
0.0






Min, Max
0.0, 80.0
0.0, 90.0









1 Confidence interval for the estradiol 10 μg-Placebo from ANOVA with treatment as a fixed effect.




2 P-value for treatment comparison from ANOVA with treatment as a fixed effect.














TABLE 8







Primary Efficacy Analysis Results of Change from Baseline (Screening) to


Day 15 in the Maturation Index of the Vaginal Smear (Intermediate Cells)

















Difference

Estradiol






Between

10 μg vs.




Estradiol

Treatment
90% CI for
Placebo


Population
Statistics
10 μg
Placebo
Means
Difference
P-value2
















Intent-to-
N
24  
24





Treat



Least-
18.7
−3.54
22.3
(11.1, 33.5)
0.0017



Squares



Mean



Mean ± SD
18.5 ± 42.7
−3.3 ± 21.6






Median
22.5
−5.0






Min, Max
−60.0, 100.0
−60.0, 20.0









1 Confidence interval for the estradiol 10 μg-Placebo from ANCOVA with treatment as a fixed effect and baseline as a covariate.




2P-value for treatment comparison from ANCOVA with treatment as a fixed effect and baseline as a covariate.







Change in pH Results


Vaginal pH was measured at Screening and Visit 3—End of treatment (day 15). The pH measurement was obtained by pressing a pH indicator strip against the vaginal wall. The subjects entering the study were required to have a vaginal pH value greater than 5.0 at screening. pH values were recorded on the subject's case report form. The subjects were advised not to have sexual activity and to refrain from using vaginal douching within 24 hours prior to the measurement. Results from these assessments are presented in Table 9.









TABLE 9







Primary Efficacy Analysis Results of Change from


Baseline (Screening) to Day 15 in Vaginal pH

















Difference

Estradiol






Between

10 μg vs.




Estradiol

Treatment
90% CI for
Placebo


Population
Statistics
10 μg
Placebo
Means
Difference1
P-value2
















Intent-to-
N
24
24





Treat



Least-
−0.974
−0.339
−0.635
(−0.900, −0.368)
0.0002



Squares



Mean



Mean ± SD
−0.917 ± 0.686
−0.396 ± 0.659






Median
−1.00
−0.500






Min, Max
−2.00, 0.500
−1.50, 0.500









1Confidence interval for the estradiol 10 μg-Placebo from ANCOVA with treatment as a fixed effect and baseline as a covariate.




2P-value for treatment comparison from ANCOVA with treatment as a fixed effect and baseline as a covariate.







Most Bothersome Symptoms Data


Subjects were asked to specify the symptom that she identified as the “most bothersome symptom.” During the screening period all of the subjects were provided with a questionnaire to self-assess the symptoms of VVA: (1) vaginal dryness; (2) vaginal or vulvar irritation, burning, or itching; (3) dysuria; (4) dysparuenia; (5) vaginal bleeding associated with sexual activity. Each symptom, with the exception of vaginal bleeding associated with sexual activity, was measured on a scale of 0 to 3, where 0=none, 1=mild, 2=moderate, and 3=severe. Vaginal bleeding associated with sexual activity was measured in a binary scale: N=no bleeding; Y=bleeding. The subject's responses were recorded. All randomized subjects were also provided a questionnaire to self-assess the symptoms of VVA at Visit 1—Randomization/Baseline (day 1) and at Visit 3—End of the treatment (day 15). Subjects recorded their self-assessments daily in a diary and answers were collected on days 8 and 15 (end of treatment). Pre-dose evaluation results obtained at Visit 1 were considered as baseline data for the statistical analyses. Data from these assessments are presented in Tables 10 and 11.









TABLE 10







Baseline Characteristics for Vaginal


Atrophy Symptoms (ITT Population)















Estradiol 10 μg




Estradiol

vs. Placebo


WA Symptom
Statistics
10 μg
Placebo
P-value1














Vaginal dryness
N of
24
24




Subjects






Mean
2.292
2.375
0.68231


Vaginal or vulvar
N of
24
24



irritation/
Subjects





burning/itching
Mean
0.875
1.333
0.08721


Pain, burning or
N of
24
24



stinging when
Subjects





urinating
Mean
0.583
0.625
0.87681


Vaginal pain
N of
12
12



associated
Subjects2





with sexual
Mean
2.083
2.333
0.54281


activity






Vaginal bleeding
N of
12
12



associated with
Subjects2





sexual activity
Percent3
25.00
33.33
0.31463






1P-value for treatment comparison from ANOVA/ANCOVA with treatment as a fixed effect and Baseline as a covariate when appropriate.




2N = number of subjects sexually active at baseline.




3Percent of subjects with bleeding, evaluated using Fisher's Exact Test.














TABLE 11







Additional Efficacy Analysis Results of Change from Baseline (Randomization)


to Day 15 in Severity of Vaginal Atrophy Symptoms











Difference

Estradiol












Least-Squares Mean
Between

10 μg vs.














Statistical
Estradiol

Treatment
90% CI for
Placebo


Symptom
Method1
10 μg
Placebo
Means
Difference2
P-value
















Vaginal
ANCOVA
0.980
0.729
0.251
−0.706, 0.204)
0.3597


dryness


Vaginal or
ANCOVA
0.694
0.514
0.180
−0.549, 0.189)
0.4159


vulvar


Irritation/


buring/


itching


Pain/Burning/
ANCOVA
0.391
0.359
0.032
−0.263, 0.200)
0.8185


Stinging


(Urination)


Vaginal pain
ANOVA
0.800
0.500
0.300
−1.033, 0.433)
0.4872


associated


with sexual


activity






1ANOVA model contained a fixed effect for treatment. ANCOVA added baseline as a covariate to the model.




2Confidence interval for the difference between estradiol 10 μg and Placebo treatment least-squares means.







Changes to the most bothersome symptom from the baseline was scored according to the evaluation of VVA symptoms generally set forth above. Tables 13 and 14 show a comparison between the pharmaceutical composition 1 and placebo generally for most bothersome symptom and vaginal atrophy symptom. It is noteworthy to point out that these measurement demonstrated a trend of improvement, though not statistically significant, at day 15.









TABLE 13







Primary Efficacy Analysis Results of Change from Baseline (Randomization)


to Day 15 in Severity of the Most Bothersome VVA

















Difference

Estradiol






Between

10 μg vs.




Estradiol

Treatment
90% CI for
Placebo


Population
Statistics
10 μg
Placebo
Means
Difference1
P-value2
















Intent-to-
N
24
24





Treat



Least-
−1.043
−1.042
−0.002
(−0.497, 0.493)
0.9951



Squares



Mean



Mean ± SD
−1.043 ± 0.928
−1.042 ± 1.08






Median
−1.00
−1.00






Min, Max
−3.00, 0.00
−3.00, 0.00









1Confidence interval for the estradiol 10 μg-Placebo from ANOVA with treatment as a fixed effect.




2P-value for treatment comparison from ANOVA with treatment as a fixed effect.














TABLE 14







Additional Efficacy Analysis Results of Change from Baseline (Randomization)


to Day 15 in Severity of Vaginal Atrophy Symptoms Symptom









TX-12-












Least-Squares
Difference

004-HR



Mean
Between

vs.














Statistical
TX-12-

Treatment
90% CI for
Placebo


Symptom
Method1
004-HR
Placebo
Means
Difference2
P-value
















Dryness
ANCOVA
−0.980
−0.729
−0.251
(−0.706, 0.204)
0.3597


Irritation
ANCOVA
−0.694
−0.514
−0.180
(−0.549, 0.189)
0.4159


Pain (Sex)
ANOVA
−0.800
−0.500
−0.300
(−1.033, 0.433)
0.4872


Pain/Burning/
ANCOVA
−0.391
−0.359
−0.032
(−0.263, 0.200)
0.8185


Stinging


(Urination)






1ANOVA model contained a fixed effect for treatment. ANCOVA added baseline as a covariate to the model.




2Confidence interval for the difference between TX-12-004-HR and Placebo treatment least-squares means.







With respect to the most bothersome symptoms data presented in Tables 13 and 14, the period over which the data was measured is generally considered insufficient to make meaningful conclusions. However, the trends observed as part of this study suggest that the data will show improvement of the most bothersome symptoms when data for a longer time period is collected.


The absence or presence of any vaginal bleeding associated with sexual activity was also measured as one of the most bothersome symptoms. The data for vaginal bleeding associated with sexual activity is reported in Table 15.









TABLE 15







Primary Efficacy Analysis Results of Change from


Baseline (Randomization) to Day 15 in Vaginal


Bleeding Associated with Sexual Activity









Baseline (Randomization) and Day 15



Summary of Vaginal Bleeding














Bleeding/
Bleeding/
No Bleeding/
No Bleeding/




No Bleeding
Bleeding
Bleeding
No Bleeding


Treatment
N*
(Success)2
(Failure)
(Failure)
(NC)
















Estradiol
10
2
(100%)
0
0
8


10 μg








Placebo
10
1
(20%)
3
1
5











P-Value for
0.1429





Estradiol 10 μg






vs. Placebo1





*N = Total number of patients within each treatment group who were sexually active at both Baseline and Day 15 and provided a response at both visits.


NC = No Change - not considered in the statistical comparison.



1P-value for treatment comparison from Fisher's Exact Test.




2Percent is based on the number of subjects classified as either a Success or a Failure (N = 2 for estradiol 10 μg; N = 5 for Placebo







Estradiol Level/Pharmacokinetics Data


In this study, the systemic exposure to estradiol following once daily intravaginal administration of estradiol 10 μg for 14 days was investigated. Descriptive statistics of the plasma estradiol concentrations taken at each sampling time and the observed Cmax and Tmax values were recorded in Tables 16 and 17. No statistically significant difference in the systemic concentration of estradiol 10 μg versus the placebo group was observed, which suggests the estradiol is not carried into the blood stream where it will have a systemic effect. Rather, it remains in localized tissues; the effect of estradiol is therefore believed be local to the location of administration (i.e., the vagina). The lower limits of detection of the assays used to measure the pharmacokinetic data may have affected the measured the accuracy of the pk values presented. Additional pk studies were performed with more accurate assays in Examples 8 and 9.


For the purpose of monitoring the estradiol level during the study blood samples were collected at 0.0, 1.0, 3.0, and 6.0 hours relative to dosing on day 1; prior to dosing on day 8; and prior to dosing on day 15. Efforts were made to collect blood samples at their scheduled times. Sample collection and handling procedures for measurement of estradiol blood level was performed according to procedure approved by the sponsor and principal investigator. All baseline and post-treatment plasma estradiol concentrations were determined using a validated bioanalytical (UPLC-MS/MS) methods. These data are shown in Tables 16 and 17.









TABLE 16







Descriptive Statistics of Estradiol Concentrations (pg/ml) at Each Sampling Time









Sampling Time


















Pre-dose
Pre-dose


Treatment
0 Hour
1 Hour
3 Hours
6 Hours
Day 8
Day 15





Estradiol








10 μg


N
24  
24  
24  
24  
24  
22  


Mean ± SD
20.1 ± 5.74
28.7 ± 5.89
25.7 ± 5.71
23.4 ± 7.91 
21.4 ± 9.28 
23.4 ± 8.72 


Median
20.2
28.9
24.7
22.3
20.7
20.7


Min, Max
2.63, 38.3
18.8, 43.9
19.3, 47.5
3.31, 52.3
2.09, 52.2
17.9, 54.7


Placebo


N
26  
26  
26  
26  
25  
24  


Mean ± SD
20.5 ± 4.29
21.0 ± 6.14
19.0 ± 5.92
26.9 ± 17.36
29.9 ± 22.51
28.1 ± 16.80


Median
20.8
20.8
20.9
21.7
21.6
21.1


Min, Max
4.03, 29.1
3.19, 41.2
3.15, 26.9
15.1, 90.0
15.0, 116.2
14.7, 81.3
















TABLE 17







Descriptive Statistics of Estradiol Cmax and Tmax on Day 1










Estradiol 10 μg
Placebo












Cmax
Tmax
Cmax
Tmax














N
24
24
26
26


Mean ± SD
30.7 ± 7.47
2.12 ± 1.73
27.5 ± 17.26
4.00 ± 2.68


Geometric
29.9

24.7



Mean






Median
29.8
1.00
22.1
6.00


Min, Max
19.7, 52.3
1.00, 6.00
15.1, 90.0
0.00, 6.00


CV %
24.3%
81.3%
62.9%
67.1%









Assessment of Vaginal Mucosa Data


The investigators rated the vaginal mucosal appearance at day 1 (pre-dose) and day 15. Vaginal color, vaginal epithelial integrity, vaginal epithelial surface thickness, and vaginal secretions were evaluated according to the following degrees of severity: none, mild, moderate, or severe using scales 0 to 3, where 0=none, 1=mild, 2=moderate, and 3=severe. Results from these investigators rated assessments are presented in Tables 18, 19, 20, and 21.









TABLE 18







Primary Efficacy Analysis Results of Change from Baseline (Randomization) to Day 15


in Investigator's Assessment of the Vaginal Mucosa (Assessment of Vaginal Color)

















Difference

Estradiol






Between

10 μg vs.




Estradiol

Treatment
90% CI for
Placebo


Population
Statistics
10 μg
Placebo
Means
Difference1
P-value2
















Intent-to-
N
24
24





Treat



Least-
−0.199
−0.009
−0.191
(−0.434, 0.052)
0.1945



squares



Mean



Mean ± SD
−0.333 ± 0.565
0.125 ± 0.741






Median
0.00
0.00






Min, Max
−2.00, 0.00
−1.00, 2.00









1Confidence interval for the estradiol 10 μg-Placebo from ANCOVA with treatment as a fixed effect and baseline as a covariate.




2P-value for treatment comparison from ANCOVA with treatment as a fixed effect and baseline as a covariate.














TABLE 19







Primary Efficacy Analysis Results of Change from Baseline (Randomization)


to Day 15 in Investigator's Assessment of the Vaginal Mucosa


(Assessment of Vaginal Epithelial Integrity)

















Difference

Estradiol






Between

10 μg vs.




Estradiol

Treatment
90% CI for
Placebo


Population
Statistics
10 μg
Placebo
Means
Difference1
P-value2
















Intent-to-
N
24
24





Treat



Least-
−0.342
0.176
−0.518
(−0.726, −0.311)
0.0001



squares



Mean



Mean ± SD
−0.417 ± 0.584
0.250 ± 0.442






Median
0.00
0.00






Min, Max
−1.00, 1.00
0.00, 1.00









1Confidence interval for the estradiol 10 μg-Placebo from ANCOVA with treatment as a fixed effect and baseline as a covariate.




2P-value for treatment comparison from ANCOVA with treatment as a fixed effect and baseline as a covariate.














TABLE 20







Primary Efficacy Analysis Results of Change from Baseline (Randomization)


to Day 15 in Investigator's Assessment of the Vaginal Mucosa


(Assessment of Vaginal Epithelial Surface Thickness)

















Difference

Estradiol






Between

10 μg vs.




Estradiol

Treatment
90% CI for
Placebo


Population
Statistics
10 μg
Placebo
Means
Difference1
P-value2
















Intent-to-
N
24
24





Treat



Least-
−0.034
−0.133
0.099
(−0.024, 0.221)
0.1820



squares



Mean



Mean ± SD
−0.125 ± 0.338
−0.042 ± 0.550






Median
0.00
0.00






Min, Max
−1.00, 0.00
−1.00, 1.00









1Confidence interval for the estradiol 10 μg-Placebo from ANCOVA with treatment as a fixed effect and baseline as a covariate.




2P-value for treatment comparison from ANCOVA with treatment as a fixed effect and baseline as a covariate.














TABLE 21







Primary Efficacy Analysis Results of Change from Baseline (Randomization) to Day 15


in Investigator's Assessment of the Vaginal Mucosa (Assessment of Vaginal Secretions)

















Difference

Estradiol






Between

10 μg vs.




Estradiol

Treatment
90% CI for
Placebo


Population
Statistics
10 μg
Placebo
Means
Difference1
P-value2
















Intent-to-
N
24
24





Treat



Least-
−0.643
−0.274
−0.369
(−0.661, −0.076)
0.0401



squares



Mean



Mean ± SD
−0.792 ± 0.779
−0.125 ± 0.741






Median
−1.00
0.00






Min, Max
−2.00, 1.00
−2.00, 2.00









1Confidence interval for the estradiol 10 μg-Placebo from ANCOVA with treatment as a fixed effect and baseline as a covariate.




2P-value for treatment comparison from ANCOVA with treatment as a fixed effect and baseline as a covariate.







Delivery Vehicle Disintegration Data


Assessment of capsule disintegration in the vagina (presence or absence) at Day 1 (6 hours after dosing) and Day 15. Results of this assessment is presented in Table 22.









TABLE 22







Capsule Disintegration State in the Vagina on Day 1 and Day 15










Estradiol 10 μg
Placebo












Day 1
Day 15
Day 1
Day 15


















No evidence
23
(95.8%)
24
(100.0%)
26
(100.0%)
24
(92.3%)


of capsule










present










Evidence
0
(0.0%)
0
(0.0%)
0
(0.0%)
0
(0.0%)


of capsule










present










Assessment
1
(4.2%)
0
(0.0%)
0
(0.0%)
22
(7.7%)


not done









Serum hormone level data was collected to measure the serum concentrations of estradiol. These data were used for screening inclusion and were determined using standard clinical chemistry methods.


Appropriateness of Measurements


The selection of the efficacy measurements used in this study was based on FDA's recommendations for studies of estrogen and estrogen/progestin drug products for the treatment of moderate to severe vasomotor symptoms associated with the menopause and moderate to severe symptoms of vulvar and vaginal atrophy associated with the menopause (Food and Drug Administration, Guidance for Industry, Estrogen and Estrogen/Progestin Drug Products to Treat Vasomotor Symptoms and Vulvar and Vaginal Atrophy Symptoms—Recommendations for Clinical Evaluation. January 2003, hereby incorporated by reference).


Standard clinical, laboratory, and statistical procedures were utilized in the trial. All clinical laboratory procedures were generally accepted and met quality standards.


Statistical Methods:


Efficacy:


Analysis of variance (ANOVA) was used to evaluate the change from baseline differences between the subjects receiving estradiol 10 μg and placebo capsules for all efficacy endpoints, except for vaginal bleeding, to estimate the effect size and variability of the effect. In some cases, for example, for some vaginal atrophy symptoms, the change from baseline (post dose response) was correlated with the baseline value (p<0.05), so baseline was included as a covariate to adjust for this correlation (Analysis of Covariance, ANCOVA). The 90% confidence intervals on the differences between estradiol 10 μg and placebo endpoint means were determined to evaluate the effect size. The change from baseline in vaginal bleeding associated with sexual activity was evaluated in terms of the proportion of subjects who had treatment success or failure. Any subject reporting bleeding at baseline who did not report bleeding at Day 15 was considered to have been successfully treated. Any subject reporting bleeding at day 15 was considered a treatment failure, regardless of whether they reported baseline bleeding or not. Subjects reporting no bleeding at both baseline and day 15 were classified as no-change and were excluded from the statistical evaluation. The difference in the proportion of subjects with success between the two treatment groups was statistically evaluated using Fisher's Exact Test. Results of this difference in proportion are presented in Table 10.


Measurements of Treatment Compliance


Subjects were required to complete a diary in order to record treatment compliance. Diaries were reviewed for treatment compliance at day 8 and day 15 visits. A total of 45 subjects (21 subjects in the estradiol 10 μg group and 24 subjects in the placebo group) were 10% compliant with the treatment regimen.


Due to the investigative nature of the study, no adjustments were made for multiplicity of endpoints.


Safety:


The frequency and severity of all adverse events were summarized descriptively by treatment group.


Results: All forty eight (48) subjects who completed the study were included in the primary efficacy analyses. The results of efficacy analyses are presented throughout Tables 5, 6, and 7.


CONCLUSIONS

Efficacy


The two-week treatment with pharmaceutical composition 10 μg led to a statistically significant greater mean decrease in percent of parabasal cells than did placebo treatment (54% vs. 5%, p<0.0001), as illustrated in Table 6. At the same time, a significantly greater mean increase in the percent of superficial cells was observed with the pharmaceutical composition (35%) than with the placebo capsules (9%), with the difference being highly statistically significant (p=0.0002), as illustrated in Table 7. The difference in pH reduction between the pharmaceutical composition (0.97 units) compared to that for the placebo (0.34 units) was only slightly greater than 0.5 units, but the difference was detected as statistically significant (p=0.0002), as illustrated in Table 9.


While the decrease in severity of the most bothersome symptom was essentially the same (˜1 unit) for both pharmaceutical composition and placebo, the reductions in the severity of the individual symptoms of vaginal dryness, irritation and pain during sexual activity were all marginally better for the active treatment than for the placebo treatment. None of the differences between the two treatments, all of which were ≤0.3 units, were detected as statistically significant. There was no difference between the two treatments in regard to reduction of pain/burning/stinging during urination (˜0.4 unit reduction). The length of the study was not long enough to show a separation between the most bothersome symptoms in the pharmaceutical composition and placebo. However, the trends of most bothersome symptoms suggest that with a suitable period of time, significantly significant differences between the two treatments would be observed.


The two-week treatment with estradiol 10 μg capsules showed no statistically detectable difference in regard to reduction of severity from baseline according to the investigator's assessment of vaginal color or vaginal epithelial surface thickness. Pharmaceutical composition capsules did demonstrate a statistically significant greater reduction than did placebo in severity of atrophic effects on vaginal epithelial integrity (−0.34 vs. 0.18, p=0.0001) and vaginal secretions (−0.64 vs. −0.27, p=0.0401).


Descriptive statistical analyses (mean, median, geometric mean, standard deviation, CV, minimum and maximum, Cmax, and Tmax) were conducted on the estradiol concentrations at each sampling time, the peak concentration on day 1 and the time of peak concentration. Results from this assessment are presented in Tables 16 and 17.


A pharmaceutical composition comprising estradiol 10 μg outperformed placebo treatment in regard to improvement in the Maturation Index, reduction in vaginal pH, reduction in the atrophic effects on epithelial integrity and vaginal secretions. The lack of statistical significance between the two treatments in regard to reduction of severity for the most bothersome symptom, and the individual vaginal atrophy symptoms of dryness, irritation, pain associated with sexual activity, and pain/burning/stinging during urination, is not unexpected given the small number of subjects in the study and the short duration of therapy. Too few subjects in the study had vaginal bleeding associated with sexual activity to permit any meaningful evaluation of this vaginal atrophy symptom.


Of the 48 subjects enrolled in the study, 45 subjects were 100% compliant with the treatment regimen. Of the remaining three subjects, one removed herself from the study due to personal reasons and the other two subjects each missed one dose due to an adverse event.


Safety


Although the Day 1 mean plasma estradiol peak concentration for the pharmaceutical composition was somewhat higher than that for the Placebo (ratio of geometric means=1.21:Test Product (estradiol 10 μg) 21%>Placebo), no statistically significant difference was determined. However, the assay methods were questionable, resulting in questionable pk data. Additional pk studies were performed in Examples 8 and 9.


There were no serious adverse events in the study.


Overall, the pharmaceutical composition comprising estradiol 10 μg was well tolerated when administered intravaginally in once daily regimen for 14 days.


Example 8: pk Study (25 μg Formulation)

A pk study was undertaken to compare the 25 μg formulation disclosed herein (Pharmaceutical Composition 3) to the RLD. The results of the pk study for estradiol are summarized in Table 23. The p values for these data demonstrate statistical significance, as shown in Table 24.









TABLE 23







Statistical Summary of the Comparative Bioavailability


Data for Unscaled Average BE studies of Estradiol, Least


Square Geometric Means of Estradiol, Ratio of Means and


90% Confidence Intervals, Fasting/Fed Bioequivalence Study


(Study No.: ESTR-1K-500-12); Dose 25 μg estradiol


















Ratio



Parameter
Test
N
RLD
N
(%)
90% C.I.
















Cmax
23.0839
36
42.7024
36
54.06
44.18-66.14


(pg/mL)


AUC0-24
89.2093
36
292.0606
36
30.54
23.72-39.34


(pg · hr/mL)
















TABLE 24







P-values for table 23










P-Value












Effect
Cmax
AUC0-24















Treatment
<.0001
<.0001



Sequence
0.4478
0.5124



Period
0.4104
0.7221










As illustrated in Table 23, baseline adjusted pk data illustrates that the formulations disclosed herein unexpectedly show a 54% decrease in Cmax and a 31% decrease in the AUC relative to the RLD. This result is desirable because the estradiol is intended only for local absorption. These data suggest a decrease in the circulating levels of estradiol relative to the RLD. Moreover, it is noteworthy to point out that the Cmax and AUC levels of estradiol relative to placebo are not statistically differentiable, which suggests that the formulations disclosed herein have a negligible systemic effect. As shown in Table 24, there was no significant difference between the test and reference products due to sequence and period effects. However, there was a significant difference due to treatment effect for both Cmax and AUC.


Pharmacokinetics for circulating total estrone, a metabolite of estradiol, is show in Table 25. These data show that the total circulating estrone for the formulations disclosed herein resulted in a 55% decrease in the Cmax for circulating estrone, and a 70% decrease in the AUC for circulating estrone.









TABLE 26







P-values for table 25










P-Value












Effect
Cmax
AUC0-24















Treatment
0.0002
<.0001



Sequence
0.1524
0.0464



Period
0.0719
0.0118

















TABLE 25







Statistical Summary of the Comparative Bioavailability


Data for Unscaled Average BE studies of Estrone, Least


Square Geometric Means, Ratio of Means and 90% Confidence


Intervals, Fasting/Fed Bioequivalence Study (Study


No.: ESTR-1K-500-12); Dose 25 μg estradiol


















Ratio



Parameter
Test
N
RLD
N
(%)
90% C.I.
















Cmax
10.7928
36
23.5794
36
45.77
32.95 to 63.59


(pg/mL)


AUC0-24
51.2491
36
165.4664
36
30.97
19.8-48.45


(pg · hr/mL)









There was a significant difference between test and reference products due to treatment effect whereas there was no significant difference due to sequence and period effects for Cmax. For AUC, there was a significant difference between test and reference products due to treatment, sequence, and period effects.


pk for circulating total estrone sulfate is shown in Table 27. These data show that the total circulating estrone sulfate for the pharmaceutical compositions disclosed herein resulted in a 33% decrease in the Cmax and a 42% decrease in the AUC for circulating estrone sulfate.









TABLE 27







Statistical Summary of the Comparative Bioavailability Data


for Unsealed Average BE studies of Estrone Sulfate, Least


Square Geometric Means of Estrone Sulfate, Ratio of Means


and 90% Confidence Intervals, Fasting/Fed Bioequivalence


Study (Study No.: ESTR-1K-500-12); Dose 25 μg estradiol


















Ratio



Parameter
Test
N
RLD
N
(%)
90% C.I.
















Cmax
490.0449
36
730.5605
36
67.08
53.84-83.57


(pg/mL)


AUC0-24
4232.9914
36
7323.0827
36
57.80
43.23-77.29


(pg · hr/mL)
















TABLE 28







P-values for table 27










P-Value












Effect
Cmax
AUC0-24















Treatment
0.0042
0.0031



Sequence
0.5035
0.9091



Period
0.1879
0.8804










There was a significant difference between test and reference products due to treatment effect whereas there was no significant difference due sequence and period effects for both Cmax and AUC.


Example 9: pk Study (10 μg Formulation)

A pk study was undertaken to compare the 10 μg formulation disclosed herein (Pharmaceutical Composition 2) to the RLD. The results of the pk study for estradiol are summarized in Table 29-40, and FIGS. 9-14.


A pk study was undertaken to compare pharmaceutical compositions disclosed herein having 10 μg of estradiol to the RLD. The results of the pk study for estradiol are summarized in tables 29-34, which demonstrate that the pharmaceutical compositions disclosed herein more effectively prevented systemic absorption of the estradiol. Table 35 shows that the pharmaceutical compositions disclosed herein had a 28% improvement over the RLD for systemic blood concentration Cmax and 72% AUC improvement over the RLD.









TABLE 29







Summary of Pharmacokinetic Parameters of Test product


(T) of Estradiol - Baseline adjusted (N = 34)













Arithmetic






Pharmacokinetic
Mean ± Standard
Coefficient


Parameter
Deviation
of Variation
Median
Minimum
Maximum















Cmax (pg/mL)
15.7176 ± 7.9179 
50.3761
13.9000
6.5000
49.6000


AUC0-24
53.0100 ± 19.5629
36.9041
49.9750
24.3000
95.1500


(pg · hr/mL)


tmax (hr)
1.98 ± 1.29
65.34
2.00
1.00
8.05
















TABLE 30







Summary of Pharmacokinetic Parameters of Reference product


(R) of Estradiol - Baseline adjusted (N = 34)













Arithmetic






Pharmacokinetic
Mean ± Standard
Coefficient


Parameter
Deviation
of Variation
Median
Minimum
Maximum















Cmax (pg/mL)
 24.1882 ± 11.9218
49.2877
24.1500
1.0000
55.3000


AUC0-24
163.8586 ± 72.0913
43.9960
158.0375
2.0000
304.8500


(pg · hr/mL)


tmax (hr)
10.53 ± 5.58
52.94
8.06
2.00
24.00
















TABLE 31







Geometric Mean of Test Product (T) and Reference product


(R) of Estradiol - Baseline adjusted (N = 34)









Geometric Mean









Pharmacokinetic Parameter
Test Product (T)
Reference Product (R)












Cmax (pg/mL)
14.3774
20.3837


AUC0-24 (pg · hr/mL)
49.6231
132.9218


tmax (hr)
1.75
9.28
















TABLE 32







Statistical Results of Test product (T) versus Reference


product (R) for Estradiol - Baseline adjusted (N = 34)










Geometric Least




Square Mean













Test
Reference
Intra
T/R
90%


Pharmacokinetic
Product
Product
Subject
Ratio
Confidence


Parameter
(T)
(R)
CV %
%
Interval















Cmax (pg/mL)
14.4490
20.1980
60.68
71.54*
56.82-90.08


AUC0-24
49.7310
131.0400
70.64
37.95*
29.21-49.31


(pg · hr/mL)





*Comparison was detected as statistically significant by ANOVA (α = 0.05).






The pk data for total estrone likewise demonstrated reduced systemic exposure when compared to the RLD. Table 33 shows the pharmaceutical compositions disclosed herein reduced systemic exposure by 25% for Cmax and 49% for AUC.









TABLE 33







Summary of Pharmacokinetic Parameters of Test product


(T) of Estrone - Baseline adjusted (N = 33)













Arithmetic






Pharmacokinetic
Mean ± Standard
Coefficient


Parameter
Deviation
of Variation
Median
Minimum
Maximum















Cmax (pg/mL)
6.8485 ± 6.5824
96.1149
5.4000
1.3000
36.3000


AUC0-24
34.7051 ± 27.9541
80.5476
30.8500
3.3500
116.7500


(pg · hr/mL)


tmax (hr)
9.12 ± 8.83
96.80
4.00
1.00
24.00
















TABLE 34







Summary of Pharmacokinetic Parameters of Reference


product (R) of Estrone - Baseline adjusted (N = 33)













Arithmetic






Pharmacokinetic
Mean ± Standard
Coefficient


Parameter
Deviation
of Variation
Median
Minimum
Maximum















Cmax (pg/mL)
8.8333 ± 7.1469
80.9086
6.7000
2.7000
30.3000


AUC0-24
63.0042 ± 46.5484
73.8814
51.2800
8.8000
214.0000


(pg · hr/mL)


tmax (hr)
11.16 ± 7.24 
64.95
10.00
4.00
24.00
















TABLE 35







Geometric Mean of Test Product (T) and Reference product


(R) of Estrone - Baseline adjusted (N = 33)









Geometric Mean









Pharmacokinetic Parameter
Test Product (T)
Reference Product (R)












Cmax (pg/mL)
5.1507
6.9773


AUC0-24 (pg · hr/mL)
24.2426
48.2377


tmax (hr)
5.87
9.07
















TABLE 36







Statistical Results of Test product (T) versus Reference


product (R) for Estrone - Baseline adjusted (N = 33)










Geometric Least




Square Mean













Test
Reference
Intra
T/R
90%


Pharmacokinetic
Product
Product
Subject
Ratio
Confidence


Parameter
(T)
(R)
CV %
%
Interval















Cmax (pg/mL)
5.1620
6.9280
47.59
74.50*
61.69-89.97


AUC0-24
24.1960
47.9020
73.66
50.51*
38.37-66.50


(pg · hr/mL)





*Comparison was detected as statistically significant by ANOVA (α = 0.05).






The pk data for estrone sulfate likewise demonstrated reduced systemic exposure when compared to the RLD. Table 37 shows the pharmaceutical compositions disclosed herein reduced systemic exposure by 25% for Cmax and 42% for AUC.









TABLE 37







Summary of Pharmacokinetic Parameters of Test product


(T) of Estrone Sulfate - Baseline adjusted (N = 24)













Arithmetic






Pharmacokinetic
Mean ± Standard
Coefficient


Parameter
Deviation
of Variation
Median
Minimum
Maximum















Cmax (ng/mL)
13.9042 ± 7.0402 
50.6339
11.1500
1.3000
39.0000


AUC0-24
97.9953 ± 80.8861
82.5408
76.2750
5.1025
338.0000


(ng · hr/mL)


tmax (hr)
6.33 ± 4.56
71.93
4.00
4.00
24.00
















TABLE 38







Summary of Pharmacokinetic Parameters of Reference product


(R) of Estrone Sulfate - Baseline adjusted (N = 24)













Arithmetic






Pharmacokinetic
Mean ± Standard
Coefficient


Parameter
Deviation
of Variation
Median
Minimum
Maximum















Cmax (ng/mL)
19.2542 ± 11.3633
59.0173
15.2000
7.0000
53.7000


AUC0-24
177.6208 ± 166.2408
93.5931
124.0000
20.0000
683.0500


(ng · hr/mL)


tmax (hr)
10.33 ± 5.58
54.05
10.00
2.00
24.00
















TABLE 39







Geometric Mean of Test Product (T) and Reference product


(R) of Estrone Sulfate - Baseline adjusted (N = 24)









Geometric Mean









Pharmacokinetic Parameter
Test Product (T)
Reference Product (R)












Cmax (ng/mL)
12.1579
16.8587


AUC0-24 (ng · hr/mL)
66.5996
121.5597


tmax (hr)
5.49
8.83
















TABLE 40







Statistical Results of Test product (T) versus Reference product


(R) for Estrone Sulfate - Baseline adjusted (N = 24)










Geometric Least




Square Mean













Test
Reference
Intra
T/R
90%


Pharmacokinetic
Product
Product
Subject
Ratio
Confidence


Parameter
(T)
(R)
CV %
%
Interval















Cmax (ng/mL)
12.3350
16.5470
48.02
74.55*
59.43-93.51


AUC0-24
68.5260
118.4170
73.87
57.87*
41.68-80.35


(ng · hr/mL)





*Comparison was detected as statistically significant by ANOVA (α = 0.05).






While the pharmaceutical compositions and methods have been described in terms of what are presently considered to be practical and preferred embodiments, it is to be understood that the disclosure need not be limited to the disclosed embodiments. It is intended to cover various modifications and similar arrangements included within the spirit and scope of the claims, the scope of which should be accorded the broadest interpretation so as to encompass all such modifications and similar embodiments. This disclosure includes any and all embodiments of the following claims.

Claims
  • 1. A method for treating vulvovaginal atrophy in a human subject comprising: intravaginally administering estradiol to the subject by manually inserting about two inches into the vagina a liquid pharmaceutical composition comprising about 4 μg to about 25 μg of estradiol, about 90 wt % of a mixture of caprylic and capric triglyceride, and about 10 wt % of a mixture of PEG-6 stearate, ethylene glycol palmitostearate and PEG-32 stearate, wherein the composition has a viscosity from about 50 cP to about 1000 cP at 25° C., wherein estradiol is the only active hormone in the composition, wherein the composition comprises all of the estradiol and is encapsulated in a capsule, and wherein the composition is inserted while the subject is in a reclining position or in a standing position; wherein administration of the composition results in a decrease in the vulvovaginal atrophy in the subject within two to six weeks from the first administration.
  • 2. The method of claim 1, wherein the capsule is a soft gelatin capsule.
  • 3. The method of claim 1, wherein administration of the composition results in an increase in the percentage of vaginal superficial cells within two weeks from the first administration.
  • 4. The method of claim 1, wherein administration of the composition results in a decrease in the percentage of vaginal parabasal cells within two weeks from the first administration.
  • 5. The method of claim 1, wherein administration of the composition results in a decrease in vaginal pH within two weeks from the first administration.
  • 6. The method of claim 1, wherein the administration comprises inserting the composition once daily for two weeks and twice weekly thereafter.
  • 7. The method of claim 1, wherein the vulvovaginal atrophy is due to menopause.
  • 8. The method of claim 1, wherein the composition contains 4 μg estradiol or 10 μg estradiol.
  • 9. The method of claim 1, wherein the viscosity of the composition is from about 50 cP to about 380 cP at 25° C.
CROSS REFERENCE TO RELATED APPLICATIONS

This application claims priority to the following U.S. Patent Applications: U.S. Provisional Application Ser. No. 61/932,140, entitled “VAGINAL INSERTED ESTRADIOL PHARMACEUTICAL COMPOSITIONS AND METHODS”, which was filed on Jan. 27, 2014; U.S. Provisional Application Ser. No. 61/894,411, entitled “SOLUBLE ESTRADIOL CAPSULE FOR VAGINAL INSERTION,” which was filed on Oct. 22, 2013; U.S. Provisional Application Ser. No. 61/745,313, entitled “SOLUBLE ESTRADIOL CAPSULE FOR VAGINAL INSERTION,” which was filed on Dec. 21, 2012; U.S. Provisional Application Ser. No. 61/889,483, entitled “Natural Combination Hormone Replacement Pharmaceutical compositions and Therapies,” which was filed on Oct. 10, 2013; U.S. Provisional Application Ser. No. 61/661,302, entitled “ESTRADIOL PHARMACEUTICAL COMPOSITIONS,” which was filed on Jun. 18, 2012; U.S. Provisional Application Ser. No. 61/662,265, entitled “PROGESTERONE PHARMACEUTICAL COMPOSITIONS,” which was filed on Jun. 20, 2012; U.S. patent application Ser. No. 13/684,002, entitled “Natural Combination Hormone Replacement Pharmaceutical compositions and Therapies,” which was filed Nov. 21, 2012; U.S. Patent Application Serial No. PCT/US2013/023309, entitled “Transdermal Hormone Replacement Therapies,” which was filed Jan. 25, 2013; and U.S. patent application Ser. No. 13/843,362, entitled “Transdermal Hormone Replacement Therapies,” which was filed Mar. 15, 2013. All aforementioned applications are hereby incorporated by reference herein in their entirety.

US Referenced Citations (1252)
Number Name Date Kind
1967351 Doisy Jan 1934 A
2232438 Butenandt Feb 1941 A
2379832 Serini et al. Jul 1945 A
2649399 Beall et al. Aug 1953 A
3198707 Nomine et al. Aug 1965 A
3478070 Stein et al. Nov 1969 A
3526648 Bertin et al. Sep 1970 A
3710795 Higuchi et al. Jan 1973 A
3729560 Hagerman Apr 1973 A
3729566 Ericsson et al. Apr 1973 A
3755573 Berman Aug 1973 A
3755575 Lerner Aug 1973 A
3903880 Higuchi et al. Sep 1975 A
3916898 Robinson Nov 1975 A
3916899 Theeuwes et al. Nov 1975 A
3921636 Zaffaroni Nov 1975 A
3923997 Meuly Dec 1975 A
3948254 Zaffaroni Apr 1976 A
3971367 Zaffaroni Jun 1976 A
3977404 Theeuwes Aug 1976 A
3993072 Zaffaroni Nov 1976 A
4008719 Theeuwes et al. Feb 1977 A
4012496 Schopflin et al. Mar 1977 A
4014334 Theeuwes et al. Mar 1977 A
4014987 Heller et al. Mar 1977 A
4016251 Higuchi et al. Aug 1977 A
4071623 van der Vies Jan 1978 A
4093709 Choi et al. Jun 1978 A
4154820 Simoons May 1979 A
4155991 Schopflin et al. May 1979 A
4196188 Besins Apr 1980 A
4215691 Wong Aug 1980 A
4237885 Wong et al. Dec 1980 A
4310510 Sherman et al. Jan 1982 A
4327725 Cortese et al. May 1982 A
4372951 Vorys Feb 1983 A
4384096 Sonnabend May 1983 A
4393871 Vorhauer et al. Jul 1983 A
4402695 Wong Sep 1983 A
4423151 Baranczuk Dec 1983 A
4449980 Millar et al. May 1984 A
4610687 Fogwell Sep 1986 A
4629449 Wong Dec 1986 A
4732763 Beck et al. Mar 1988 A
4738957 Laurent et al. Apr 1988 A
4756907 Beck et al. Jul 1988 A
4762717 Crowley, Jr. Aug 1988 A
4788062 Gale et al. Nov 1988 A
4816257 Buster et al. Mar 1989 A
4822616 Zimmermann et al. Apr 1989 A
4865848 Cheng et al. Sep 1989 A
4900734 Maxson et al. Feb 1990 A
4906475 Kim Mar 1990 A
4942158 Sarpotdar et al. Jul 1990 A
4961931 Wong Oct 1990 A
5030629 Rajadhyaksha Jul 1991 A
5043331 Hirvonen et al. Aug 1991 A
5059426 Chiang Oct 1991 A
5064654 Berner et al. Nov 1991 A
5108995 Casper Apr 1992 A
5128138 Blank Jul 1992 A
5130137 Crowley, Jr. Jul 1992 A
5140021 Maxson et al. Aug 1992 A
5164416 Nagai et al. Nov 1992 A
5208225 Boissonneault et al. May 1993 A
5211952 Spicer et al. May 1993 A
5252334 Chiang et al. Oct 1993 A
5280023 Ehrlich et al. Jan 1994 A
5288496 Lewis Feb 1994 A
5295945 Miller Mar 1994 A
5340584 Spicer et al. Aug 1994 A
5340585 Pike et al. Aug 1994 A
5340586 Pike et al. Aug 1994 A
5362497 Yamada et al. Aug 1994 A
5382573 Casper Jan 1995 A
5393528 Staab Feb 1995 A
5393529 Hoffmann et al. Feb 1995 A
5419910 Lewis May 1995 A
5453279 Lee et al. Sep 1995 A
5468736 Hodgen Nov 1995 A
5474783 Miranda et al. Dec 1995 A
5480776 Dullien Jan 1996 A
5514673 Heckenmueller et al. May 1996 A
5516528 Hughes et al. May 1996 A
5527534 Myhling Jun 1996 A
5529782 Staab Jun 1996 A
5538736 Barth et al. Jul 1996 A
5543150 Bologna et al. Aug 1996 A
5547948 Barcomb Aug 1996 A
5556635 Grognet et al. Sep 1996 A
5565199 Page et al. Oct 1996 A
5567831 Li Oct 1996 A
5569652 Beier et al. Oct 1996 A
5580572 Liorzou et al. Dec 1996 A
5582592 Kendrick Dec 1996 A
5585370 Casper Dec 1996 A
5595759 Wright et al. Jan 1997 A
5595970 Garfield et al. Jan 1997 A
5605702 Math et al. Feb 1997 A
5607691 Solas et al. Mar 1997 A
5607693 Bonte et al. Mar 1997 A
5609617 Cady et al. Mar 1997 A
5620705 Dong et al. Apr 1997 A
5626866 Heiber et al. May 1997 A
5629021 Wright May 1997 A
5633011 Dong et al. May 1997 A
5633242 Oettel et al. May 1997 A
5639743 Kaswan et al. Jun 1997 A
5645856 Lacy et al. Jun 1997 A
5653983 Bonte et al. Aug 1997 A
5656286 Miranda et al. Aug 1997 A
5660839 Allec et al. Aug 1997 A
5662927 Ehrlich et al. Sep 1997 A
5663160 Dumas et al. Sep 1997 A
5676968 Lipp et al. Oct 1997 A
5677292 Li et al. Oct 1997 A
5686097 Crisologo et al. Nov 1997 A
5693335 Xia et al. Dec 1997 A
5694947 Lehtinen et al. Dec 1997 A
5700480 Hille et al. Dec 1997 A
5709844 Arbeit et al. Jan 1998 A
5719197 Mantelle et al. Feb 1998 A
5735801 Caillouette Apr 1998 A
5739176 Dunn et al. Apr 1998 A
5744463 Bair Apr 1998 A
5747058 Tipton et al. May 1998 A
5762614 Caillouette Jun 1998 A
5770176 Nargessi Jun 1998 A
5770219 Chiang et al. Jun 1998 A
5770220 Meconi et al. Jun 1998 A
5770227 Dong et al. Jun 1998 A
5776495 Duclos et al. Jul 1998 A
5780044 Tipton et al. Jul 1998 A
5780050 Jain et al. Jul 1998 A
5788980 Nabahi Aug 1998 A
5788984 Schmidt et al. Aug 1998 A
5789442 Garfield et al. Aug 1998 A
5811416 Chwalisz et al. Sep 1998 A
5811547 Nakamichi et al. Sep 1998 A
5814329 Shah Sep 1998 A
5820878 Shinmura et al. Oct 1998 A
5827200 Caillouette Oct 1998 A
5840327 Gale et al. Nov 1998 A
5843468 Yum et al. Dec 1998 A
5843979 Wille et al. Dec 1998 A
5858394 Lipp et al. Jan 1999 A
5863552 Yue Jan 1999 A
5866603 Li et al. Feb 1999 A
5869084 Paradissis et al. Feb 1999 A
5882676 Yum et al. Mar 1999 A
5885612 Meconi et al. Mar 1999 A
5888533 Dunn Mar 1999 A
5891462 Carrara Apr 1999 A
5891868 Cummings et al. Apr 1999 A
5898038 Yallampalli et al. Apr 1999 A
5902603 Chen et al. May 1999 A
5904931 Gunther et al. May 1999 A
5906830 Farinas et al. May 1999 A
5912010 Wille et al. Jun 1999 A
5916176 Caillouette Jun 1999 A
RE36247 Plunkett et al. Jul 1999 E
5919477 Bevan et al. Jul 1999 A
5922349 Elliesen et al. Jul 1999 A
5928666 Farinas et al. Jul 1999 A
5942243 Shah Aug 1999 A
5942531 Diaz et al. Aug 1999 A
5952000 Fikstad et al. Sep 1999 A
5958446 Miranda et al. Sep 1999 A
5962445 Stewart Oct 1999 A
5968919 Gyurik et al. Oct 1999 A
5972372 Saleh et al. Oct 1999 A
5985311 Cordes et al. Nov 1999 A
5985850 Falk et al. Nov 1999 A
5985861 Levine et al. Nov 1999 A
5993856 Ragavan et al. Nov 1999 A
5989568 De Lacharriere et al. Dec 1999 A
6001846 Edwards et al. Dec 1999 A
6007835 Bon-Lapillonne et al. Dec 1999 A
6010715 Pollock et al. Jan 2000 A
6013276 Teillaud et al. Jan 2000 A
6022562 Autant et al. Feb 2000 A
6024974 Li Feb 2000 A
6024976 Miranda et al. Feb 2000 A
6028057 Burns Feb 2000 A
6030948 Mann Feb 2000 A
6039968 Nabahi Mar 2000 A
6040340 Garfield et al. Mar 2000 A
6056972 Hermsmeyer May 2000 A
6060077 Meignant May 2000 A
6068853 Berner et al. May 2000 A
6074625 Hawthorne et al. Jun 2000 A
6077531 Salin-Drouin Jun 2000 A
6080118 Blythe Jun 2000 A
6083178 Caillouette Jul 2000 A
6086916 Agnus et al. Jul 2000 A
6087352 Trout Jul 2000 A
6090404 Meconi et al. Jul 2000 A
6096338 Lacy et al. Jul 2000 A
6106848 Willcox et al. Aug 2000 A
6117446 Place Sep 2000 A
6117450 Dittgen et al. Sep 2000 A
6124362 Bradbury et al. Sep 2000 A
6133251 Dittgen et al. Oct 2000 A
6133320 Yallampalli et al. Oct 2000 A
6139868 Hoffmann Oct 2000 A
6139873 Hughes, Jr. et al. Oct 2000 A
6149935 Tenzel et al. Nov 2000 A
6153216 Cordes et al. Nov 2000 A
6165491 Grasset et al. Dec 2000 A
6165975 Adams et al. Dec 2000 A
6187323 Aiache et al. Feb 2001 B1
6187339 de Haan et al. Feb 2001 B1
6190331 Caillouette Feb 2001 B1
6201072 Rathi et al. Mar 2001 B1
6217886 Rubinstein et al. Apr 2001 B1
6225297 Stockemann et al. May 2001 B1
6227202 Matapurkar May 2001 B1
6228383 Hansen et al. May 2001 B1
6228852 Shaak May 2001 B1
6242509 MacQueen et al. Jun 2001 B1
6245811 Horrobin et al. Jun 2001 B1
6262115 Guittard et al. Jul 2001 B1
6267984 Hamlin et al. Jul 2001 B1
6274165 Meconi et al. Aug 2001 B1
6277418 Marakverich et al. Aug 2001 B1
6283927 Caillouette Sep 2001 B1
6284263 Place Sep 2001 B1
6287588 Shih et al. Sep 2001 B1
6287693 Savoir et al. Sep 2001 B1
6294188 Ragavan et al. Sep 2001 B1
6294192 Patel et al. Sep 2001 B1
6294550 Place et al. Sep 2001 B1
6299900 Reed et al. Oct 2001 B1
6303132 Nelson Oct 2001 B1
6303588 Danielov Oct 2001 B1
6306841 Place et al. Oct 2001 B1
6306914 de Ziegler et al. Oct 2001 B1
6309669 Setterstrom et al. Oct 2001 B1
6309848 Howett et al. Oct 2001 B1
6312703 Orthoefer Nov 2001 B1
6328987 Marini Dec 2001 B1
6342491 Dey et al. Jan 2002 B1
6344211 Hille Feb 2002 B1
6372209 Chrisope Apr 2002 B1
6372245 Vo et al. Apr 2002 B1
6372246 Wei et al. Apr 2002 B1
6387390 Deaver et al. May 2002 B1
6402705 Caillouette Jun 2002 B1
6416778 Ragavan et al. Jul 2002 B1
6420352 Knowles Jul 2002 B1
6423039 Rathbone et al. Jul 2002 B1
6423683 Heaton et al. Jul 2002 B1
6432438 Shukla Aug 2002 B1
6436633 Kreider et al. Aug 2002 B1
6440454 Santoro et al. Aug 2002 B1
6444224 Rathbone et al. Sep 2002 B1
6444234 Kirby et al. Sep 2002 B1
6451300 Leyba et al. Sep 2002 B1
6451339 Patel et al. Sep 2002 B2
6451779 Hesch Sep 2002 B1
6455246 Howett et al. Sep 2002 B1
6455517 Tanabe et al. Sep 2002 B1
6465004 Houze et al. Oct 2002 B1
6465005 Biali et al. Oct 2002 B1
6465006 Zhang et al. Oct 2002 B1
6468526 Chrisope Oct 2002 B2
6469016 Place et al. Oct 2002 B1
6472434 Place et al. Oct 2002 B1
6479232 Howett et al. Nov 2002 B1
6495160 Esposito et al. Dec 2002 B2
6500814 Hesch Dec 2002 B1
6503896 Tanabe et al. Jan 2003 B1
6511969 Hermsmeyer Jan 2003 B1
6521250 Seibertz et al. Feb 2003 B2
6526980 Tracy et al. Mar 2003 B1
6528094 Savoir et al. Mar 2003 B1
6531149 Meconi et al. Mar 2003 B1
6537580 Savoir et al. Mar 2003 B1
6538039 Laurent Mar 2003 B2
6544196 Caillouette Apr 2003 B2
6544553 Hsia et al. Apr 2003 B1
6548053 Murray et al. Apr 2003 B1
6548491 Tanabe et al. Apr 2003 B2
6551611 Sen et al. Apr 2003 B2
6555131 Wolff et al. Apr 2003 B1
6562367 Wolff et al. May 2003 B1
6562370 Luo et al. May 2003 B2
6562790 Chein May 2003 B2
6569463 Patel et al. May 2003 B2
6583129 Mazer et al. Jun 2003 B1
6586006 Roser et al. Jul 2003 B2
6589549 Shih et al. Jul 2003 B2
6593317 de Ziegler et al. Jul 2003 B1
6599519 Seo et al. Jul 2003 B1
6610325 Meignant Aug 2003 B1
6610652 Adams et al. Aug 2003 B2
6610670 Backensfeld et al. Aug 2003 B2
6610674 Schreiber Aug 2003 B1
6635274 Carter et al. Oct 2003 B1
6638528 Kanios Oct 2003 B1
6638536 Savoir et al. Oct 2003 B2
6645528 Straub et al. Nov 2003 B1
6649155 Dunlop et al. Nov 2003 B1
6653298 Potter et al. Nov 2003 B2
6656929 Agnus et al. Dec 2003 B1
6660726 Hill et al. Dec 2003 B2
6663608 Rathbone et al. Dec 2003 B2
6663895 Savoir et al. Dec 2003 B2
6664296 Meignant Dec 2003 B1
6682757 Wright Jan 2004 B1
6692763 Cummings et al. Feb 2004 B1
6708822 Muni Mar 2004 B1
6716454 Meignant Apr 2004 B2
6720001 Chen et al. Apr 2004 B2
6737081 Avoir et al. May 2004 B2
6740333 Beckett et al. May 2004 B2
6743448 Kryger Jun 2004 B2
6743815 Huebner et al. Jun 2004 B2
6747018 Tanabe et al. Jun 2004 B2
6750291 Kim et al. Jun 2004 B2
6756208 Griffin et al. Jun 2004 B2
6776164 Bunt et al. Aug 2004 B2
6787152 Kirby et al. Sep 2004 B2
6805877 Massara et al. Oct 2004 B2
6809085 Elson et al. Oct 2004 B1
6818226 Reed et al. Nov 2004 B2
6821524 Marini Nov 2004 B2
6841716 Tsutsumi Jan 2005 B1
6844334 Hill et al. Jan 2005 B2
6855703 Hill et al. Feb 2005 B1
6860859 Mehrotra et al. Mar 2005 B2
6866865 Hsia et al. Mar 2005 B2
6869969 Heubner et al. Mar 2005 B2
6878518 Whitehead Apr 2005 B2
6901278 Notelovitz May 2005 B1
6905705 Palm et al. Jun 2005 B2
6911211 Tamarkin et al. Jun 2005 B2
6911438 Wright Jun 2005 B2
6923988 Patel et al. Aug 2005 B2
6924274 Lardy et al. Aug 2005 B2
6932983 Straub et al. Aug 2005 B1
6939558 Massara et al. Sep 2005 B2
6943021 Klausner et al. Sep 2005 B2
6958327 Hillisch et al. Oct 2005 B1
6960337 Pike et al. Nov 2005 B2
6962691 Lulla et al. Nov 2005 B1
6962908 Aloba et al. Nov 2005 B2
6967194 Matsuo et al. Nov 2005 B1
6974569 Boyd et al. Dec 2005 B2
6977250 Rodriguez Dec 2005 B2
6978945 Wong et al. Dec 2005 B2
6987129 Mak et al. Jan 2006 B2
6995149 Reilhac et al. Feb 2006 B1
7004321 Hackbarth et al. Feb 2006 B1
7005429 Dey et al. Feb 2006 B2
7011846 Shojaei et al. Mar 2006 B2
7018992 Koch et al. Mar 2006 B2
7030104 Paris et al. Apr 2006 B2
7030157 Ke et al. Apr 2006 B2
RE39104 Duclos et al. May 2006 E
7074779 Sui et al. Jul 2006 B2
7083590 Bunt et al. Aug 2006 B1
7091213 Metcalf, III et al. Aug 2006 B2
7094228 Zhang et al. Aug 2006 B2
7097853 Keister et al. Aug 2006 B1
7101342 Caillouette Sep 2006 B1
7105573 Krajcik et al. Sep 2006 B2
7135190 Piao et al. Nov 2006 B2
7153522 Ikeura et al. Dec 2006 B1
7163681 Giles-Komar et al. Jan 2007 B2
7163699 Besse Jan 2007 B2
7175850 Cevc Feb 2007 B2
7179799 Hill et al. Feb 2007 B2
7196074 Blye et al. Mar 2007 B2
7198800 Ko Apr 2007 B1
7198801 Carrara et al. Apr 2007 B2
7226910 Wilson et al. Jun 2007 B2
7247625 Zhang et al. Jul 2007 B2
7250446 Sangita et al. Jul 2007 B2
7267829 Kirby et al. Sep 2007 B2
7300926 Prokai et al. Nov 2007 B2
7303763 Ho Dec 2007 B2
7317037 Fensome et al. Jan 2008 B2
7329654 Kanojia et al. Feb 2008 B2
7335650 Potter et al. Feb 2008 B2
7374779 Chen et al. May 2008 B2
7378404 Peters et al. May 2008 B2
7381427 Ancira et al. Jun 2008 B2
7387789 Klose et al. Jun 2008 B2
7388006 Schmees et al. Jun 2008 B2
7414043 Kosemund et al. Aug 2008 B2
7427413 Savoir et al. Sep 2008 B2
7427609 Leonard Sep 2008 B2
7429576 Labrie Sep 2008 B2
7431941 Besins et al. Oct 2008 B2
7456159 Houze et al. Nov 2008 B2
7459445 Hill et al. Dec 2008 B2
7465587 Imrich Dec 2008 B2
7470433 Carrara et al. Dec 2008 B2
7485666 Villaneuva et al. Feb 2009 B2
7497855 Ausiello et al. Mar 2009 B2
7498303 Arnold et al. Mar 2009 B2
7534765 Gregg et al. May 2009 B2
7534780 Ring et al. May 2009 B2
7550142 Giles-Komar et al. Jun 2009 B2
7563565 Matsuo et al. Jul 2009 B1
7569274 Alphonse et al. Aug 2009 B2
7572779 Aloba et al. Aug 2009 B2
7572780 Hermsmeyer Aug 2009 B2
7589082 Savoir et al. Sep 2009 B2
7671027 Loumaye Mar 2010 B2
7674783 Hermsmeyer Mar 2010 B2
7687281 Roth et al. Mar 2010 B2
7687485 Levinson et al. Mar 2010 B2
7694683 Callister et al. Apr 2010 B2
7704983 Hodgen et al. Apr 2010 B1
7727720 Dhallan Jun 2010 B2
7732408 Josephson et al. Jun 2010 B2
7749989 Hill et al. Jul 2010 B2
7767656 Shoichet et al. Aug 2010 B2
7799769 White et al. Sep 2010 B2
7815936 Hasenzahl et al. Oct 2010 B2
7815949 Cohen Oct 2010 B2
7829115 Besins et al. Nov 2010 B2
7829116 Frye et al. Nov 2010 B2
RE42012 Deaver et al. Dec 2010 E
7850992 Hwang, II et al. Dec 2010 B2
7854753 Kraft et al. Dec 2010 B2
7858607 Mamchur Dec 2010 B2
RE42072 Deaver et al. Jan 2011 E
7862552 McIntyre et al. Jan 2011 B2
7867990 Schultz et al. Jan 2011 B2
7871643 Lizio et al. Jan 2011 B2
7879830 Wiley Feb 2011 B2
7884093 Creasy et al. Feb 2011 B2
7925519 Greene Apr 2011 B2
7939104 Barbera et al. May 2011 B2
7943602 Bunschoten et al. May 2011 B2
7943604 Coelingh Bennink et al. May 2011 B2
7945459 Grace et al. May 2011 B2
7960368 Rao et al. Jun 2011 B2
7989436 Hill et al. Aug 2011 B2
7989487 Welsh et al. Aug 2011 B2
8022053 Mueller et al. Sep 2011 B2
8048017 Xu Nov 2011 B2
8048869 Bunschoten et al. Nov 2011 B2
8063030 Ellman Nov 2011 B2
8071576 Visser et al. Dec 2011 B2
8071729 Giles-Komar et al. Dec 2011 B2
8075916 Park et al. Dec 2011 B2
8075917 Park et al. Dec 2011 B2
8076317 Kulmann Dec 2011 B2
8076319 Leonard Dec 2011 B2
8080553 Auspitz et al. Dec 2011 B2
8088605 Beudet et al. Jan 2012 B2
8096940 Iverson et al. Jan 2012 B2
8101209 Legrand et al. Jan 2012 B2
8101773 Smith et al. Jan 2012 B2
8114152 Furst Feb 2012 B2
8114434 Sasaki et al. Feb 2012 B2
8114442 Tucker et al. Feb 2012 B2
8119741 Pavlin Feb 2012 B2
8121886 Azar Feb 2012 B2
8124118 Lennernaes et al. Feb 2012 B2
8124595 Boissonneault Feb 2012 B2
8147561 Binmoeller Apr 2012 B2
8148546 Baasner et al. Apr 2012 B2
8158613 Staniforth et al. Apr 2012 B2
8158614 Lambert et al. Apr 2012 B2
8163722 Savoir et al. Apr 2012 B2
8177449 Watkinson et al. May 2012 B2
8182833 Hermsmeyer May 2012 B2
8187615 Friedman May 2012 B2
8195403 Wood, Jr. et al. Jun 2012 B2
8202736 Mousa et al. Jun 2012 B2
8217024 Ahmed et al. Jul 2012 B2
8221785 Chien Jul 2012 B2
8222008 Thoene Jul 2012 B2
8222237 Narkunan et al. Jul 2012 B2
8227454 Hill et al. Jul 2012 B2
8227509 Castro et al. Jul 2012 B2
8241664 Dudley et al. Aug 2012 B2
8247393 Ahmed et al. Aug 2012 B2
8257724 Cromack et al. Sep 2012 B2
8257725 Cromack et al. Sep 2012 B2
8268352 Karan et al. Sep 2012 B2
8268806 Labrie Sep 2012 B2
8268878 Johnson et al. Sep 2012 B2
8273730 Fernandez et al. Sep 2012 B2
8287888 Song et al. Oct 2012 B2
8288366 Gonzalez et al. Oct 2012 B2
8318898 Fasel et al. Nov 2012 B2
8324193 Lee Sepsick et al. Dec 2012 B2
8329680 Evans et al. Dec 2012 B2
8337814 Osbakken et al. Dec 2012 B2
8344007 Chui et al. Jan 2013 B2
8349820 Zeun et al. Jan 2013 B2
8353863 Imran Jan 2013 B2
8357723 Satyam Jan 2013 B2
8361995 Schramm Jan 2013 B2
8362091 Besonov et al. Jan 2013 B2
8372424 Berry et al. Feb 2013 B2
8372806 Bragagna et al. Feb 2013 B2
8377482 Laurie et al. Feb 2013 B2
8377994 Drechsler et al. Feb 2013 B2
8394759 Barathur et al. Mar 2013 B2
8415332 Reape et al. Apr 2013 B2
8420111 Hermsmeyer Apr 2013 B2
8435561 Besins et al. May 2013 B2
8435972 Sayeed et al. May 2013 B2
8449879 Laurent Applegate et al. May 2013 B2
8450108 Boyce May 2013 B2
8454945 Narain et al. Jun 2013 B2
8455468 Kellermann et al. Jun 2013 B2
8461138 Boissonneault Jun 2013 B2
8476252 Pickersgill et al. Jul 2013 B2
8481488 Carter Jul 2013 B2
8486374 Zlatkis et al. Jul 2013 B2
8486442 Yamaji et al. Jul 2013 B2
8492368 Lewandowski et al. Jul 2013 B2
8507467 Ueda et al. Aug 2013 B2
8512693 Azevedo et al. Aug 2013 B2
8512754 Needham Aug 2013 B2
8518376 Schuz et al. Aug 2013 B2
8536159 Zeng et al. Sep 2013 B2
8540967 Trivedi et al. Sep 2013 B2
8541400 Joabsson et al. Sep 2013 B2
8551462 Marenus et al. Oct 2013 B2
8551508 Lee et al. Oct 2013 B2
8557281 Tuominen et al. Oct 2013 B2
8568374 De Graaff et al. Oct 2013 B2
8591951 Kohn et al. Nov 2013 B2
8613951 Troiano et al. Dec 2013 B2
8633178 Cacace et al. Jan 2014 B2
8633180 Zeng et al. Jan 2014 B2
8636787 Sabaria Jan 2014 B2
8636982 Schuz et al. Jan 2014 B2
8653129 Fein et al. Feb 2014 B2
8658627 Voskuhl Feb 2014 B2
8658628 Baucom Feb 2014 B2
8663681 Ahmed et al. Mar 2014 B2
8663692 Mueller et al. Mar 2014 B1
8663703 Moldavski et al. Mar 2014 B2
8664207 Zheng et al. Mar 2014 B2
8669293 Sharoni et al. Mar 2014 B2
8679552 Guthery Mar 2014 B2
8694358 Tryfon Apr 2014 B2
8697127 Sah Apr 2014 B2
8697710 Zeng et al. Apr 2014 B2
8703105 Besonov et al. Apr 2014 B2
8709385 Schuz et al. Apr 2014 B2
8709451 Rapoport et al. Apr 2014 B2
8715735 Funke et al. May 2014 B2
8721331 Raghuprasad May 2014 B2
8722021 Eini et al. May 2014 B2
8734846 Hrkach et al. May 2014 B2
8735381 Podolski May 2014 B2
8741336 Dipierro et al. Jun 2014 B2
8741373 Rao et al. Jun 2014 B2
8753661 Gassner et al. Jun 2014 B2
8784882 Mattern Jul 2014 B2
8846648 Bernick et al. Sep 2014 B2
8846649 Bernick et al. Sep 2014 B2
8933059 Bernick et al. Jan 2015 B2
8987237 Bernick et al. Mar 2015 B2
8987238 Bernick et al. Mar 2015 B2
8993548 Bernick et al. Mar 2015 B2
8993549 Bernick et al. Mar 2015 B2
9005597 Hansen et al. Apr 2015 B2
9006222 Bernick et al. Apr 2015 B2
9012434 Bernick et al. Apr 2015 B2
9114145 Bernick et al. Aug 2015 B2
9114146 Bernick et al. Aug 2015 B2
9180091 Bernick et al. Nov 2015 B2
9248136 Bernick et al. Feb 2016 B2
9289382 Bernick et al. Mar 2016 B2
9301920 Bernick et al. Apr 2016 B2
9931349 Shadiack et al. Apr 2018 B2
10052386 Bernick et al. Aug 2018 B2
10098894 Amadio et al. Oct 2018 B2
10206932 Bernick et al. Feb 2019 B2
10258630 Mirkin et al. Apr 2019 B2
10398708 Mirkin et al. Sep 2019 B2
10471072 Bernick et al. Nov 2019 B2
10568891 Mirkin et al. Feb 2020 B2
10835487 Bernick et al. Nov 2020 B2
10888516 Bernick et al. Jan 2021 B2
11065197 Bernick et al. Jul 2021 B2
20010005728 Guittard et al. Feb 2001 A1
20010009673 Gunther et al. Jul 2001 A1
20010021816 Caillouette Sep 2001 A1
20010023261 Ryoo et al. Sep 2001 A1
20010027189 Bennink et al. Oct 2001 A1
20010029357 Bunt et al. Oct 2001 A1
20010031747 de Ziegler et al. Oct 2001 A1
20010032125 Bhan et al. Oct 2001 A1
20010034340 Pickar Oct 2001 A1
20120269878 Cantor et al. Oct 2001 A2
20010053383 Sablotsky et al. Dec 2001 A1
20010056068 Chwalisz et al. Dec 2001 A1
20020012710 Lansky Jan 2002 A1
20020026158 Rathbone et al. Feb 2002 A1
20020028788 Bunt et al. Mar 2002 A1
20020035070 Gardlik et al. Mar 2002 A1
20020058648 Hammerly May 2002 A1
20020058926 Rathbone et al. May 2002 A1
20020064541 Lapidot et al. May 2002 A1
20020076441 Shih et al. Jun 2002 A1
20020102308 Wei et al. Aug 2002 A1
20020107230 Waldon et al. Aug 2002 A1
20020114803 Deaver et al. Aug 2002 A1
20020119174 Gardlik et al. Aug 2002 A1
20020119198 Gao et al. Aug 2002 A1
20020132801 Heil et al. Sep 2002 A1
20020137749 Levinson et al. Sep 2002 A1
20020142017 Simonnet Oct 2002 A1
20020151530 Leonard et al. Oct 2002 A1
20020156394 Mehrotra et al. Oct 2002 A1
20020169150 Pickar Nov 2002 A1
20020169205 Garfield et al. Nov 2002 A1
20020173510 Levinson et al. Nov 2002 A1
20020193356 Van Beek et al. Dec 2002 A1
20020193758 Sandberg Dec 2002 A1
20020197286 Brandman et al. Dec 2002 A1
20030003139 Gunther et al. Jan 2003 A1
20030004145 Leonard Jan 2003 A1
20030007994 Bunt et al. Jan 2003 A1
20030027772 Breton Feb 2003 A1
20030091620 Venkateshwaran et al. Feb 2003 A1
20030044453 Volkel et al. Mar 2003 A1
20030049307 Gyurik Mar 2003 A1
20030052799 Weigl Mar 2003 A1
20030064097 Patel et al. Apr 2003 A1
20030064975 Koch et al. Apr 2003 A1
20030072760 Sirbasku Apr 2003 A1
20030073248 Roth et al. Apr 2003 A1
20030073673 Hesch Apr 2003 A1
20030077297 Chen et al. Apr 2003 A1
20030078245 Bennink et al. Apr 2003 A1
20030091640 Ramanathan et al. May 2003 A1
20030092691 Besse et al. May 2003 A1
20030096012 Besse et al. May 2003 A1
20030104048 Patel et al. Jun 2003 A1
20030109507 Beckmann et al. Jun 2003 A1
20030113268 Buenafae et al. Jun 2003 A1
20030114420 Salvati et al. Jun 2003 A1
20030114430 MacLeod et al. Jun 2003 A1
20030124182 Shojaei et al. Jul 2003 A1
20030124191 Besse et al. Jul 2003 A1
20030130558 Massara et al. Jul 2003 A1
20030144258 Heil et al. Jul 2003 A1
20030157157 Luo et al. Aug 2003 A1
20030166509 Batycky et al. Sep 2003 A1
20030170295 Yoon et al. Sep 2003 A1
20030175329 Mak et al. Sep 2003 A1
20030175333 Shefer et al. Sep 2003 A1
20030180352 Patel et al. Sep 2003 A1
20030181353 Nyce Sep 2003 A1
20030181728 Salvati et al. Sep 2003 A1
20030191096 Leonard et al. Oct 2003 A1
20030195177 Leonard et al. Oct 2003 A1
20030215496 Patel et al. Nov 2003 A1
20030219402 Rutter Nov 2003 A1
20030220297 Bernstein et al. Nov 2003 A1
20030224057 Martin-Letellier et al. Dec 2003 A1
20030224059 Lerner et al. Dec 2003 A1
20030225047 Friedman et al. Dec 2003 A1
20030225048 Friedman et al. Dec 2003 A1
20030225050 Eichardt et al. Dec 2003 A1
20030228686 Klausner et al. Dec 2003 A1
20030229057 Caubel et al. Dec 2003 A1
20030235596 Gao et al. Dec 2003 A1
20030236236 Chen et al. Dec 2003 A1
20040009960 Heil et al. Jan 2004 A1
20040022820 Anderson Feb 2004 A1
20040034001 Karara Feb 2004 A1
20040037881 Guittard et al. Feb 2004 A1
20040039356 Maki et al. Feb 2004 A1
20040043043 Schlyter et al. Mar 2004 A1
20040043943 Guittard et al. Mar 2004 A1
20040044080 Place et al. Mar 2004 A1
20040048900 Flood Mar 2004 A1
20040052824 Abou Chacra-Vernet et al. Mar 2004 A1
20040073024 Metcalf, III et al. Apr 2004 A1
20040077605 Salvati et al. Apr 2004 A1
20040077606 Salvati et al. Apr 2004 A1
20040087548 Salvati et al. May 2004 A1
20040087564 Wright et al. May 2004 A1
20040089308 Welch May 2004 A1
20040092494 Dudley May 2004 A9
20040092583 Shanahan-Prendergast May 2004 A1
20040093261 Jain et al. May 2004 A1
20040097468 Wimalawansa May 2004 A1
20040101557 Gibson et al. May 2004 A1
20040106542 Deaver et al. Jun 2004 A1
20040110732 Masini Eteve et al. Jun 2004 A1
20040131670 Gao Jul 2004 A1
20040138103 Patt Jul 2004 A1
20040142012 Bunt et al. Jul 2004 A1
20040146539 Gupta Jul 2004 A1
20040146894 Warrington et al. Jul 2004 A1
20040147578 Calvet Jul 2004 A1
20040161435 Gupta Aug 2004 A1
20040176324 Salvati et al. Sep 2004 A1
20040176336 Rodriguez Sep 2004 A1
20040185104 Piao et al. Sep 2004 A1
20040191207 Lipari et al. Sep 2004 A1
20040191276 Muni Sep 2004 A1
20040198706 Carrara et al. Oct 2004 A1
20040210280 Liedtke Oct 2004 A1
20040213744 Lulla et al. Oct 2004 A1
20040219124 Gupta Nov 2004 A1
20040225140 Sciano et al. Nov 2004 A1
20040234606 Levine et al. Nov 2004 A1
20040241219 Hille et al. Dec 2004 A1
20040243437 Grace et al. Dec 2004 A1
20040253319 Netke et al. Dec 2004 A1
20040259817 Waldon et al. Dec 2004 A1
20040266745 Schwanitz et al. Dec 2004 A1
20050003003 Deaver et al. Jan 2005 A1
20050004088 Hesch Jan 2005 A1
20050009800 Thumbeck et al. Jan 2005 A1
20050014729 Pulaski Jan 2005 A1
20050020550 Latif et al. Jan 2005 A1
20050020552 Aschkenasay et al. Jan 2005 A1
20050021009 Massara et al. Jan 2005 A1
20050025833 Aschkenasay et al. Feb 2005 A1
20050031651 Gervais et al. Feb 2005 A1
20050042173 Besse et al. Feb 2005 A1
20050042268 Aschkenasay et al. Feb 2005 A1
20050048116 Straub et al. Mar 2005 A1
20050054991 Paterson et al. Mar 2005 A1
20050079138 Chickering et al. Apr 2005 A1
20050085453 Govindarajan Apr 2005 A1
20050101579 Shippen May 2005 A1
20050113350 Duesterberg et al. May 2005 A1
20050118244 Theobald et al. Jun 2005 A1
20050118272 Besse et al. Jun 2005 A1
20050129756 Podhaisky et al. Jun 2005 A1
20050152956 Dudley Jul 2005 A1
20050153946 Hirsh et al. Jul 2005 A1
20050164977 Coelingh Bennink Jul 2005 A1
20050182105 Nirschl et al. Aug 2005 A1
20050186141 Gonda et al. Aug 2005 A1
20050187267 Hamann et al. Aug 2005 A1
20050192253 Salvati et al. Sep 2005 A1
20050192310 Gavai et al. Sep 2005 A1
20050196434 Brierre Sep 2005 A1
20050207990 Funke et al. Sep 2005 A1
20050209209 Koch et al. Sep 2005 A1
20050214384 Juturu et al. Sep 2005 A1
20050220825 Funke et al. Oct 2005 A1
20050220900 Wuttke et al. Oct 2005 A1
20050222106 Bracht Oct 2005 A1
20050228692 Hodgdon Oct 2005 A1
20050228718 Austin Oct 2005 A1
20050239747 Le et al. Oct 2005 A1
20050239758 Roby Oct 2005 A1
20050244360 Billoni Nov 2005 A1
20050244522 Carrara et al. Nov 2005 A1
20050245902 Cornish et al. Nov 2005 A1
20050250746 Iammatteo Nov 2005 A1
20050250750 Cummings et al. Nov 2005 A1
20050250753 Fink et al. Nov 2005 A1
20050256028 Yun et al. Nov 2005 A1
20050266078 Jorda et al. Nov 2005 A1
20050266088 Frijlink et al. Dec 2005 A1
20050271597 Keith Dec 2005 A1
20050271598 Friedman et al. Dec 2005 A1
20050272685 Hung Dec 2005 A1
20050272712 Grubb et al. Dec 2005 A1
20060009428 Grubb et al. Jan 2006 A1
20060014728 Chwalisz et al. Jan 2006 A1
20060018937 Friedman et al. Jan 2006 A1
20060019978 Balog Jan 2006 A1
20060020002 Salvati et al. Jan 2006 A1
20060030615 Fensome et al. Feb 2006 A1
20060034889 Jo et al. Feb 2006 A1
20060034904 Weimann Feb 2006 A1
20060040904 Ahmed et al. Feb 2006 A1
20060051391 Dvoskin et al. Mar 2006 A1
20060052341 Cornish et al. Mar 2006 A1
20060069031 Loumaye Mar 2006 A1
20060078618 Constantinides et al. Apr 2006 A1
20060083778 Allison et al. Apr 2006 A1
20060084704 Shih et al. Apr 2006 A1
20060088580 Seibertz et al. Apr 2006 A1
20060089337 Casper et al. Apr 2006 A1
20060093678 Chickering, III et al. May 2006 A1
20060100180 Bohlmann et al. May 2006 A1
20060106004 Brody et al. May 2006 A1
20060110415 Gupta May 2006 A1
20060111424 Salvati et al. May 2006 A1
20060121102 Chiang Jun 2006 A1
20060121626 Imrich Jun 2006 A1
20060134188 Podhaisky et al. Jun 2006 A1
20060135619 Kick et al. Jun 2006 A1
20060165744 Anyarambhatla et al. Jul 2006 A1
20060193789 Tamarkin et al. Aug 2006 A1
20060194775 Tofovic et al. Aug 2006 A1
20060204557 Gupta et al. Sep 2006 A1
20060233743 Kelly Oct 2006 A1
20060233841 Pushpala et al. Oct 2006 A1
20060235037 Purandare et al. Oct 2006 A1
20060240111 Fernandez et al. Oct 2006 A1
20060246122 Langguth et al. Nov 2006 A1
20060247216 Haj-Yehia Nov 2006 A1
20060247221 Coelingh Bennink et al. Nov 2006 A1
20060251581 Madenjian et al. Nov 2006 A1
20060252049 Shuler et al. Nov 2006 A1
20060257472 Neilsen Nov 2006 A1
20060275218 Besonov et al. Dec 2006 A1
20060275360 Ahmed et al. Dec 2006 A1
20060276414 Coelingh Bennink et al. Dec 2006 A1
20060280771 Groenewegen et al. Dec 2006 A1
20060280797 Shoichet et al. Dec 2006 A1
20060280800 Nagi et al. Dec 2006 A1
20060292223 Mc Ilroy et al. Dec 2006 A1
20070004693 Woolfson et al. Jan 2007 A1
20070004694 Woolfson et al. Jan 2007 A1
20070009559 Alosio et al. Jan 2007 A1
20070009594 Grubb et al. Jan 2007 A1
20070010550 McKenzie Jan 2007 A1
20070014839 Bracht Jan 2007 A1
20070015698 Goldstein et al. Jan 2007 A1
20070021360 Nyce et al. Jan 2007 A1
20070027201 McComas et al. Feb 2007 A1
20070031491 Levine et al. Feb 2007 A1
20070036843 Hirsh et al. Feb 2007 A1
20070037780 Anigbogu et al. Feb 2007 A1
20070037782 Suzuki et al. Feb 2007 A1
20070042038 Besse Feb 2007 A1
20070049567 Wiley Mar 2007 A1
20070060589 Purandare et al. Mar 2007 A1
20070066628 Zhang et al. Mar 2007 A1
20070066637 Zhang et al. Mar 2007 A1
20070066675 Zhang et al. Mar 2007 A1
20070071777 Bromer et al. Mar 2007 A1
20070078091 Hubler et al. Apr 2007 A1
20070088029 Balog et al. Apr 2007 A1
20070093548 Diffendal et al. Apr 2007 A1
20070116729 Palepu May 2007 A1
20070116829 Prakash et al. May 2007 A1
20070128263 Wall et al. Jun 2007 A1
20070154533 Dudley Jul 2007 A1
20070167418 Ferguson Jul 2007 A1
20070178166 Bernstein et al. Aug 2007 A1
20070184558 Roth et al. Aug 2007 A1
20070185068 Ferguson et al. Aug 2007 A1
20070190022 Chiao et al. Aug 2007 A1
20070191319 Ke et al. Aug 2007 A1
20070191321 Ahmed Aug 2007 A1
20070196415 Houston et al. Aug 2007 A1
20070196433 Ron et al. Aug 2007 A1
20070207225 Squadrito Sep 2007 A1
20070225281 Zhang et al. Sep 2007 A1
20070232574 Bernard et al. Oct 2007 A1
20070238713 Gast et al. Oct 2007 A1
20070243229 Smith et al. Oct 2007 A1
20070248658 Bracht et al. Oct 2007 A1
20070254858 Cronk Nov 2007 A1
20070255197 Wilkins et al. Nov 2007 A1
20070264309 Chollet et al. Nov 2007 A1
20070264345 Eros et al. Nov 2007 A1
20070264349 Lee et al. Nov 2007 A1
20070270394 El-Alfy et al. Nov 2007 A1
20070281008 Lin Dec 2007 A1
20070286819 DeVries et al. Dec 2007 A1
20070287688 Chan et al. Dec 2007 A1
20070287789 Jones et al. Dec 2007 A1
20070292359 Schuz et al. Dec 2007 A1
20070292387 Jon et al. Dec 2007 A1
20070292461 Danziger et al. Dec 2007 A1
20070292493 Brierre Dec 2007 A1
20070298089 Yoshinaga et al. Dec 2007 A1
20080026035 Chollet et al. Jan 2008 A1
20080026040 Rivera et al. Jan 2008 A1
20080026062 Farr et al. Jan 2008 A1
20080038219 Carlson et al. Feb 2008 A1
20080038350 Gerecke et al. Feb 2008 A1
20080039405 Joseph et al. Feb 2008 A1
20080050317 Besonov et al. Feb 2008 A1
20080051351 Ghisalberti Feb 2008 A1
20080063607 Berman et al. Mar 2008 A1
20080069779 Schuz et al. Mar 2008 A1
20080069791 Beissert Mar 2008 A1
20080085877 Bortz Apr 2008 A1
20080095831 Mc Graw Apr 2008 A1
20080095838 Abou Chacra-Vernet Apr 2008 A1
20080119537 Zhang et al. May 2008 A1
20080125402 Dilberti May 2008 A1
20080138379 Jennings-Spring Jun 2008 A1
20080138390 Gricenko et al. Jun 2008 A1
20080139392 Yuan et al. Jun 2008 A1
20080145423 Khan et al. Jun 2008 A1
20080153789 Dmowski et al. Jun 2008 A1
20080175814 Phiasivongsa et al. Jul 2008 A1
20080175905 Baksh et al. Jul 2008 A1
20080175908 Baksh et al. Jul 2008 A1
20080188829 Creasy Aug 2008 A1
20080206156 Cronk Aug 2008 A1
20080206159 Schuz et al. Aug 2008 A1
20080206161 Tamarkin et al. Aug 2008 A1
20080214512 Seitz et al. Sep 2008 A1
20080220069 Allison Sep 2008 A1
20080226698 Beste et al. Sep 2008 A1
20080227763 Paris et al. Sep 2008 A1
20080234199 Katamreddy Sep 2008 A1
20080234240 Duesterberg et al. Sep 2008 A1
20080255078 Katamreddy Oct 2008 A1
20080255089 Katamreddy Oct 2008 A1
20080261931 Stenlof et al. Oct 2008 A1
20080113953 DeVries et al. Dec 2008 A1
20080114050 Fensome et al. Dec 2008 A1
20080299220 Tamarkin et al. Dec 2008 A1
20080306036 Katamreddy Dec 2008 A1
20080312197 Rodriguez Dec 2008 A1
20080312198 Rodriguez Dec 2008 A1
20080319078 Katamreddy Dec 2008 A1
20090004246 Woolfson et al. Jan 2009 A1
20090010968 Peyrot et al. Jan 2009 A1
20090011041 Musaeva et al. Jan 2009 A1
20090017120 Brisco et al. Jan 2009 A1
20090022683 Park et al. Jan 2009 A1
20090047357 Tomohira et al. Feb 2009 A1
20090053294 Prendergast Feb 2009 A1
20090060982 Ron et al. Mar 2009 A1
20090060997 Seitz et al. Mar 2009 A1
20090068118 Eini et al. Mar 2009 A1
20090074859 Patel Mar 2009 A1
20090081206 Leibovitz Mar 2009 A1
20090081278 De Graaff et al. Mar 2009 A1
20090081303 Savoir et al. Mar 2009 A1
20090092656 Klamerus et al. Apr 2009 A1
20090093440 Murad Apr 2009 A1
20090098069 Vacca Apr 2009 A1
20090099106 Phiasivongsa et al. Apr 2009 A1
20090099149 Kresevic et al. Apr 2009 A1
20090130029 Tamarkin et al. May 2009 A1
20090131385 Voskuhl May 2009 A1
20090136574 Diaz-Astruc et al. May 2009 A1
20090137478 Bernstein et al. May 2009 A1
20090137538 Klamerus et al. May 2009 A1
20090143344 Chang Jun 2009 A1
20090164341 Sunvoid et al. Jun 2009 A1
20090175799 Tamarkin et al. Jul 2009 A1
20090181088 Song et al. Jul 2009 A1
20090186081 Slot et al. Jul 2009 A1
20090197843 Notelovitz et al. Aug 2009 A1
20090203658 Rose et al. Aug 2009 A1
20090214474 Jennings Aug 2009 A1
20090227025 Nichols et al. Sep 2009 A1
20090227550 Mattern Sep 2009 A1
20090232897 Sahoo et al. Sep 2009 A1
20090258096 Cohen Oct 2009 A1
20090264395 Creasy Oct 2009 A1
20090269403 Shaked et al. Oct 2009 A1
20090285772 Phiasivongsa et al. Nov 2009 A1
20090285869 Trimble Nov 2009 A1
20090318558 Kim et al. Dec 2009 A1
20090324714 Kresevic et al. Dec 2009 A1
20090325916 Zhang et al. Dec 2009 A1
20100008985 Vermeulen et al. Jan 2010 A1
20100028360 Atwood Feb 2010 A1
20100034838 Staniforth et al. Feb 2010 A1
20100034880 Sintov et al. Feb 2010 A1
20100040671 Ahmed et al. Feb 2010 A1
20100048523 Bachman et al. Feb 2010 A1
20100055138 Jacobs et al. Mar 2010 A1
20100074959 Hansom et al. Mar 2010 A1
20100086501 Chang et al. Apr 2010 A1
20100086599 Huempel et al. Apr 2010 A1
20100092568 Lerner et al. Apr 2010 A1
20100105071 Laufer et al. Apr 2010 A1
20100119585 Hille et al. May 2010 A1
20100129320 Phiasivongsa et al. May 2010 A1
20100136105 Chen et al. Jun 2010 A1
20100137265 Leonard Jun 2010 A1
20100137271 Chen et al. Jun 2010 A1
20100143420 Lee et al. Jun 2010 A1
20100143481 Shenoy et al. Jun 2010 A1
20100150993 Theobald et al. Jun 2010 A1
20100152144 Hermsmeyer Jun 2010 A1
20100168228 Bose et al. Jul 2010 A1
20100183723 Laurent-Applegate et al. Jul 2010 A1
20100184736 Coelingh Bennink et al. Jul 2010 A1
20100190758 Fauser et al. Jul 2010 A1
20100204326 D Souza Aug 2010 A1
20100210994 Zarif Aug 2010 A1
20100221195 Ziv et al. Sep 2010 A1
20100227797 Danielsson et al. Sep 2010 A1
20100240626 Kulkarni et al. Sep 2010 A1
20100247482 Chen et al. Sep 2010 A1
20100247632 Dong et al. Sep 2010 A1
20100247635 Schmidt et al. Sep 2010 A1
20100255085 Liu et al. Oct 2010 A1
20100273730 Hsu et al. Oct 2010 A1
20100278759 Murad Nov 2010 A1
20100279988 Setiawan et al. Nov 2010 A1
20100291191 Lapitsky et al. Nov 2010 A1
20100292199 Leverd et al. Nov 2010 A1
20100303825 Sirbasku Dec 2010 A9
20100312137 Gilmour et al. Dec 2010 A1
20100316724 Whitfield et al. Dec 2010 A1
20100322884 Wilkins et al. Dec 2010 A1
20100330168 Gicquel et al. Dec 2010 A1
20110028439 Witt-Enderby et al. Feb 2011 A1
20110039814 Ross et al. Feb 2011 A1
20110053845 Levine et al. Mar 2011 A1
20110066473 Bernick et al. Mar 2011 A1
20110076775 Stewart et al. Mar 2011 A1
20110076776 Stewart et al. Mar 2011 A1
20110086825 Chatroux Apr 2011 A1
20110087192 Uhland et al. Apr 2011 A1
20110091555 De Luigi Bruschi et al. Apr 2011 A1
20110098258 Canet et al. Apr 2011 A1
20110098631 McIntyre et al. Apr 2011 A1
20110104268 Segot et al. May 2011 A1
20110104289 Savoir Vilboeuf et al. May 2011 A1
20110130372 Marliani et al. Jun 2011 A1
20110135719 Besins et al. Jun 2011 A1
20110142945 Chen et al. Jun 2011 A1
20110152840 Lee et al. Jun 2011 A1
20110158920 Fisher et al. Jun 2011 A1
20110171140 Illum et al. Jul 2011 A1
20110182997 Lewis et al. Jul 2011 A1
20110190201 Wood, Jr. et al. Aug 2011 A1
20110195031 Du Aug 2011 A1
20110195114 Carrara et al. Aug 2011 A1
20110195944 Mura et al. Aug 2011 A1
20110217341 Sah Sep 2011 A1
20110238003 Karabelas et al. Sep 2011 A1
20110244043 Wang et al. Oct 2011 A1
20110250256 Hyun Oh et al. Oct 2011 A1
20110250259 Buckman Oct 2011 A1
20110250274 Shaked et al. Oct 2011 A1
20110256092 Phiasivongsa et al. Oct 2011 A1
20110262373 Umbert Millet Oct 2011 A1
20110262494 Achleitner et al. Oct 2011 A1
20110268665 Tamarkin et al. Nov 2011 A1
20110275584 Volkmann et al. Nov 2011 A1
20110281832 Wennogle et al. Nov 2011 A1
20110287094 Penhasi et al. Nov 2011 A1
20110293720 General et al. Dec 2011 A1
20110294738 Kuliopulos et al. Dec 2011 A1
20110300167 Covic et al. Dec 2011 A1
20110301087 McBride et al. Dec 2011 A1
20110306579 Stein Dec 2011 A1
20110311592 Birbara Dec 2011 A1
20110312927 Nachaegari et al. Dec 2011 A1
20110312928 Nachaegari et al. Dec 2011 A1
20110318405 Erwin Dec 2011 A1
20110318431 Gulati Dec 2011 A1
20120009276 De Groote Jan 2012 A1
20120015350 Nabatiyan et al. Jan 2012 A1
20120021041 Rossi et al. Jan 2012 A1
20120028888 Janz et al. Feb 2012 A1
20120028910 Takruri et al. Feb 2012 A1
20120028936 Popova et al. Feb 2012 A1
20120045532 Cohen Feb 2012 A1
20120046264 Lieb et al. Feb 2012 A1
20120046518 Yoakum et al. Feb 2012 A1
20120052077 Truitt, III et al. Mar 2012 A1
20120058171 Zeeman et al. Mar 2012 A1
20120058962 Sparrow et al. Mar 2012 A1
20120058979 Auspitz et al. Mar 2012 A1
20120064135 Harms et al. Mar 2012 A1
20120065179 Andersson Mar 2012 A1
20120065221 Babul Mar 2012 A1
20120087872 Schuz et al. Apr 2012 A1
20120101073 Mannion et al. Apr 2012 A1
20120121517 Kim et al. May 2012 A1
20120121692 Fang et al. May 2012 A1
20120122829 Masini et al. May 2012 A1
20120128625 Shalwitz et al. May 2012 A1
20120128654 Terpstra et al. May 2012 A1
20120128683 Shantha May 2012 A1
20120128733 Perrin et al. May 2012 A1
20120128777 Keck et al. May 2012 A1
20120129773 Geier et al. May 2012 A1
20120129819 Vancaillie et al. May 2012 A1
20120136013 Wennogle et al. May 2012 A1
20120142645 Marx Jun 2012 A1
20120148670 Lee et al. Jun 2012 A1
20120149748 Shanler et al. Jun 2012 A1
20120172343 Schuermann et al. Jul 2012 A1
20120184515 Schwede et al. Jul 2012 A1
20120231052 Brinton et al. Sep 2012 A1
20120232011 Kneissel Sep 2012 A1
20120232042 Krenz et al. Sep 2012 A1
20120263679 Wallace et al. Oct 2012 A1
20120269721 Weng et al. Oct 2012 A1
20120277249 Tarrand et al. Nov 2012 A1
20120277727 Doshi et al. Nov 2012 A1
20120283671 Shibata et al. Nov 2012 A1
20120295911 Mannion et al. Nov 2012 A1
20120301517 Warner et al. Nov 2012 A1
20120301538 Latere et al. Nov 2012 A1
20120302535 Caufriez et al. Nov 2012 A1
20120316130 Hendrix Dec 2012 A1
20120316496 Horres et al. Dec 2012 A1
20120321579 Edelson et al. Dec 2012 A1
20120322779 Voskuhl Dec 2012 A9
20120328549 Edelson et al. Dec 2012 A1
20120329738 Liu Dec 2012 A1
20130004619 Goh et al. Jan 2013 A1
20130011342 Hazot et al. Jan 2013 A1
20130017239 Fernandez et al. Jan 2013 A1
20130022674 Dudley et al. Jan 2013 A1
20130023505 Garfield et al. Jan 2013 A1
20130023823 Volland et al. Jan 2013 A1
20130028850 Hazot et al. Jan 2013 A1
20130029947 Nachaegari et al. Jan 2013 A1
20130029957 Venkateshwaran et al. Jan 2013 A1
20130045266 Kang et al. Feb 2013 A1
20130045953 Grenier et al. Feb 2013 A1
20130059795 Lo et al. Mar 2013 A1
20130064897 Binay Mar 2013 A1
20130072466 Choi et al. Mar 2013 A1
20130084257 Ishida et al. Apr 2013 A1
20130085123 Zhao et al. Apr 2013 A1
20130089574 Stock et al. Apr 2013 A1
20130090318 Gainer et al. Apr 2013 A1
20130102781 Ely et al. Apr 2013 A1
20130108551 Gruell et al. May 2013 A1
20130116215 Lleo et al. May 2013 A1
20130116222 Altomari et al. May 2013 A1
20130122051 Gullapalli et al. May 2013 A1
20130123175 McKee et al. May 2013 A1
20130123220 Queiroz et al. May 2013 A1
20130123351 Dewitt May 2013 A1
20130129818 Bernick et al. May 2013 A1
20130131027 Schmitz et al. May 2013 A1
20130131028 Snyder et al. May 2013 A1
20130131029 Baltussen et al. May 2013 A1
20130149314 Bullerdiek et al. Jun 2013 A1
20130150334 Sun et al. Jun 2013 A1
20130164225 Besonov et al. Jun 2013 A1
20130164346 Son et al. Jun 2013 A1
20130165744 Carson et al. Jun 2013 A1
20130178452 King Jul 2013 A1
20130183254 Cochran et al. Jul 2013 A1
20130183325 Sforzini et al. Jul 2013 A1
20130189193 Besonov et al. Jul 2013 A1
20130189196 Tamarkin et al. Jul 2013 A1
20130189230 Kooy et al. Jul 2013 A1
20130189368 Mosqueira et al. Jul 2013 A1
20130210709 Covic et al. Aug 2013 A1
20130216550 Penninger et al. Aug 2013 A1
20130216596 Fernandez et al. Aug 2013 A1
20130224177 Kim et al. Aug 2013 A1
20130224257 Sah et al. Aug 2013 A1
20130224268 Jaikaria et al. Aug 2013 A1
20130224300 Maggio Aug 2013 A1
20130225412 Sardari Lodriche et al. Aug 2013 A1
20130225542 Frick et al. Aug 2013 A1
20130226113 Langguth et al. Aug 2013 A1
20130243696 Wang et al. Sep 2013 A1
20130245253 Mook et al. Sep 2013 A1
20130245570 Jackson Sep 2013 A1
20130261096 Merian et al. Oct 2013 A1
20130266645 Schoenecker et al. Oct 2013 A1
20130267485 Da Silva Maia Filho et al. Oct 2013 A1
20130273167 Kim et al. Oct 2013 A1
20130274211 Prusthy et al. Oct 2013 A1
20130280213 Voskuhl Oct 2013 A1
20130316374 Menon et al. Nov 2013 A1
20130317065 Seto et al. Nov 2013 A1
20130317315 Tsang et al. Nov 2013 A1
20130324565 Zhao et al. Dec 2013 A1
20130331363 Zhao et al. Dec 2013 A1
20130338122 Bernick et al. Dec 2013 A1
20130338123 Bernick et al. Dec 2013 A1
20130338124 Zhao et al. Dec 2013 A1
20130345187 Rodriguez Oquendo Dec 2013 A1
20140018335 Keto et al. Jan 2014 A1
20140024590 Taylor et al. Jan 2014 A1
20140031289 Kim et al. Jan 2014 A1
20140031323 Perez Jan 2014 A1
20140066416 Leunis et al. Mar 2014 A1
20140072531 Oh et al. Mar 2014 A1
20140079686 Prouty et al. Mar 2014 A1
20140088051 Bernick et al. Mar 2014 A1
20140088058 Maurizio Mar 2014 A1
20140088059 Santha et al. Mar 2014 A1
20140094426 Drummond et al. Apr 2014 A1
20140094440 Bernick et al. Apr 2014 A1
20140094441 Bernick et al. Apr 2014 A1
20140099362 Bernick et al. Apr 2014 A1
20140100159 Conrad Apr 2014 A1
20140100204 Bernick et al. Apr 2014 A1
20140100205 Bernick et al. Apr 2014 A1
20140100206 Cacace et al. Apr 2014 A1
20140113889 Haine et al. Apr 2014 A1
20140127185 Sayeed et al. May 2014 A1
20140127280 Jukarainen et al. May 2014 A1
20140127308 Opara et al. May 2014 A1
20140128798 Malanchine et al. May 2014 A1
20140148491 Valia et al. May 2014 A1
20140186332 Ezrin et al. Jul 2014 A1
20140187487 Shoichet et al. Jul 2014 A1
20140193523 Henry Jul 2014 A1
20140194396 Wennogle et al. Jul 2014 A1
20140206616 Ko et al. Jul 2014 A1
20140213565 Bernick et al. Jul 2014 A1
20140329783 Bernick et al. Nov 2014 A1
20140370084 Bernick et al. Dec 2014 A1
20140371182 Bernick et al. Dec 2014 A1
20140371183 Bernick et al. Dec 2014 A1
20140371184 Bernick et al. Dec 2014 A1
20140371185 Bernick et al. Dec 2014 A1
20150031654 Amadio Jan 2015 A1
20150133421 Bernick et al. May 2015 A1
20150148323 Bernick et al. May 2015 A1
20150164789 Bernick et al. Jun 2015 A1
20150224117 Bernick et al. Aug 2015 A1
20150224118 Bernick et al. Aug 2015 A1
20150297733 Oberegger et al. Oct 2015 A1
20150302435 Bernick et al. Oct 2015 A1
20150342963 Bernick et al. Dec 2015 A1
20150352126 Bernick et al. Dec 2015 A1
20150359737 Bernick et al. Dec 2015 A1
20160030449 Persicaner et al. Feb 2016 A1
20160213685 Bernick et al. Jul 2016 A1
20170056418 Thorsteinsson et al. Mar 2017 A1
20170216310 Mirkin et al. Aug 2017 A1
20170281645 Shadiack et al. Oct 2017 A1
20170281646 Inskeep et al. Oct 2017 A1
20170281647 Shadiack et al. Oct 2017 A1
20170281776 Shadiack et al. Oct 2017 A1
20180161343 Mirkin et al. Jun 2018 A1
20180161344 Bernick et al. Jun 2018 A1
20180161345 Bernick et al. Jun 2018 A1
20180221389 Amadio et al. Aug 2018 A1
20180256598 Mirkin et al. Sep 2018 A1
20180280410 Amadio et al. Oct 2018 A1
20180289723 Bernick et al. Oct 2018 A1
20190022107 Mirkin et al. Jan 2019 A1
20190046542 Bernick et al. Feb 2019 A1
20190070197 Amadio et al. Mar 2019 A1
20190142844 Bernick et al. May 2019 A1
20190247401 Amadio et al. Aug 2019 A1
20190343771 Mirkin et al. Nov 2019 A1
20190343845 Bernick et al. Nov 2019 A1
20190358243 Mirkin et al. Nov 2019 A1
20200069700 Bernick et al. Mar 2020 A1
20200147104 Bernick May 2020 A1
20200230153 Bernick et al. Jul 2020 A1
20200230154 Bernick et al. Jul 2020 A1
20200276210 Amadio et al. Sep 2020 A1
20200281849 Bernick et al. Sep 2020 A1
20200297736 Bernick et al. Sep 2020 A1
Foreign Referenced Citations (200)
Number Date Country
PI1001367 Jul 2012 BR
2044371 Dec 1991 CA
2612380 Dec 2006 CA
102258455 Nov 2011 CN
0261429 Mar 1988 EP
275716 Jul 1988 EP
0279977 Aug 1988 EP
622075 Nov 1994 EP
785211 Jul 1997 EP
785212 Jul 1997 EP
811381 Dec 1997 EP
0904064 Mar 1999 EP
0813412 Dec 1999 EP
0750495 Dec 2002 EP
1300152 Apr 2003 EP
1094781 Jul 2008 EP
2191833 Jun 2010 EP
452238 Aug 1936 GB
720561 Dec 1954 GB
848881 Sep 1960 GB
874368 Aug 1961 GB
1589946 May 1981 GB
2005KO00053 Aug 2005 IN
216026 Mar 2008 IN
244217 Nov 2010 IN
H4-503810 Sep 1990 JP
H2-264725 Oct 1990 JP
2002 510336 Apr 2002 JP
2006 513182 Apr 2006 JP
2155582 Sep 2000 RU
199010425 Sep 1990 WO
1990011064 Oct 1990 WO
1993017686 Sep 1993 WO
1994022426 Oct 1994 WO
1995005807 Mar 1995 WO
1995030409 Nov 1995 WO
1996009826 Apr 1996 WO
1996019975 Jul 1996 WO
1996030000 Oct 1996 WO
1997005491 Feb 1997 WO
1997040823 Nov 1997 WO
1997043989 Nov 1997 WO
1998010293 Mar 1998 WO
1998032465 Jul 1998 WO
1998041217 Sep 1998 WO
1998051280 Nov 1998 WO
199922680 May 1999 WO
1999032072 Jul 1999 WO
1999039700 Aug 1999 WO
1999042109 Aug 1999 WO
1999043304 Sep 1999 WO
1999048477 Sep 1999 WO
1999052528 Oct 1999 WO
1999053910 Oct 1999 WO
1999055333 Nov 1999 WO
1999062497 Dec 1999 WO
1999063974 Dec 1999 WO
2000001351 Jan 2000 WO
2000006175 Feb 2000 WO
2000038659 Jun 2000 WO
2000045795 Aug 2000 WO
2000050007 Aug 2000 WO
2000059577 Oct 2000 WO
2000076522 Dec 2000 WO
2001037808 May 2001 WO
2001054699 Aug 2001 WO
2001060325 Aug 2001 WO
2001087276 Nov 2001 WO
2001091757 Dec 2001 WO
2002007700 Jan 2002 WO
2002011768 Feb 2002 WO
2002022132 Mar 2002 WO
2002040008 May 2002 WO
2002041878 May 2002 WO
2002053131 Jul 2002 WO
2002078602 Oct 2002 WO
2002078604 Oct 2002 WO
2003028667 Apr 2003 WO
2003041718 May 2003 WO
2003041741 May 2003 WO
2003068186 Aug 2003 WO
2003077923 Sep 2003 WO
2003082254 Oct 2003 WO
2003092588 Nov 2003 WO
2004014397 Feb 2004 WO
2004014432 Feb 2004 WO
2004017983 Mar 2004 WO
2004032897 Apr 2004 WO
2004032942 Apr 2004 WO
2004052336 Jun 2004 WO
2004054540 Jul 2004 WO
2004054576 Jul 2004 WO
2004080413 Sep 2004 WO
2004105694 Dec 2004 WO
2004110402 Dec 2004 WO
2004110408 Dec 2004 WO
2005027911 Mar 2005 WO
2005030175 Apr 2005 WO
2005081825 Sep 2005 WO
2005087194 Sep 2005 WO
2005087199 Sep 2005 WO
2005105059 Nov 2005 WO
2005115335 Dec 2005 WO
2005120470 Dec 2005 WO
2005120517 Dec 2005 WO
2006013369 Feb 2006 WO
2006034090 Mar 2006 WO
2006036899 Apr 2006 WO
2006053172 May 2006 WO
2006105615 Oct 2006 WO
2006113505 Oct 2006 WO
2006138686 Dec 2006 WO
2006138735 Dec 2006 WO
2007045027 Apr 2007 WO
2007076144 Jul 2007 WO
2007103294 Sep 2007 WO
2007120868 Oct 2007 WO
2007123790 Nov 2007 WO
2007124250 Nov 2007 WO
2007144151 Dec 2007 WO
2008049516 May 2008 WO
2008152444 Dec 2008 WO
2009002542 Dec 2008 WO
2009036311 Mar 2009 WO
2009040818 Apr 2009 WO
2009069006 Jun 2009 WO
2009098072 Aug 2009 WO
2009133352 Nov 2009 WO
2010033188 Mar 2010 WO
2010146872 Dec 2010 WO
2011000210 Jan 2011 WO
2011073995 Jun 2011 WO
2011120084 Oct 2011 WO
2011128336 Oct 2011 WO
2012009778 Jan 2012 WO
2012024361 Feb 2012 WO
2012055814 May 2012 WO
2012055840 May 2012 WO
2012065740 May 2012 WO
2012098090 Jul 2012 WO
2012116277 Aug 2012 WO
2012118563 Sep 2012 WO
2012120365 Sep 2012 WO
2012127501 Sep 2012 WO
2012156561 Nov 2012 WO
2012156822 Nov 2012 WO
2012158483 Nov 2012 WO
2012166909 Dec 2012 WO
2012170578 Dec 2012 WO
2013011501 Jan 2013 WO
2013025449 Feb 2013 WO
2013028639 Feb 2013 WO
2013035101 Mar 2013 WO
2013044067 Mar 2013 WO
2013045404 Apr 2013 WO
2013059285 Apr 2013 WO
2013063279 May 2013 WO
2013064620 May 2013 WO
2013071281 May 2013 WO
2013078422 May 2013 WO
2013088254 Jun 2013 WO
2013102665 Jul 2013 WO
2013106437 Jul 2013 WO
2013112947 Aug 2013 WO
2013113690 Aug 2013 WO
2013124415 Aug 2013 WO
2013127727 Sep 2013 WO
2013127728 Sep 2013 WO
2013144356 Oct 2013 WO
2013149258 Oct 2013 WO
2013158454 Oct 2013 WO
2013170052 Nov 2013 WO
2013178587 Dec 2013 WO
2013181449 Dec 2013 WO
2013192248 Dec 2013 WO
2013192249 Dec 2013 WO
2013192250 Dec 2013 WO
2013192251 Dec 2013 WO
2014001904 Jan 2014 WO
2014004424 Jan 2014 WO
2014009434 Jan 2014 WO
2014018569 Jan 2014 WO
2014018570 Jan 2014 WO
2014018571 Jan 2014 WO
2014018856 Jan 2014 WO
2014018932 Jan 2014 WO
2014031958 Feb 2014 WO
2014041120 Mar 2014 WO
2014052792 Apr 2014 WO
2014056897 Apr 2014 WO
2014066442 May 2014 WO
2014074846 May 2014 WO
2014076231 May 2014 WO
2014076569 May 2014 WO
2014081598 May 2014 WO
2014086739 Jun 2014 WO
2014093114 Jun 2014 WO
2014104784 Jul 2014 WO
2015179782 Nov 2015 WO
2016018993 Feb 2016 WO
Non-Patent Literature Citations (437)
Entry
US 6,214,374 B1, 04/2001, Schmirler et al. (withdrawn)
UNC, “Inserting of suppositories”, Nov. 3, 2008, https://pharmlabs.unc.edu/labs/suppository/inserting.htm, retrieved on Apr. 4, 2021. (Year: 2008).
Office Action dated Jan. 30, 2017, in U.S. Appl. No. 14/649,818 Inventors, Bernick, B.A., Jun. 4, 2015, 12 pages.
Office Action dated Nov. 2, 2017, in U.S. Appl. No. 14/649,818 Inventors, Bernick, B.A., Jun. 4, 2015, 21 pages.
Office Action dated Jun. 15, 2018, in U.S. Appl. No. 14/649,818 Inventors, Bernick, B.A., Jun. 4, 2015, 21 pages.
Office Action dated Jun. 3, 2017, in U.S. Appl. No. 14/649,818 Inventors, Bernick, B.A., Jun. 4, 2015, 15 pages.
Office Action dated Jan. 5, 2016, in U.S. Appl. No. 14/521,002 Inventors, Bernick, B.A., Oct. 22, 2014, 9 pages.
Office Action dated Jan. 6, 2017, in U.S. Appl. No. 14/521,002 Inventors, Bernick, B.A., Oct. 22, 2014, 10 pages.
Office Action dated Oct. 5, 2017, in U.S. Appl. No. 14/521,002 Inventors, Bernick, B.A., Oct. 22, 2014, 13 pages.
Office Action dated Jul. 20, 2018, in U.S. Appl. No. 14/521,002 Inventors, Bernick, B.A., Oct. 22, 2014, 16 pages.
Office Action dated Jun. 3, 2019, in U.S. Appl. No. 14/521,002 Inventors, Bernick, B.A., Oct. 22, 2014, 12 pages.
Office Action dated Apr. 2, 2020, in U.S. Appl. No. 14/521,002 Inventors, Bernick, B.A., Oct. 22, 2014, 6 pages.
Kingsburg, S.A et al., “Treating dyspareunia caused by vaginal atrophy: a review of treatment options using vaginal estrogen therapy,” International Journal of Women's Health 1:105-111, Dove Press, England (2009).
ACTIVELLA® (estradiol/ norethindrone acetate) prescribing information (Nov. 2017) FDA Label, 39 pages.
PROMETRIUM® (progesterone, USP) prescribing information (Jun. 2009) FDA Label, 33 pages.
VAGIFEM® (estradiol vaginal tablets) prescribing information (Nov. 2009) FDA Label, 14 pages.
MacBride, M.B., et al., “Vulvovaginal Atrophy,” Mayo Clinic Proceedings, 85(1): 87-94, Elsevier, Netherlands (2010).
De Vries, T.P.G.M., et al., “Guide to Good Prescribing: A Practical Manual,” Essential Medicines and Health Products Information Portal, World Health Organization, Annex 3 (“How to explain the use of some dosage forms”), Checklist 11 (“Vaginal tablet without applicator”) available at https://apps.who.int/iris/handle/10665/59001 (4 pages)(1994).
Rioux, J.E., et al “17 beta-Estradiol Vaginal Tablet Versus Conjugated Equine Estrogen Vaginal Cream to Relieve Menopausal Atrophic Vaginitis,” Menopause, 7(3): 156-161, The North American Menopause Society, United States (2000).
Cicinelli, E. et al “Placement of the vaginal 17 beta-estradiol tablets in the inner or outer one third of the vagina affects the preferential delivery of 17 beta-estradiol toward the uterus or periurethral areas, thereby modifying efficacy and endometrial safety,” Am. J. Obstet. Gynecol, 189: 55-58 (2003).
Office Action dated Apr. 30, 2021, in U.S. Appl. No. 16/834,780, Bernick, B. A., et al., filed Apr. 30, 2021, 13 pages.
Office Action dated Jun. 25, 2020, in U.S. Appl. No. 16/834,844, Bernick, B. A., et al., filed Mar. 30, 2020, 12 pages.
Notice of Allowance dated Aug. 21, 2020, in U.S. Appl. No. 16/834,844, Bernick, B. A., et al., filed Mar. 30, 2020, 6 pages.
Office Action dated Aug. 7, 2020, in U.S. Appl. No. 16/833,213, Bernick, B. A., et al., filed Mar. 27, 2020, 14 pages.
Bassi, P., and Kaur, G., “Innovations in bioadhesive vaginal drug delivery system,” Expert Opinion on Therapeutic Patents 22(9): 1019-1032, Taylor and Francis Ltd., United Kingdom (published online Aug. 2012, published in print Sep. 2012).
Office Action dated Apr. 8, 2021, in U.S. Appl. No. 16/833,188, Bernick, B. A., et al., filed Mar. 27, 2020, 16 pages.
UNC School of Pharmacy, “Preparation of Suppositories,” PharmLabs.unc.edu, accessed at http://pharmlabs.unc.edu/labs/suppository/inserting.htm on Apr. 16, 2021, 1 page.
Final Office Action dated Oct. 8, 2021, in U.S. Appl. No. 16/834,780, Bernick, B. A., et al., filed Mar. 30, 2020, 12 pages.
Notice of Allowance dated Oct. 7, 2020, in U.S. Appl. No. 16/833,213, Bernick, B. A., et al., filed Mar. 27, 2020, 5 pages.
Gullapalli, R.P., “Soft Gelatin Capsules (Softgels),” J. Pharm. Sci., 99, No. 10, Oct. 2010.
Office Action dated Nov. 27, 2020, in U.S. Appl. No. 16/837,929, Bernick, B. A., et al., filed Apr. 1, 2020, 15 pages.
Notice of Allowance dated Mar. 12, 2021, in U.S. Appl. No. 16/837,929, Bernick, B. A., et al., filed Apr. 1, 2020, 8 pages.
Office Action dated Jul. 10, 2020, in U.S. Appl. No. 16/837,937, Bernick, B. A., et al., filed Apr. 1, 2020, 14 pages.
Final Office Action dated Nov. 30, 2020, in U.S. Appl. No. 16/837,937, Bernick, B. A., et al., filed Apr. 1, 2020, 15 pages.
Notice of Allowance dated Mar. 17, 2021, in U.S. Appl. No. 16/837,937, Bernick, B. A., et al., filed Apr. 1, 2020, 7 pages.
Office Action dated Oct. 6, 2020, in U.S. Appl. No. 16/837,933, Bernick, B. A., et al., filed Apr. 1, 2020, 14 pages.
Final Office Action dated Feb. 4, 2021, in U.S. Appl. No. 16/837,933, Bernick, B. A., et al., filed Apr. 1, 2020, 15 pages.
Notice of Allowance dated Mar. 12, 2021, in U.S. Appl. No. 16/837,933, Bernick, B. A., et al., filed Apr. 1, 2020, 7 pages.
Office Action dated Apr. 30, 2021 in U.S. Appl. No. 16/875,030, Bernick, B. A., et al., filed May 15, 2020, 17 pages.
Dugal et al., “Comparison of usefulness of estradiol vaginal tablets and estriol vagitories for treatment of vaginal atrophy” Acta Obstericia et Gynecologia Scandinavica, 79, 2000, pp. 293-297.
Office Action dated Apr. 8, 2021, in U.S. Appl. No. 16/677,831, Bernick, B. A., et al., filed Nov. 8, 2019, 19 pages.
Rodriguez-Tenreiro, C. et al., “Cyclodextrin/carbopol micro-scale interpenetrating networks for dmg delivery” J. of Controlled Release 123, 2007, pp. 56-66.
Rodriguez-Tenreiro, C. et al., “Estradiol sustained release from high affinity cyclodextrin hydrogels” Eur. J. of Pharmaceutics and Biopharmaceutics 66, 2007, 55-62.
Notice of Allowance dated Sep. 8, 2020, in U.S. Appl. No. 14/521,002, Bernick, B. A., et al., filed Oct. 22, 2014, 6 pages.
Office Action dated Apr. 7, 2021, in U.S. Appl. No. 16/746,434, Bernick, B. A., et al., filed Jan. 17, 2020, 14 pages.
Notice of Allowance dated Oct. 6, 2021 in U.S. Appl. No. 16/746,434, Bernick, B. A., et al., filed Jan. 17, 2020, 10 pages.
Notice of Allowance dated Oct. 20, 2021 in U.S. Appl. No. 16/677,831, Bernick, B. A., et al., filed Oct. 20, 2021, 7 pages.
Notice of Allowance dated Oct. 6, 2021 in U.S. Appl. No. 16/833,188, Bernick, B. A., et al., filed Mar. 27, 2020, 10 pages.
Abbas et al., “Regression of endometrial implants treated with vitamin D3 in a rat model of endometriosis,” European J of Pharma, Elsevier, 2013, 715:72-75.
Abitec, CapmulMCM, EP, Technical Data Sheet, version 10, 2014, Columbus, OH, 1 page.
Abitec, CapmulMCM, NF, Technical Data Sheet, version 6, 2014, Columbus, OH. 1 page.
Abitec, CapmulMCM, Safety Data Sheet, 2011, Janesville, WI, 5 pages.
Abitec, CapmulMCM, Technical Data Sheet, version 17, 2014, Columbus, OH, 1 page.
Abitec, CapmulPG8, CAS No. 31565-12-5, version 11, 2006, Columbus, OH, 2 pages.
Abitec, Excipients for the Pharmaceutical Industry—Regulatory and Product Information, 2013, 2 pages.
Acarturk, Fusun, “Mucoadhesive Vaginal Drug Delivery System,” Recent Patents on Drug Delivery & Formulation, 2009, 3:193-205.
Alabi et al., “Analysis of Fatty Acid Composition of Thevetia peruviana and Hura crepitans Seed oils using GC-FID,” Fountain Journal of Nat. and Appl. Sciences, 2013, 2(2):32-37.
Alexander, KS, Com Oil, CAS No. 8001-30-7, Jan. 2009, 2 pages.
Alvarez et al., “Ectopic uterine tissue as a chronic pain generator,” Neuroscience, Dec. 6, 2012, 225:269-282.
Application Note FT-IR: JI-Ap-FT0508-008, CD spectra of pharmaceuticals substances—Steroids (2), JASCO International Co., Ltd., 2 pages.
Araya-Sibaja, Andrea Manela, et al., “Chemical Properties of Progesterone Selected Refer.,” SciFinder, 2014, American Chemical Society & US Natl. Lib. of Med., 6 pages.
Araya-sibaja, andrea m.a., “Morphology Study of Progesterone Polymorphs Prepared by Polymer-Induced Heteronucleation (PIHn),” Scanning, 2013, 35:213-21, Wiley Period., Inc.
Araya-sibaja, andrea manela, et al., “Polymorphism in Progesterone Selected References,” SciFinder, Feb. 24, 2014, pp. 1-12, American Chem. Society & Natl. Lib. of Med.
Araya-sibaja, andrea manela, et al., “Polymorphism in Progesterone,” SciFinder, Feb. 24, 2014, pp. 1-46, American Chem. Society & Natl. Lib. of Med.
Araya-sibaja et al., “Crystallization of progesterone polymorphs using polymer-induced heteronucleation (PIHn) method,” Drug Development and Industrial Pharmacy, Early Online, 2014, pp. 1-8.
Archer et al., “Effects of ospemifene on the female reproductive and urinary tracts: translation from preclinical models into clinical evidence,” Menopause: The Journal of the North American Menopause Society, 2015, 22(77):1-11.
Archer et al., “Estrace® vs Premarin® for Treatment of Menopausal Symptoms: Dosage Comparison Study,” Advances In Therapy®, Jan./Feb. 1992, 9(1):21-31.
Ashburn et al., “Cardiovascular, Hepatic and Renal Lesions in Mice Receiving Cortisone, Estrone and Progesterone,” Yale J Bilogy and Medicine, Feb. 1963, 35:329-340.
Azeem, adnan et al., “Microemulsions as a Surrogate Carrier for Dermal Drug Delivery” (abstract only), Drug Development and Industrial Pharmacy, May 2000, 35(5):525-547 http://informahealthcare.com/doi/abs/10.1080/03639040802448646.
Azure Pharma, Inc., ELESTRINTM—Estradiol Gel, Drug Info, http://dailymed.nlm.nih.gov/dailymed/archives/fdaDrugInfo.cfm?archiveid=11885, Aug. 2009, 25 pages.
Bakhmutova-albert, Ekaterina, et al., “Enhancing Aqueous Dissolution Rates of Progesterone via Cocrystallization,” SSCI, Division of Aptuit, Poster No. R6247, West Lafayette (publicly available before application filing date of Oct. 22, 2014), 1 page.
Banerjee, Sila, et al., On the Stability of Salivary Progesterone Under Various Conditions of Storage, Steroids, Dec. 1985, 46(6):967-974.
Barnett, Steven M, “Pressure-tuning infared and solution Raman spectroscopic studies of 17β-estradiol and several A-ring . . . ,” Vibrational Spectroscopy 8, Elsevier, 1995, pp. 263-277.
Bartosova, “Transdermal Drug Delivery In Vitro Using Diffusion Cells,” Current Medicinal Chemistry, 2012, 19:4671-4677, Bentham Science Publishers.
Benbow et al., “Distribution and Metabolism of Maternal Progesterone in the Uterus, Placenta, and Fetus during Rat Pregnancy,” Biology of Reproduction, 1995, 52:1327-1333.
Bernabei et al., “Release of progesterone polymorphs from dimethylpoly siloxane polymeric matrixes,” Bollettino Chimico Farmaceutico, 1983, 122(1):20-6, SciFinder (abstract only).
Bhavnani et al., “Misconception and Concerns about Bioidentical Hormones Used for Custom-Compounded Hormone Therapy,” J Clin Endocrin Metab, first published ahead of print Dec. 28, 2011 as doi: 10.1210/jc.2011-2492, Mar. 2012, 97(3), 4 pages.
Bhavnani et al., “Structure Activity Relationships and Differential Interactions and Functional Activity of Various Equine Estrogens Mediated via Estrogen Receptors (ERs) ERα and ERβ,” Endocrinology, Oct. 2008, 149(10):4857 4870.
Bhavnani, b.r., Stanczyk, f.z., “Pharmacology of conjugated equine estrogens: Efficacy, safety and mechanism of action,” J. Steroid Biochem. Mol. Biol., 2013, 14 pages. Elsevier.
Bhavnani, B.R., Stanczyk, F.Z., “Use of medroxyprogesterone acetate for hormone therapy in postmenopausal women: Is it safe?, ” J. Steroid Biochem. Mol. Biol., 2013, 9 pages, Elsevier.
BioMed Central, Solubility of Progesterone in Organic Solvents, Online PDF, http://www.biomedcentral.com/content/supplementary/1475-2859-11-106-S2.pdf, 1 page (publicly available before application filing date of Oct. 22, 2014).
Blake et al., “Single and multidose pharmacokinetic study of a vaginal micronized progesterone insert (Endometrin) compared with vaginal gel in healthy reproductiveaged female subjects,” Fertility and Sterility#, 94(4), Sep. 2010, Elsevier, 6 pages.
Borka, laszlo, “Crystal Polymorphism of Pharmaceuticals,” Acta Pharm. Jugosl., 1990, 40:71-94.
Brandstatter-kuhnert, M, “Zur mikroskopischen Identitatsprufung und zur Polymorphie der Sexualhormone,” Acta, vol. 6, pp. 847-853, 1959, Univ. Innsbruck.
Brinton, l.a., felix, a.s., “Menopausal hormone therapy and risk of endometrial cancer,” 2013, J. Steroid Biochem. Mol. Biol., Elsevier, 7 pages.
British Pharmacopoeia 2014 Online, Refined Maize Oil, Ph. Eur. Monograph 1342, vol. I & II, Monographs: Medicinal and Pharmaceutical Substances, http://www.pharmacopoeia.co.uk/bp2014/ixbin/bp.cgi?a=print&id=7400&tab=a-z%20index[Feb. 3, 2014 1:37:50 PM], 2 pages.
Burry, “Percutaneous absorption of progesterone in postmenopausal women treated with transdermal estrogen,” Am J Obstet Gynecol, 1999, vol. 180(6) part 1, pp. 1504-1511.
Busetta, “Structure Cristalline et Moleculair de 1'Oestradiol Hemihydrate,” Acta Cryst., B28 pp. 560, 1972, Bis(dimethyl-o-thiolophenylarsine)palladium(II).
Busetta, “Structure Cristalline et Moleculaire du Complexe Oestradiol-Propanol,” Acta Cryst., B28 pp. 1349, 1972, J.A. Kanters and J. Kroon.
Campsteyn et al., “Structure Cristalline et Moleculaire de la Progesterone C21H30O2,” Acta Cryst., B28 pp. 3032-3042, 1972 (with English abstract).
Cendejas-Santana et al., “Growth and characterization of progesterone crystallites,” Revista Mexicana de Fisica, 2004, 50, Suplemento 1 pp. 1-3.
ChemPro, Top-Notch Technology in Production of Oils and Fats, Chempro-Edible-Oil-Refining-ISO-TUV-Austria, 3 pages (publicly available before application filing date of Oct. 22, 2014).
Christen et al., “Phase I/Pharmacokinetic Study of High-Dose Progesterone and Doxorubicin,” J Clin Oncol, 1993, 11:2417-2426.
Christensson et al., “Limonene hydroperoxide analogues differ in allergenic activity,” Contact Dermatitis 2008, 59:344-352.
Christensson et al., “Limonene hydroperoxide analogues show specific patch test reactions,” Contact Dermatitis, 2014, 70, 291-299.
Christensson et al., “Positive patch test reactions to oxidized limonene: exposure and relevance,” Contact Dermatitis, 2014, 71, 264-272.
Chun et al., “Transdermal Delivery of Estradiol and Norethrindrone Acetate: Effect of Vehicles . . . , ” J. Kor. Pharm. Sci., 2005, 35(3):173-174.
Cicinelli et al., “Direct Transport of Progesterone From Vagina to Uterus, Obstetrics & Gynecology,” 95(3), Mar. 2000, pp. 403-406.
Cole, Wayne & Julian, Percy l, Sterols. I. A Study of the 22-Ketosteroids, Cont. of the Research Lab. of the Glidden Co., Soya Prod. Div., vol. 67 pp. 1369-1375, Aug. 1945, Chicago.
Committee Opinion, Incidentally Detected Short Cervical Length, Committee of Obstetric Practice, Obstetrics & Gynecology, Acog, vol. 119, No. 4, Apr. 2012, pp. 879-882.
Commodari, Fernando, “Comparison of 17β-estradiol structures from x-ray diffraction and solution NMR,” Magn. Reson. Chem., 2005, 43:444-50, Wiley InterScience.
Cooper et al., “Systemic absorption of progesterone from Progest cream in postmenopausal women,” The Lancet, vol. 351, pp. 1255-1256, Research Letters, Apr. 25, 1998.
Corbett et al., “Trends in Pharmacy Compounding for Women's Health in North Carolina: Focus on Vulvodynia,” Southern Medical Journal, Jul. 2014, 107(7):433-436.
Corn Refiners Association, Corn Oil, 5th Edition, Washington, D.C., 2006, 24 pages.
Critchley et al., “Estrogen Receptor β, But Not Estrogen Receptor α, Is Present in the Vascular Endothelium of the Human and Nonhuman Primate Endometrium,” The Journal of Clinical Endocrinology & Metabolism, 2001, 86(3): 1370-1378.
Dauqan, Eqbal m. A., et al., “Fatty Acids Composition of Four Different Vegetable Oils (Red Palm Olein, Palm Olein, Corn Oil,” IPCBEE, vol. 14, 2011, pp. 31-34 IACSIT Press, Singapore.
Dideberg et al., “Crystal data on progesterone (C21H30O2), desoxycorticosterone (C21H30O3), corticosterone (C21H30O4) and aldosterone . . . ,” J. Appl. Cryst., 1971, 4:80.
Diramio, “Polyethylene Glycol Methacrylate/Dimetacrylate Hydrogels for Controlled Release of Hydrophobic Drugs,” Masters of Science Thesis, University of Georgia, Athens, Georgia, 2002, 131 pages.
Drakulic, “Role of complexes formation between drugs and penetration enhancers in transdermal . . . ,” Inter. Journal of Pharmaceutics, Elsevier, 2009, 363:40-49.
Du et al., “Percutaneous progesterone delivery via cream or gel application in postmenopausal women: a randomized cross-over study of progesterone levels in serum, whole blood, saliva, and capillary blood,” Menopause: The Journal of the North American Menopause Society, 2013, 20(11):1-7.
Duax et al., “Conformation of Progesterone Side Chain: Conflict between X-ray Data and Force-Field Calculations,” J. Am. Chem. Soc., Jun. 1981, 103, pp. 6705-6712.
Duclos et al., “Polymorphism of Progesterone: Influence of the carrier and of the solid dispersion manufacturing . . . ,” J. Thermal Anal., 1991, 37:1869-75, Wiley.
Ebian, “Ebian Article: Polymorphism and solvation of ethinyl estradiol,” SciFinder, Pharmaceutica Acta Helvetiae, vol. 54(4), pp. 111-114, 1979, Alexandria, Egypt (abstract only).
Eisenberger, a., Westhoff, c., “Hormone replacement therapy and venous thromboembolism,” J. Steroid Biochem. Mol. Biol., 2013, Elsevier, 7 pages.
Engelhardt et al., “Conceptus Influences the Distribution of Uterine Leukocytes During Early Porcine Pregnancy,” Biology of Reproduction, 2002, 66:1875-1880.
Ettinger et al., “Comparison of endometrial growth produced by unopposed conjugated estrogens or by micronized estradiol in postmenopausal women,” Am J Obstet Gynecol 1997, 176:112-117.
Excipients for Pharmaceuticals, Sasol Olefins & Surfactants Gmbh, 2010, 28 pages.
Faassen, Fried, “Physicochemical Properties and Transport of Steroids across Caco-2 Cells,” Pharmaceutical Research, vol. 20(2), 2003, Plenum Pub. Corp., 10 pages.
FDA, Draft Guidance on Progesterone, Recommended Apr. 2010, Revised Feb. 2011 http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM209294.pdf, 8 pages.
Ferrari, Roseli Ap., et al., “Oxidative Stability of Biodiesel From Soybean Oil Fatty Acid Ethyl Esters,” Sci. Agric., 2005, vol. 62(3), pp. 291-295, Piracicaba, Braz.
Filipsson et al., “Concise International Chemical Assessment Document 5: Limonene,” first draft, World Health Organization, Geneva, 1998, 36 pages.
Final Report on the Safety Assessment of BHT, International Journal of Toxicology, 2002, 21(Suppl. 2):19-94.
Flyvholm, “Sensitizing risk of butylated hydroxytoluene based on exposure and effect data,” Contact Dermatitis, 1990:23:341-345.
Fotherby, “Bioavailability of Orally Administered Sex Steroids Used in Oral Contraception and Hormone Replacement Therapy,” Contraception, 1996; 54:59-69.
Franklin et al., “Characterization of immunoglobulins and cytokines in human cervical mucus: influence of exogenous and endogenous hormones,” Journal of Reproductive Immunology, 1999, 42: 93-106, Elsevier.
Franz et al., “Use of Excised Human Skin to Assess the Bioequivalence of Topical Products,” Skin Pharmacol Physiol 2009, 22:276-286.
Freedman, “Menopausal hot flashes: Mechanisms, endocrinology, treatment,” J. Steroid Biochem. Mol. Biol., 2013, 6 pages, Elsevier.
Fuchs et al., “The Effects of an Estrogen and Glycolic Acid Cream on the Facial Skin of Postmenopausal Women: A Randomized Histologic Study,” Cutis. Jun. 2003, 71(6):481-8.
Fugh-Berman, Adriane, “Bioidentical Hormones for Menopausal Hormone Therapy: Variation on a Theme, Journal of General Internal Medicine,” 2007, vol. 1030-34.
Furness et al.,“Hormone therapy in postmenopausal women and risk of endometrial hyperplasia (Review),” 2012, 208 pages, The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.
Gäfvert et al., “Free radicals in antigen formation: reduction of contact allergic response to hydroperoxides by epidermal treatment with antioxidants,” British Journal of Dermatology, 2002, 146:649-656.
Ganam-Quintanar et al., “Evaluation of the transepidermal permeation of diethylene glycol monoethyl ether and skin water loss,” International Journal of Pharmaceutics, 147(2), Feb. 28, 1997, pp. 165-171 (abstract only).
Gattefossé SAS, Material Safety Data Sheet, Gelot 64, 2012, 8 pages.
Gattefossé SAS, Regulatory Data Sheet, Gelot 64, 2012, 6 pages.
Gattefossé SAS, Regulatory Data Sheet, Lauroglycol 90, 2012, 5 pages.
Gattefossé, “Excipients for Safe and Effective Topical Delivery, Drug Development and Delivery” Jul./Aug. 2012, http://drug-dev.com/Main/Back-Issues/Transdermal-Topical-Sutecutaneous-NonInvasive-Deliv-5.aspx#, 2 pages.
Gillet et al., “Induction of amenorrhea during hormone replacement therapy: optimal micronized progesterone dose,” A multicenter study, 1994, Maturitas, 19:103-115.
Giron-Forest et al., “Thermal analyis methods for pharmacopoeial materials,” J. Pharmaceutical & Biomedical Anal., 1989, vol. 7(12) pp. 1421-1433, Pergamon Press, Gr. Britain.
Giron-Forest, “Thermal analysis and calorimetric methods in the characterisation of polymorphs and solvates,” Thermochimica Acta, 1995, 248:1-59, Elsevier.
Glaser et al., “Pilot Study: Absorption and Efficacy of Multiple Hormones Delivered in a Single Cream Applied to the Mucous Membranes of the Labia and Vagina,” Gynecol Obstet Invest 2008; 66:111-118.
Golatowski et al., “Comparative evaluation of saliva collection methods for proteome analysis,” Clinica Chimica Acta, 2013, 419:42-46.
Graham et al., “Physiological Action of Progesterone in Target Tissues, Endocrine Reviews,” 1997, 18(4):502-519.
Groothuis et al., “Estrogen and the endometrium: lessons learned from gene expression profiling in rodents and human,” Human Reproduction Update, 2007, 13(4):405-417.
Gunstone et al., “Vegetable Oils in Food Technology: Composition, Properties and Uses, Blackwell Publishing,” CRC Press, 2002, 21 pages.
Gurney et al., “The Women's Health Initiative trial and related studies: 10 years later: A clinician's view,” J.Steroid Biochem. Mol. Biol., 2013, 8 pages Elsevier.
Hamid et al. “The Effects of Common Solubilizing Agents on the Intestinal Membrane Barrier Functions and Membrane Toxicity in Rats,” International Journal of Pharmaceutics, 2009 379:100-108, Elsevier.
Haner, “Crystal data (I) for some pregnenes and pregnadienes,” Acta Cryst., 1964, 17:1610.
Hapgood et al., “Potency of progestogens used in hormonal therapy: Toward understanding differential actions,” J. Steroid Biochem. Mol. Biol., 2013, Elsevier, 9 pages.
Hargrove et al., “Menopausal Hormone Replacement Therapy with Continuous Daily Oral Micronize Estradiol and Progesterone,” Obstet Gynecol, Apr. 1989, 73(4):606-612.
Hatton et al., “Safety and efficacy of a lipid emulsion containing medium-chain triglycerides,” Clinical Pharmacy, 1990, 9(5):366-371.
He et al., “Apoptotic Signaling Pathways in Uteri of Rats with Endometrial Hyperplasia Induced by Ovariectomy Combined with Estrogen,” Gynecol Obstet Invest 2013;76:51-56.
Helbling et al., “The Optimization of an Intravaginal Ring Releasing Progesterone Using a Mathematical Model,” Pharm Res, 2014, 31:795-808, Springer Science.
Helmy et al., “Estrogenic Effect of Soy Phytoestrogens on the Uterus of Ovariectomized Female Rats,” Clinic Pharmacol Biopharmaceut, 2014, S2, 7 pages.
Henderson, “Alzheimer's disease: Review of hormone therapy trials and implications for treatment and prevention after . . . ,” J. Steroid Biochem. Mol. Biol., 2013, 8 pages, Elsevier.
Henriksen et al., “An ENDOR Sturdy of Radiation-Induced Molecular Damage to Progesterone,” Jour. of Mag. Resonance, 1985, 63:333-42, Acedemic Press, Inc.
Hodis, H.N., Mack, W.J., “Hormone replacement therapy and the association with heart disease and overall mortality: Clinical . . . ,” J. Steroid Biochem. Mol. Biol., 2013, 8 pages, Elsevier.
Hospital et al., “X-ray Crystallography of Estrogens and Their Binding to Receptor Sites,” Mol. Pharmacology, 1972, 8:438-45, Acedemic Press, Inc.
Hostynek, “Predicting absorption of fragrance chemicals through human skin,” J. Soc.CosmeCt.hem.,4 6, 221-229, Jul./Aug. 1995.
Hulsmann, “Stability of Extruded 17B-Estradiol Solid Dispersions,” Pharmaceutical Development and Tech., 2001, 6(2):223-29, (9 total pages), Marcel Dekker, Inc.
Hurn et al., “Estrogen as a Neuroprotectant in Stroke,” Journal of Cerebral Blood Flow and Metabolism, 2000, 20:631-652, Lippincott Williams & Wilkins, Inc., Philadelphia.
Hyder et al., “Synthetic Estrogen 17α-Ethinyl Estradiol Induces Pattern of Uterine Gene Expression Similar to Endogenous Estrogen 17β-Estradiol,” JPET, 1999, 290(2):740-747.
Idder et al., “Physicochemical properties of Progesterone,” SciFinder, Feb. 24, 2014, pp. 1-26, American Chem. Society & US Natl. Lib. of Med.
International Search Report issued in International Application No. PCT/US12/66406, dated Jan. 24, 2013, 3 pages.
International Search Report issued in International Application No. PCT/US13/023309, dated Apr. 9, 2013, 12 pages.
International Search Report and written Opinion issued in PCT/US/13/46442, dated Nov. 1, 2013, 10 pages.
International Search Report and written Opinion issued in PCT/US/13/46443, dated Oct. 31, 2013, 11 pages.
International Search Report and written Opinion issued in PCT/US/13/46444, dated Oct. 31, 2013, 10 pages.
International Search Report and written Opinion issued in PCT/US/13/46445, dated Nov. 1, 2013, 9 pages.
Johanson, “Toxicity Review of Ethylene Glycol Monomethyl Ether and its Acetate Ester,” Critical Reviews in Toxicology, 2000, 30(3):307-345 (abstract only). http://informahealthcare.com/doi/abs/10.1080/10408440091159220.
Johnson et al., “Racemic Progesterone,” Tetrahedron Letters No. 4, 1963, pp. 193-196, Pergamon Press Ltd., Great Britain.
Joshi et al., “Detection and synthesis of a progestagen-dependent protein in human endometrium,” J Reprod Fert, 1980, 59:273-285.
Kanno et al., “The OECD Program to Validate the Rat Uterotrophic Bioassay to Screen Compounds for in Vivo Estrogenic Responses: Phase 1,” Environmental Health Perspectives, Aug. 2001, 109(8):785-794.
Karlberg et al., “Air oxidation of d-limonene (the citrus solvent) creates potent allergens,” Contact Dermatitis, 1992:26:332-340.
Karlberg et al., “Influence of an anti-oxidant on the formation of allergenic compounds during auto-oxication of d-limonene,” Ann. Occup. Hyg., 1994, 38(2):199-207.
Kaunitz, “Extended duration use of menopausal hormone therapy,” Menopause: The Journal of the North American Menopause Society, 2014, 21(6):1-3.
Khalil, “Stability and Dissolution Rates of Corticosteroids in Polyethylene Glycol Solid Dispersions,” Drug Dev. & Indus. Pharm., 1984, 10(5):771-87, Marcel Dekker.
Kharode et al., “The Pairing of a Selective Estrogen Receptor Modulator, Bazedoxifene, with Conjugated Estrogens as a New Paradigm for the Treatment of Menopausal Symptoms and Osteoporosis Prevention,” Endocrinology, 2008, 149(12):6084-6091.
Kim et al., “Safety Evaluation and Risk Assessment of d-Limonene,” Journal of Toxicology and Environmental Health, Part B: Critical Reviews, 2013, 16:1, pp. 17-38 http://dx.doi.org/10.1080/10937404.2013.769418.
Kincl et al., “Increasing Oral Bioavailability of Progesterone by Formulation,” Journal of Steroid Biochemistry, 1978, 9:83-84.
Knuth et al., “Hydrogel delivery systems for vaginal and oral applications: Formulation and biological considerations,” Advanced Drug Delivery Reviews, Jul.-Aug. 1993, 11(1-2):137-167 (abstract only).
Koga et al., “Enhancing mechanism of Labrasol on intestinal membrane permeability of the hydrophilic drug gentamicin sulfate,” European Journal of Pharmaceutics and Biopharmaceutics, 2006, 64:82-91.
Komm et al., “Bazedoxifene Acetate: A Selective Estrogen Receptor Modulator with Improved Selectivity,” Endocrinology, 2005, 146(9):3999-4008.
Korkmaz, “Byophysical Studies of Progesterone-Model Membrane Interactions,” Thesis, Grad. School of Nat. and App. Sci. of the Middle East Tech. University, Sep. 2003, 143 pages.
Kotiyan, “Stability indicating HPTLC method for the estimation of estradiol,” Journal of Pharmaceutical and Biomedical Analysis, 2000, 22: 667-671, Elsevier.
Krzyminiewski et al., “EPR Study of the Stable Radical in a y-Irradiated Single Crystal of Progesterone,” Jour. of Mag. Resonance, 1982, 46:300-05, Acedemic Press.
Kubli-Garfias et al., “Ab initio calculations of the electronic structure of glucocorticoids,” Jour. of Mol. Structure, Theochem, 1998, 454:267-75, Elsevier.
Kubli-Garfias, “Ab initio study of the electronic structure of progesterone and related progestins, Jour. of Mol. Structure,” Theochem, 1998, 425:171-79, Elsevier.
Kuhnert-Brandstaetter and Kofler, Zur Unterscheidung von losungsmittelhaltigen pseudopolymorphen Kristallformen und polymorphen Modiflkationen bei Steroidhormonen.II. vol. 1 pp. 127-139, 1968, Mikrochimica Acta.
Kuhnert-Brandstaetter, M & Lnder, R, Zur Hydratbildung bei Steroidhormonen, Sci. Pharm., vol. 41(2) pp. 109-116, 1973.
Kuhnert-Brandstatter, “Thermo-microscopic and spectrophotometric: Determination of steroid hormones,” Microchemical Journal, 1965, 9:105-33.
Kumasaka et al., “Effects of Various Forms of Progestin on the the Estrogen-Primed, Ovariectomized Rat,” Endocrine Journal, 1994, 41(2):161-169.
Kuon et al., “A Novel Optical Method to Assess Cervical Changes during Pregnancy and Use to Evaluate the Effects of Progestins on Term and Preterm Labor,” Am J Obstet Gynecol. Jul. 2011, 205(1): 82.e15-82.e20.
Kuon et al., “Actions of progestins for the inhibition of cervical ripening and uterine contractions to prevent preterm birth,” FVV in Obgyn, 2012, 4 (2):110-119.
Kuon et al., “Pharmacological actions of progestins to inhibit cervical ripening and prevent delivery depend upon their properties, the route of administration and the vehicle,” Am J Obstet Gynecol. May 2010, 202(5):455.e1-455.e9.
Labrie et al., “Intravaginal prasterone (DHEA) provides local action without clinically significant changes in serum concentrations of estrogens or androgens,” Journal of Steroid Biochemistry & Molecular Biology, 2013, 38:359-67, Elsevier.
Lacey, “The WHI ten year's later: An epidemiologist's view,” J. Steroid Biochem. Mol. Biol., 2013, 4 pages, Elsevier.
Lahiani-Skiba, “Solubility and Dissolution Rate of Progesterone-Cyclodextrin . . . , Drug Development and Industrial Pharmacy,” Informa Healthcare, 2006, 32:1043-1058.
Lancaster et al., “The Polymorphism of Progesterone: Stabilization of a ‘Disappearing’ Polymorph by . . . ,” Jour. of Pharm. Sci., 2007, 96(12):3419-31, Wiley-Liss.
Land, “The influence of water content of triglyceride oils on the solubility of steriods,” Pharmaceutical Research, May 2005, 22(5) Springer Science+Business Media, pp. 784-788.
Lauer et al., “Evaluation of the hairless rat as a model for in vivo percutaneous absorption,” Journal of Pharmaceutical Sciences, Jan. 1997, 86(1):13-18.
Leonetti et al., “Transdermal progesterone cream as an alternative progestin in hormone therapy,” Alternative Therapies, Nov./Dec. 2005, 11(6):36-38.
Leonetti, “Topical progesterone cream has an antiproliferative effect on estrogen-stimulated endometrium,” Fertility and Sterility, Jan. 2003, 79(1):221-222.
Lewis et al., “Caution on the use of saliva measurements to monitor absorption of progesterone from transdermal creams in postmenopausal women,” Maturitas, The European Menopause Journal, 2002, 41:1-6.
Li, “Solid-state NMR analysis of steroidal conformation of 17a- and 17B-estradiol in the absence and presence of lipi . . . ,” Steroids, Elsevier, 2012, 77:185-92.
Lobo, Foreword, J. Steroid Biochem. Mol. Biol., 2014, 1 page, Elsevier.
López-Belmonte, “Corrigendum to Comparative uterine effects on ovariectomized rats after repeated treatment with different vaginal estrogen formulations,” [Maturitas 72 (2012) 353-358], Maturitas 74, 2013, p. 393, Elsevier.
Lucy et al., “Gonadotropin-releasing hormone at estrus: lutenizing hormone, estradiol, and progesterone during, . . . ,” Biol Reprod, Sep. 1986;35(2):300-311 (abstract only).
LVova, “Thermal Analysis in the Quality Control and Standardization of Some Drugs,” J Thermal Anal., 1993, 40:405-11, Wiley.
Madishetti et al., “Development of domperidone bilayered matrix type transdermal patches physicochemical,” in vitro and ex vivo characterization, DARU, 2010, 18(3):221-229.
Magness et al., “Estrone, Estradiol-17β and Progesterone Concentrations in Uterine Lymph and Systematic Blood throughout the Porcine Estrone Estrous Cycle,” Journal of Animal Science, 1983, 57:449-55, ISU.
Mcguffy, “Softgel Technology as a Lipid-Based Delivery Tool for Bioavailability Enhancement,” Catalent Pharma Solutions, Somerset, NJ, Mar. 2011, 35 pages.
Estradiol, The Merck Index Online, Royal Society of Chemistry, https://www.rsc.org/Merck-Index/monograph/mono1500003758/estradiol?q=unauthorize, 2013, 2 pages.
Progesterone, The Merck Index Online, Royal Society of Chemistry, 2013, search Feb. 17, 2014 https://www.rsc.org/Merck-Index/monograph/print/mono1500007889/progesterone?q=authorize, 2 pages.
Mesley, “Clathrate Formation from Steroids,” Chemistry and Industry, Sep. 1965, 37:1594-95.
Miao et al., “Chemical Properties of Progesterone,” SciFinder, 2014, American Chemical Society & US Natl. Lib. of Med., 36 pages.
Miles et al., “Pharmacokinetics and endometrial tissue levels of progesterone after administration bv'Intramuscular and vaginal routes: a comparative study,” Fertility and Sterility, Sep. 1994, 62(3):485-490.
Miller et al., “Safety and Feasibility of Topical Application of Limonene as a Massage Oil to the Breast,” Journal of Cancer Therapy, 2012, 3:749-754.
Mueck et al., “Genomic and non-genomic actions of progestagens in the breast,” J. Steroid Biochem. Mol.Biol, 2013, Elsevier, 6 pages.
Muramatsu, “Thermodynamic Relationship between a- and B—Forms of Crystalline Progesterone,” J. Pharmaceutical Sciences, 1979, 68(2):175-76, Amer. Pharm. Assoc.
Ng et al., “Advances in biodiesel fuel for application in compression ignition engines,” Clean Techn Environ Policy, 2010, 12:459-93, Springer-Verlag.
Nicklas, “Preparation and characterization of marine sponge collagen nanoparticles and employment for the trans . . . ,” Drug Devel. & Indust. Pharmacy, 2009, 35(9):1035-1042.
Nilsson et al., “Analysis of Contact Allergenic Compounds in Oxidized d-Limonene,” Chromatographia, Feb. 1996, 42(3/4):199-205.
Notelovitz et al., “Initial 17-b-Estradiol Dose for Treating Vasomotor Symptoms, Obstetrics & Gynecology,” vol. 95(5):726-31, part 1, May 2000, Elsevier.
NuGen, What is NuGen HP Hair Growth System, accessed from the Internet on Mar. 7, 2013, copyright 1997-2013, 3 pages.
NuGest900, NuGest 900™, accessed Mar. 5, 2013, copyright 1998-2009, 4 pages.
O'leary, “Salivary, but not serum or urinary levels of progesterone are elevated after topical application of pregersterone cream to pre-and post-menopausal women,” Clinical Endocrinology, 2000, 53: 615-20, Blackwell Science (abstract only).
Open Notebook, Science Solubility Challenge, Jul. 16, 2013, Solubility of progesterone in organic solvents, http://lxsrv7.oru.edu/˜alang/onsc/solubility/allsolvents.php?solute=progesterone, 1 page.
Opinion on the Diethylene Glycol Momoethyl Ether (DEGEE), Scientific Committee on Consumer Products, Dec. 19, 2006, 27 pages.
Outterson, “The Drug Quality and Security Act—Mind the Gaps,” NEngl J med , Jan. 9, 2014, 370(2):97-99, nejm.org.
Palamakula et al., “Preparation and In Vitro Characterization of Self-Nanoemulsified Drug Delivery Systems of Coenzyme Q10 Using Chiral Essential Oil Components,” Pharmaceutical Technology Oct. 2004, pp. 74-88.
Panay et al., “The 2013 British Menopause Society & Women's Health Concern recommendations on hormone replacement therapy,” Menopause International: The Integrated Journal of Postreproductive Health, published online May 23, 2013, Sage Publications. http://min.sagepub.com/content/early/2013/05/23/1754045313489645.1, 11 pages.
Panchangnula et al., “Development and evaluation of an intracutaneous depot formulation of corticosteroids using Transcutol . . . ,” J Pharm Pharmacol. Sep. 1991, 43(9):609-614 (abstract only).
Parasuraman et al., “Blood sample collection in small laboratory animals,” Journal of Pharmacology & Pharmacotherapeutics, Jul.-Dec. 2010, 1(2):87-93.
Park, “Solvent effects on physicochemical behavior of estradiols recrystalized for transdermal delivery,” Arch Pharm Res, 2008, 31(1): 111-16.
Park, “Use of CP/MAS solid-state NMR for the characterization of solvate . . . ,” European Journal of Pharmaceutics and Biopharmaceutics, 2005, 60:407-12.
Parrish, “A new estra—1,3,5(10)-triene-3,17b-diol solvate: estradiol-methanol-water, Crystal Structure Comm.,” Intn'l Union of Crystallography, ISSN 0108-2701, 2003, pp. o80-o82.
Patel et al., “Transdermal Drug Delivery System: A Review,” www.thepharmajournal.com, vol. 1, No. 4, 2012, pp. 78-87.
Payne et al., “Examples of successful crystal structure prediction: polymorphs of primidone and progesterone,” Intl. Jourl of Pharma., 1999, 177:231-45, Elsevier.
PCCA, Apothogram, PCCA, May 2014, 14 pages, Houston, TX.
Persson et al., “Physicochemical Properties of Progesterone Selecte,” SciFinder, pp. 1-5, Feb. 24, 2014, American Chem. Society & US Natl. Lib. of Med.
Pfaus et al., “Selective facilitation of sexual solicitation in the female rat by a melanocortin receptor agonist,” PNAS, Jul. 6, 2004, 101(27):10201-10204.
Pheasant, “Polymorphism of 17-Ethinylestradiol, Schering Corporation,” Bloomfield, NJ, May 1950, pp. 4303-4304.
Pickles, “Cutaneous reactions to injection of progesterone solutions into the skin,” Br Med Journal, Aug. 16, 1952, pp. 373-374.
Pinkerton et al., “What are the concerns about custom-compounded “bioidentical” hormone therapy?” Menopause: The Journal of the North American Menopause Society, 2014, 21(12):1-3.
Pinkerton, J.V., Thomas, S., “Use of SERMs for treatment in postmenopausal women,” J. Steroid Biochem. Mol. Biol., 2014, 13 pages Elsevier.
Pisegna, “A High-pressure Vibrational Spectroscopic Study of Polymorphism in Steroids . . . ,” Thesis, McGill University, Dept. of Chem, Nov. 1999, Natl. Lib. of Canada, 134 pages.
Position Statement, Management of symptomatic vulvovaginal atrophy: 2013 position statement of the North American Menopause Society (NAMS), Menopause, 2013, 20(9):888-902.
Practice Bulletin No. 141, Management of Menopausal Symptoms, Obstetrics & Gynecology, Acog, Jan. 2014, 123(1): 202-216.
Prajapati et al., “A comparative Evaluation of Mono-, Di- and Triglyceride of Medium Chain Fatty Acids by Lipid/Surfactant/Water Phase Diagram, Solubility Determination and Dispersion Testing for Application in Pharmaceutical Dosage Form Development,” Springerlink.com, Apr. 2011, 21 pages.
Prausnitz et al., “Transdermal drug delivery,” Nat Biotechnol. Nov. 2008 ; 26(11), 18 pages.
Price, “The computational prediction of pharmaceutical crystal structures and polymorphism,” Adv. Drug Delivery Reviews, 2004, 56:301-19, Elsevier.
Product Information Sheet, Body Balance Cream, Tahitian Noni International, 2013, 1 page.
Product Safety Assessment: Diethylene Glycol Monoethyl Ether, Created: Sep. 24, 2007 the Dow Chemical Company Page, 5 pages.
Progynova TS 100, available online at file:///C:/Users/Call%20Family/Desktop/Progynova%/20TS%20100%2012%20Patches_Pack%20%28Estradiol%20Hemihydrate%29.html, 2010, 6 pages.
Provider Data Sheet, About Dried Blood Spot Testing, ZRT Laboratory, 2014, 3 pages.
Rahn et al., “Vaginal Estrogen for Genitourinary Syndrome of Menopause a Systematic Review,” Obstet Gynecol, 2014, 124(6):1147-56.
Reisman et al., “Topical Application of the Synthetic Triterpenoid RTA 408 Protects Mice from Radiation-Induced Dermatitis,” Radiation Researc, 2014, 181:512-520.
Rosilio et al., “Physical Aging of Progesterone-Loaded Poly(D,L,-lactide-co-glycolide) Microspheres,” Pharmaceutical Research, 1998, 15(5):794-99, Plenum Pub. Corp.
Ross et al., “Randomized, double-blind, dose-ranging study of the endometrial effects of a vaginal progesterone gel in estrogen-treated postmenopausal women,” AnnJ Obstet Gynecol, Oct. 1997, 177(4):937-941.
Ruan et al., “Systemic progesterone therapy—Oral, vaginal, injections and even transdermal?” Maturitas, 2014, 79:248-255, Elsevier.
Salem, “Sustained-release progesterone nanosuspension following intramuscular injection in ovariectomized rats,” International Journal of Nanomedicine 2010, 5:943-954, Dove Press.
Salole, “Estradiol, Analytical Profiles of Drug Substances,” 1986, 15:283-318.
Salole, “The physicochemical properties of oestradiol, Journal of Pharmaceutical & Biomedical Analysis,” 1987, 5(7):635-648.
Santen, “Menopausal hormone therapy and breast cancer,” J. Steroid Biochem. Mol. Biol., 2013, Elsevier, 10 pages.
Santen, “Vaginal administration of estradiol: effects of dose, preparation and timing on plasma estradiol levels,” CLIMACTERIC, 2014; 17:1-14.
Sarkar et al., “Chemical Stability of Progesterone in Compounded Topical Preparations using PLO Transdermal Cream™ and HRT Cream™ Base . . . ,” J Steroids Horn Sci, 2013, 4:2, 3 pages.
Sarrel et al., “The Mortality Toll of Estrogen Avoidance: An Analysis of Excess Deaths Among Hysterectomized Women Aged 50 to 59 Years,” American Journal of Public Health, Research and Practice, Published online ahead of print Jul. 18, 2013, pp. e1-e6.
Satyanarayana et al., “Aqueous Solubility Predictions of Aliphatic Alcohols, Alkyl Substituted Benzoates and Steroids,” Asian J. Chem., 1997, 9(3):418-26.
Scavarelli et al., “Progesterone and Hydrate or Solvate,” SciFinder, pp. 1-2, Feb. 24, 2014, American Chem. Society.
Schindler, “The “newer” progestogens and postmenopausal hormone therapy (HRT),” J. Steroid Biochem.Mol. Biol., 2013, Elsevier, 4 pages.
Schutte et al., “A tissue engineered human endometrial stroma that responds to cues for secretory differentiation, decidualization and menstruation,” Fertil Steril, Apr. 2012 ; 97(4):997-1003, Elsevier.
Schweikart et al., “Comparative Uterotrophic Effects of Endoxifen and Tamoxifen in Ovariectomized Sprague-Dawley Rats, Toxicologic Pathology,” 2014, 42: 1188-1196.
SciFinder Scholar Prednisone Chemical Properties, SciFinder, 2014, pp. 1-7, National Library of Medicine.
SciFinder Scholar Prednisone Physical Properties, SciFinder, 2014, pp. 1-10, Natioinal Library of Medicine.
SciFinder Scholar Progesterone Experimental Properties, SciFinder, pp. 1-9, Feb. 24, 2014, American Chem. Society.
Serantoni et al., “4-Pregnen-3,20-dione (progesterone, form II),” Crystal Structure Comm., 1975, 4(1):189-92, CAPLUS Database (abstract only).
Shao et al., “REVIEW Open Access Direct effects of metformin in the endometrium: a hypothetical mechanism for the treatment of women with PCOS and endometrial carcinoma,” Journal of Experimental & Clinical Cancer Research, 2014, 33(1):41, 11 pages.
Sharma et al., “Physical Properties of Progesterone Selected Refer,” SciFinder, pp. 1-5, Feb. 24, 2014, American Chern. Society & US Natl. Lib. of Med.
Shrier et al., “Mucosal Immunity of the Adolescent Female Genital Tract,” Journal of Adolescent Health, 2003, 32:183-186.
Shufelt et al., “Hormone therapy dose, formulation, route delivery, and risk of cardiovascular events in women: findings from the Women's Health Initiative Observational Study,” Menopause: The Journal of The North American Menopause Society, 2014, 21(3): 1-7.
Siew, “Adeline, moderator, Bioavailability Enhancement with Lipid-Based Drug-Delivery Systems,” Pharmaceutical Technology, Aug. 2014, pp. 28, 30-31.
Sigma-Aldrich, Progesterone-Water Soluble: powder, BioReagent, suitable for cell culture), MSDS available online: hitp://www.sigmaaldrich.com/catalog/product/sigma/p7556,, 2015, 1 page.
Simon et al., “Effective Treatment of Vaginal atrophy with an Ultra-low-dose estradiol vaginal tablet,” Obstetrics & Gynocology, Nov. 2008, 112(5):1053-1060.
Simon, “What if the Women's Health Initiative had used transdermal estradiol and oral progesterone instead?” Menopause: The Journal of The North American Menopause Society, 2014, 21(7):1-15.
Sitruk-Ware et al., “Progestogens in hormonal replacement therapy: new molecules, risks, and benefits,” Menopause: The Journal of The North American Menopause Society. 2002, 9(1):6-15, 2002.
Sitruk-Ware, “Oral Micronized Progesterone—Bioavailability pharmacokinetics, pharmacological and therapeutic implications—A review,” Contraception, Oct. 1987, 36(4):373-402.
Smith et al., “Lower Risk of Cardiovascular Events in Postmenopausal Women Taking Oral Estradiol Compared with Oral Conjugated Equine Estrogens,” JAMA Internal Medicine, Published online Sep. 30, 2013, E1-E7. jamainternalmedicine.com.
Smyth et al., “Summary of Toxicological Data, A 2-Yr Study of Diethylene Glycol Monoethyl Ether in Rats,” Fd Cosmet. Toxicol., 1964, vol. 2, pp. 641-642.
Stanczyk et al., “Therapeutically equivalent pharmacokinetic profile across three application sistes for AG200-15, a novel low-estrogen dose contraceptive patch,” Contraception, 2013, 87:744-749.
Stanczyk et al., Ethinyl estradiol and 17β-estradiol in combined oral contraceptives pharmacokinetics, pharmacodynamics and risk assessment, Contraception, Jun. 2013, 87(6):706-727.
Stanczyk, F.Z., Bhavnani, B.R., “Current views of hormone therapy for the management and treatment of postmenopausal women,” J. Steroid Biochem. Mol. Biol., 2014, Elsevier, 3 pages.
Stein et al., Progesterone Physical Properties, SciFinder, pp. 1-46, Feb. 24, 2014, American Chem. Society & US Natl. Lib. of Med.
Strickley, “Solubilizing excipients in oral and injectable formulations,” Pharmaceutical Research Feb. 2004, 21 (2):201-230 (abstract only).
Strocchi, “Fatty Acid Composition, and Triglyceride Structure of Corn Oil, Hydrogenated Corn Oil, and Corn Oil Margarine,” Journal of Food Science, 1981, 47:36-9.
Struhar et al., “Estradiol Benzoate: Preparation of an injection suspension . . . ,” SciFinder, Cesko-Slovenska Farmacie, 1978, 27(6):245-9, Bratislava, Czech (abstract only).
Sullivan et al., “A review of the nonclinical safety of Transcutol®, a highly purified form of diethylene glycol monoethyl ether (DEGEE) used as a pharmaceutical excipient,” Food and Chemical Toxicology, 2014, 72:40-50.
Sun, D-Limonene: Safety and Clinical Applications, Alternative Medicine Review, 2007, 12(3):259-264.
Tait, “Characterization of the Prod. from the Oxidation of Progesterone with Osmium Tetroxide,” Dept of Investigative Med., Univ. Cambridge, Gt. Britain, 1972, pp. 531-542.
Takacs M. et al., “The light sensitivity of corticosteroids in crystalline form,” Pharmaceutica acta Helvetiae, vol. 66 (5-6) pp. 137-140, 1991, Hardin Library.
Tan et al., “A Sensitive Method for the Determination of Progesterone in Human Plasma by LC-MS-MS,” M1025, Cedra Corporation, Austin, Texas (publicly available before application filing date of Oct. 22, 2014), 1 page.
Tang et al., “Effect of Estrogen and Progesterone on the Development of Endometrial Hyperplasia in the Fischer Rat,” Biology of Reproduction, 1984, 31:399-413.
Tas et al., “Comparison of antiproliferative effects of metformine and progesterone on estrogen-induced endometrial hyperplasia in rats,” Gynecol Endocrinol, Early Online: pp. 1-4, 2013. http://informahealthcare.com/gye.
Telia, S.H., Gallagher, J.C., “Prevention and treatment of postmenopausal osteoporosis,” J. Steroid Biochem. Mol. Biol., 2013, Elsevier, 16 pages.
Thomas et al., The effect of water solubility of solutes on their flux through human skin in vitro: An . . . , Intl. J. of Pharmaceut., 2007, vol. 339 pp. 157-167, Elsevier.
Thomas, “Characteristics of membrane progestin receptor alpha (mPRα) and progesterone membrane receptor component 1 (PGMRC1) and their roles in mediating rapid progestin actions,” Frontiers in Neuroendocrinology, 2008, 29:292-312.
Tripathi et al., “Study of Polymorphs of Progesterone by Novel Melt Sonocrystallization Technique: A Technical Note,” AAPS PhamSciTech, Sep. 2010, 11(3):1493-1498.
Trommer et al., “Overcoming the stratum Comeum: The modulation of Skin Penetration,” Skin Pharmacol Physiol, 2006, 19:106-121.
Tuleu et al., “Comparative Bioavailability Study in Dogs of a Self-Emulsifying Formulation of Progesterone Presented in a Pellet and Liquid Form Compared with an Aqueous Suspension of Progesterone,” Journal of Pharmaceutical Sciences, Jun. 2004, 93(6):1495-1502.
Ueda et al., “Topical and Transdermal Drug Products,” Pharmacopeial Forum, vol. 35(3), May-Jun. 2009, 750-764.
USP, 401 Fats and Fixed Oils, Chemical Tests, Second Suplement to USP36-NF 31, pp. 6141-6151, 2013.
USP, Lauroyl Polyoxylglycerides, Safety Data Sheet, US, 5611 Version #02, pp. 1-9, 2013.
USP, Official Monographs, Corn Oil, NF 31, pp. 1970-1971, Dec. 2013.
USP, Official Monographs, Lauroyl Polyoxylglycerides, NF 31, pp. 2064-2066, Dec. 2013.
USP, Official Monographs, Medium Chain Triglycerides, NF 31, pp. 2271-2272, Dec. 2013.
USP, Official Monographs, Mono- and Di-glycerides, NF 31, p. 2101, Dec. 2013.
USP, USP Certificate-Corn Oil, Lot G0L404, Jul. 2013, 2 pages.
USP Monographs: Progesterone, USP29, www.pharmacopeia.cn/v29240/usp29nf24s0_m69870.html, search done: Feb. 25, 2014, 2 pages.
USPTO, Final Office Action issued in U.S. Appl. No. 13/684,002, dated Jul. 16, 2013, 13 pages.
USPTO, Non-Final Office Action issued in U.S. Appl. No. 13/684,002, dated Mar. 20, 2013, 14 pages.
USPTO, Notice of Allowance issued in U.S. Appl. No. 13/684,002, dated Dec. 6, 2013, 7 pages.
USPTO, Non-Final Office Action issued in U.S. Appl. No. 14/099,545, dated Feb. 18, 2014, 7 pages.
USPTO, Restriction/Election Requirement issued in U.S. Appl. No. 14/099,562, dated Feb. 20, 2014, 6 pages.
USPTO, Restriction/Election Requirement issued in U.S. Appl. No. 14/099,623, dated Mar. 5, 2014, 9 pages.
USPTO, Office Action issued in U.S. Appl. No. 12/561,515, dated Dec. 12, 2011, 14 pages.
USPTO, Final Office Action issued in U.S. Appl. No. 12/561,515, dated Oct. 26, 2013, 14 pages.
USPTO, Advisory Action issued in U.S. Appl. No. 12/561,515, dated Jan. 29, 2013, 3 pages.
USPTO, Notice of Allowance issued in U.S. Appl. No. 12/561,515, dated Sep. 11, 2013, 12 pages.
Utian et al., “Relief of vasomotor symptoms and vaginal atrophy with lower doses of conjugated equine estrogens,” Fertility and Sterility, Jun. 2001, 75(6), 15 pages.
Voegtline et al., “Dispatches from the interface of salivary bioscience and neonatal research,” Frontiers in Endocrinology, Mar. 2014, vol. 5, article 25, 8 pages.
Waddell et al., “Distribution and metabolism of topically applied progesterone in a rat model,” Journal of Steroid Biochemistry & Molecular Biology, 2002, 80:449-455.
Waddell et al., “The Metabolic Clearance of Progesterone in the Pregnant Rat: Absence of a Physiological Role for the Lung,” Biology of Reproduction, 1989, 40:1188-1193.
Walter et al., “The role of progesterone in endometrial angiogenesis in pregnant and ovariectomised mice,” Reproduction, 2005, 129:765-777.
Weber, “Corn Lipids,” Cereal Chem.,Sep.-Oct. 1978, 55(5):572-584, The American Assoc of Cereal Chem.
Weber, et al., “Cognition and mood in perimenopause: A systematic review and meta-analysis,” J. Steroid Biochem. Mol. Biol., 2013, 9 pages, Elsevier.
Whitehead et al., “Absorption and metabolism of oral progesterone,” The British Medical Journal, vol. 280, No. 6217 (Mar. 22, 1980), pp. 825-827, BMJ Publishing Group.
Wiranidchapong, “Method of preparation does not affect the miscibility between steroid hormone and polymethacrylate,” Thermochimica Acta 485, Elsevier, 2009, p. 57.
Wood et al., “Effects of estradiol with micronized progesterone or medroxyprogesterone acetate on risk markers for breast cancer in postmenopausal monkeys,” Breast Cancer Res Treat, 2007, 101:125-134.
Wren et al., “Effect of sequential transdermal progesterone cream on endometrium, bleeding pattern, and plasma progesterone and salivary progesterone levels in postmenopausal women,” Climacteric, 2000, 3(3), pp. 155-160. http://dx.doi.org/10.1080/13697130008500109.
Wu et al., “Gene Expression Profiling of the Effects of Castration and Estrogen Treatment in the Rat Uterus,” Biology of Reproduction, 2003, 69:1308-1317.
Yalkowsky, Samuel H, & Valvani, Shri C, “Solubility and Partitioning I: Solubility of Nonelectrolytes in Water,” J. of Pharmaceutical Sciences, 1980, 69(8):912-22.
Yalkowsky, Handbook of Acqueous Solubility Data, Solutions, pp. 1110-1111, CRC Press, Boca Raton, London, New York, Wash. D.C., Jan. 2003.
Yue, “Genotoxic metabolites of estradiol in breast: potential mechanism of estradiol induced carcinogenesis,” Journal of Steroid Biochem & Mol Biology, 2003, 86:477-86.
Zava et al.,“Percutaneous absorption of progesterone,” Maturitas, 2014, 77:91-92, Elsevier.
Zava, “Topical Progesterone Delivery and Levels in Serum, Saliva, Capillary Blood, and Tissues,” Script, ZRT Laboratory, pp. 4-5. (publicly available before application filing date of Oct. 22, 2014).
Geelen, Math J.H. et al., “Dietary medium-chain fatty acids raise and (n-3) polyunsaturated fatty acids lower hepatic triacylglycerol synthesis in rats,” The Journal of Nutrition, 1995, 125(10):2449-2456.
Herman, Anna et al., “Essential oils and their constituents as skin penetration enhancer for transdermal drug delivery: a review,” 2014 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, pp. 1-13.
Manson, JoAnn E. et al., “Menopausal hormone therapy and health outcomes during the intervention and extended poststopping phases of the women's health initiative randomized trials,” JAMA, Oct. 2, 2013, vol. 310, No. 13, pp. 1353-1368.
Portman, David et al., One-year treatment persistence with local estrogen therapy in postmenopausal women diagnosed as having vaginal atrophy, Menopause, vol. 22, No. 11, 2015, pp. 000/000 (8 pages).
Rao, Rajeswara et al., “Intra Subject Variability of Progesterone 200 mg Soft Capsules in Indian Healthy Adult Postmenopausal Female Subjects under Fasting Conditions,” J Bioequiv Availab. 2014, 6: 139-143.
Schindler, Aldof E. et al., Classification and pharmacology of progestins, Maturitas 46S1 (2003) S7-S16.
Sitruk-Ware, Regine, “Pharmacological profile of progestins,” Maturitas 47 (2004) 277-283.
Stanczyk, F.Z. et al., “Percutaneous administration of progesterone: blood levels and endometrial protection,” Menopause: The Journal of The North American Menopause Society, 2005, vol. 12, No. 2, pp. 232-237.
Stanczyk, F.Z., “All progestins are not created equal,” Steroids 68 (2003) 879-880.
Stanczyk, F.Z., “Treatment of postmenopausal women with topical progesterone creams and gels: are they effective?” Climacteric 2014; 17(Suppl 2):8-11.
Stephenson et al., “Transdermal progesterone: Effects on Menopausal symptoms and on thrombotic, anticoagulant, and inflammatory factors in postmenopausal women,” Int J Pharmaceutical Compounding, vol. 12, No. 4, Jul./Aug. 2008, pp. 295-304.
U.S. Appl. No. 13/843,362_Mar. 16, 2015_Restriction_Requirement.
U.S. Appl. No. 13/843,428_Apr. 14, 2015_Restriction_Requirement.
U.S. Appl. No. 14/099,545_Jul. 14, 2014_Notice_of_Allowance.
U.S. Appl. No. 14/099,562_Mar. 27, 2014_Non-Final_Office_Action.
U.S. Appl. No. 14/099,562_Jul. 2, 2014_Final_Office_Action.
U.S. Appl. No. 14/099,562_Dec. 10, 2014_Notice_of_Allowance.
U.S. Appl. No. 14/099,571_Mar. 28, 2014_Restriction_Requirement.
U.S. Appl. No. 14/099,571_Jul. 15, 2014_Notice_of_Allowance.
U.S. Appl. No. 14/099,582_Apr. 29, 2014_Restriction_Requirement.
U.S. Appl. No. 14/099,582_Jun. 17, 2014_Non-Final_Office_Action.
U.S. Appl. No. 14/099,582_Nov. 7, 2014_Notice_of_Allowance.
U.S. Appl. No. 14/099,582_Jan. 22, 2015_Notice_of_Allowance.
U.S. Appl. No. 14/099,598_May 13, 2014_Restriction_Requirement.
U.S. Appl. No. 14/099,598_Jul. 3, 2014_Non-Final_Office_Action.
U.S. Appl. No. 14/099,598_Dec. 10, 2014_Notice_of_Allowance.
U.S. Appl. No. 14/099,612_Mar. 20, 2014_Restriction_Requirement.
U.S. Appl. No. 14/099,612_Oct. 30, 2014_Non-Final_Office_Action.
U.S. Appl. No. 14/099,612_Nov. 26, 2014_Notice_of_Allowance.
U.S. Appl. No. 14/099,623_Jul. 18, 2014_Non-Final_Office_Action.
U.S. Appl. No. 14/099,623_Dec. 15, 2014_Notice_of_Allowance.
U.S. Appl. No. 14/103,355_Dec. 8, 2014_Non-Final_Office_Action.
U.S. Appl. No. 14/106,655_Jul. 3, 2014_Restriction_Requirement.
U.S. Appl. No. 14/125,554_Dec. 5, 2014_Restriction_Requirement.
U.S. Appl. No. 14/125,554_Apr. 14, 2015_Non-Final_Office_Action.
U.S. Appl. No. 14/136,048_Nov. 4, 2014_Restriction_Requirement.
U.S. Appl. No. 14/136,048_Mar. 12, 2015_Non-Final_Office_Action.
U.S. Appl. No. 14/475,814_Oct. 1, 2014_Non-Final_Office_Action.
U.S. Appl. No. 14/475,814_Feb. 13, 2015_Notice_of_Allowance.
U.S. Appl. No. 14/475,864_Feb. 11, 2014_Notice_of_Allowance.
U.S. Appl. No. 14/475,864_Oct. 2, 2014_Non-Final_Office_Action.
U.S. Appl. No. 14/476,040_Mar. 26, 2014_Restriction_Requirement.
U.S. Appl. No. 14/521,230_Dec. 12, 2014_Restriction_Requirement.
U.S. Appl. No. 14/521,230_Feb. 18, 2015_Non-Final_Office_Action.
U.S. Appl. No. 14/624,051_Apr. 7, 2015_Non-Final_Office_Action.
Weintraub, Arlene, “Women fooled by untested hormones from compounding pharmacies,” Forbes, Feb. 20, 2015; retrieved online at http://onforb.es/1LIUm1V on Feb. 23, 2015, 3 pages.
Castelo-Branco Camil et al., “Treatment of atrophic vaginitis,” Therapy, 2007, vol. 4, No. 3, pp. 349-353.
Chambin et al., Interest of Multifunctional Lipid Excipients: Case of Gelucire® 44/14, Drug Development and Industrial Pharmacy, vol. 31, No. 6, pp. 527-534 (Year: 2005).
Cho, Y.A. et al., Transdermal Delivery of Ketorolac Tromethamine: Effects of Vehicles and Penetration Enhancers, Drug Development and Industrial Pharmacy, 30(6):557-564, Jun. 2004.
Cicinelli et al., “First uterine pass effect” is observed when estradiol is placed in the upper but not lower third of the vagina, Fertility and Sterility, vol. 81, No. 5, May 2004, pp. 1414-1416.
Cicinelli, Intravaginal oestrogen and progestin administration: advantages and disadvantages, Best Practices & Research Clinical Obstretrics and Gynaecology vol. 22, No. 2, 2008, pp. 391-405.
Crandall, Carolyn, “Vaginal Estrogen Preparations: A Review of Safety and Efficacy for Vaginal Atrophy,” Journal of Women's Health, 2002, vol. 11, No. 10, pp. 857-877.
Garad S. et al., “Preclinical Development for Suspensions,” A.K. Kulshreshtha et al. (eds), Pharmaceutical Suspensions: From Formulation Development to Manufacturing, Springer, New York 2010, pp. 127-176.
Holm et al., “Examination of oral absorption and lymphatic transport of halofantrine in a triple-cannulated canine model after administration in self-microemulsifying drug delivery systems (SMEDDS) containing structured triglycerides,” European Journal of Pharmaceutical Sciences 20 (2003) 91-97.
Humberstone, Andrew et al., “Lipid-based vehicles for the oral delivery of poorly water soluble drugs,” Advanced Drug Delivery Reviews, 25 (1997) 103-128.
Karande, et al., Enhancement of transdermal drug delivery via synergistic action of chemicals, Biochimica et Biophysica Acta, 1788:2362-2373, Sep. 2009.
Knuth et al., Hydrogel delivery systems for vaginal and oral applications: Formulation and biological considerations, Advanced Drug Delivery Reviews, vol. 11, No. 1-2, Jul.-Aug. 1993, pp. 137-167.
Lane, Majella E., “Skin penetration enhancers,” International Journal of Pharmaceutics 447 (2013) 12-21.
Lindmark, Tuulikki et al., “Absorption Enhancement through Intracellular Regulation of Tight Junction Permeability by Medium Chain Fatty Acids in Caco-2 Cells,” JPET 284(1):362-369, 1998.
Lindmark, Tuulikki et al., “Mechanisms of Absorption Enhancement by Medium Chain Fatty Acids in Intestinal Epithelial Caco-2 Cell Monolayers,” JPET 275(2):958-964, 1995.
Lopes, Luciana B. et al., Enhancement of transdermal delivery of progesterone using medium-chain mono and diglycerides as skin penetration enhancers, Pharmaceutical Development and Technology, 14:5, 524-529, Mar. 2009.
Mac Bride, Maire B. et al., “Vulvovaginal Atrophy,” Mayo Clin Proc, Jan. 2010, 85(1):87-94.
Monti, D. et al., Effect of different terpene-containing essential oils on permeation of estradiol through hairless mouse skin, International Journal of Pharmaceutics, 237:209-24, 2002.
Pachman et al., “Management of menopause-associated vasomotor symptoms: Current treatment options, challenges and future directions,” International Journal of Women's Health, May 7, 2010.
Potluri, Praveen and Guru V. Betageri, “Mixed-micellar proliposomal systems for enhanced oral delivery of progesterone,” Drug Delivery, 2006, vol. 13, No. 3, pp. 227-232.
Prajapati Hetal N. et al., “A Comparative Evaluation of Mono-, Di- and Triglyceride of Medium Chain Fatty Acids by Lipid/Surfactant/Water Phase Diagram, Solubility Determination and Dispersion Testing for Application in Pharmaceutical Dosage Form Development,” Pharm Res. Jan. 2012; 29(1): 285-305. Published online Aug. 23, 2011. doi: 10.1007/s11095-011-0541-3.
Prajapati Hetal N. et al., “Effect of Difference in Fatty Acid Chain Lengths of Medium-Chain Lipids on Lipid/Surfactant/Water Phase Diagrams and Drug Solubility,” J. Excipients and Food Chem. 2 (3) 2011:73-88.
Rao, R. et al., “The Affect of Capmul, Labrafil and Transcutol on Progesterone 100 Mg Soft Capsules Bioavailability in Indian Healthy Adult Postmenopausal Female Subjects Under Fasting Conditions,” Bioequivalence & Bioavailability, 7(2):095-107, 2015.
Sallee, Verney L. et al., “Determinants of intestinal mucosal uptake of short- and medium-chain fatty acids and alcohols,” Journal of Lipid Research, 1973, vol. 14, 475-484.
Sarpal, K. et al., “Self emulsifying drug delivery systems: a strategy to improve oral bioavailability,” Current Research & Information on Pharmaceuticals Sciences (CRIPS), 2010, vol. 11, No. 3, pp. 42-49.
Search Report, Extended European Search Report for EP13741053.6, dated Jul. 1, 2015.
Search Report, Extended European Search Report for EP13807188.1, dated Nov. 23, 2015.
Search Report, International Search Report and Written Opinion for PCT/US14/61811, dated Jan. 21, 2015.
Search Report, International Search Report and Written Opinion for PCT/US15/23041, dated Jun. 30, 2015.
Search Report, International Search Report and Written Opinion for PCT/US15/42621, dated Oct. 29, 2015.
U.S. Appl. No. 12/561,515 Dec. 12, 2011 Non-Final Office Action.
U.S. Appl. No. 12/561,515 Oct. 26, 2012 Final Office Action.
U.S. Appl. No. 12/561,515 Sep. 11, 2013 Notice of Allowance.
U.S. Appl. No. 13/843,428 Jul. 2, 2015 Non-Final Office Action.
U.S. Appl. No. 14/106,655 Jun. 19, 2015 Final Office Action.
U.S. Appl. No. 14/690,955 Feb. 1, 2016 Non-Final Office Action.
Ettinger et al., “Measuring symptom relief in studies of vaginal and vulvar atrophy: the most bothersome symptom approach,” Menopause, vol. 15, No. 5, 2008, pp. 885-889.
Eugster-Hausmann et al., “Minimized estradiol absorption with ultra-low-dose 10 μg 17β-estradiol vaginal tablets,” Climacteric 2010;13:219-227.
Martelli, Mary Elizabeth, “Vaginal Medicine Administration,” The Gale Encyclopedia of Nursing and Allied Health, Gale Group, 2002, pp. 2542-2543.
Regidor, P., “Progesterone in Peri- and Postmenopause: A Review,” Geburtshilfe Frauenheilkd, Nov. 2014. 74(11):995-1002.
Simon, James A. et al., “A vaginal estradiol softgel capsule, TX-004HR, has negligible to verylow systemic absorption of estradiol: Efficacy and pharmacokineticdata review,” Maturitas 99 (2017) 51-58.
Stefanick, “Estrogens and progestins: background and history, trends in use, and guidelines and regimens approved by the US Food and Drug Administration,” The American Journal of Medicine (2005) vol. 118 (12B), 64S-73S.
Hitchcock, Christine L. et al., “Oral micronized progesterone for vasomotor symptoms—a placebo-controlled randomized trial in healthy postmenopausal women,” Menopause: The Journal of The North American Menopause Society. 19(8):886-893, Aug. 2012.
Hosmer, Jaclyn et al., “Microemulsions Containing Medium-Chain Glycerides as Transdermal Delivery Systems for Hydrophilic and Hydrophobic Drugs,” AAPS PharmSciTech, 2009, vol. 10, No. 2, pp. 589-596.
March, Charles M. et al., “Roles of Estradiol and Progesterone in Eliciting the Midcycle Luteinizing Hormone and Follicle-Stimulating Hormone Surges,” The Journal of Clinical Endocrinology & Metabolism, vol. 49, Issue 4, Oct. 1, 1979, pp. 507-513.
Sofi, Showkat Hussain et al., “Gelucire: A Versatile Formulation Excipient,” Ijppr.Human, 2017; vol. 10 (3): 55-73.
Tang et al., “Pharmacokinetics of different routes of administration of misoprostol,” Human Reproduction, 2002; 17(2):332-226.
Wang et al., “Pharmacokinetics of hard micronized progesterone capsules via vaginal or oral route compared with soft micronized capsules in healthy postmenopausal women: a randomized open-label clinical study,” Drug Des Devel Ther., 2019; 13: 2475-2482.
Related Publications (1)
Number Date Country
20200297735 A1 Sep 2020 US
Provisional Applications (3)
Number Date Country
61932140 Jan 2014 US
61894411 Oct 2013 US
61745313 Dec 2012 US
Continuations (2)
Number Date Country
Parent 16746434 Jan 2020 US
Child 16833186 US
Parent 14521230 Oct 2014 US
Child 16746434 US
Continuation in Parts (1)
Number Date Country
Parent PCT/US2013/046443 Jun 2013 US
Child 14521230 US