CA2474698C - Polymer-based compositions for sustained release - Google Patents

Polymer-based compositions for sustained release Download PDF

Info

Publication number
CA2474698C
CA2474698C CA002474698A CA2474698A CA2474698C CA 2474698 C CA2474698 C CA 2474698C CA 002474698 A CA002474698 A CA 002474698A CA 2474698 A CA2474698 A CA 2474698A CA 2474698 C CA2474698 C CA 2474698C
Authority
CA
Canada
Prior art keywords
fsh
polymer
formulation
stabilized
sustained release
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
CA002474698A
Other languages
French (fr)
Other versions
CA2474698A1 (en
Inventor
Mark A. Tracy
Henry R. Costantino
Maria Figueiredo
Kevin L. Ward
David S. Scher
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Merck Serono SA
Alkermes Controlled Therapeutics Inc
Original Assignee
Merck Serono International SA
Alkermes Controlled Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Merck Serono International SA, Alkermes Controlled Therapeutics Inc filed Critical Merck Serono International SA
Publication of CA2474698A1 publication Critical patent/CA2474698A1/en
Application granted granted Critical
Publication of CA2474698C publication Critical patent/CA2474698C/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/1605Excipients; Inactive ingredients
    • A61K9/1629Organic macromolecular compounds
    • A61K9/1641Organic macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, poloxamers
    • A61K9/1647Polyesters, e.g. poly(lactide-co-glycolide)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/24Follicle-stimulating hormone [FSH]; Chorionic gonadotropins, e.g. HCG; Luteinising hormone [LH]; Thyroid-stimulating hormone [TSH]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/02Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/06Drugs for disorders of the endocrine system of the anterior pituitary hormones, e.g. TSH, ACTH, FSH, LH, PRL, GH
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/10Drugs for disorders of the endocrine system of the posterior pituitary hormones, e.g. oxytocin, ADH
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/1605Excipients; Inactive ingredients
    • A61K9/1611Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/1605Excipients; Inactive ingredients
    • A61K9/1617Organic compounds, e.g. phospholipids, fats
    • A61K9/1623Sugars or sugar alcohols, e.g. lactose; Derivatives thereof; Homeopathic globules
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/1682Processes
    • A61K9/1694Processes resulting in granules or microspheres of the matrix type containing more than 5% of excipient
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions

Abstract

This invention relates to sustained release compositions, and methods of forming and using said compositions, in particular for the sustained release of Follicle Stimulating Hormone (FSH). The sustained release compositions comprise a polymeric matrix of a biodegradable biocompatible polymer and stabilized FSH. The method of the invention for forming a sustained release composition includes, dissolving a biodegradable biocompatible polymer in a polymer solvent to form a polymer solution; adding biologically active stabilized FSH; removing the solvent; and solidifying the polymer to form a polymer matrix containing stabilized FSH dispersed therein. Also described is a method for providing a therapeutically effective amount of stabilized FSH in a patient in need of for a sustained period comprising administering to the patient a dose of the sustained release compositions of the invention. The sustained release composition of FSH can be used to promote maturation of follicles, promote spermatogenesis and to treat fertility disorders.

Description

POLYMER-BASED COMPOSITIONS FOR SUSTAINED RELEASE

BACKGROUND OF THE INVENTION
, Follicle Stimulating Hormone (FSH) is a heterodimeric glycoprotein hormone consisting of non-covalently attached a and (3 subunits. The alpha subunit, a 92 amino acid polypeptide with 5 disulfide bonds, is comnion to the glycoprotein hormone family, which additionally includes chorionic gonadotropin, thyroid stimulating hormone and luteinizing hormone. The beta subunit, a 111 amino acid polypeptide with 6 disulfide bonds, is unique to FSH. Each subunit has two asparagine-linlced glycosylation sites.

Human FSH has been isolated from pituitary glands and from post-menopausal urine (EP 322,438) and has recently been produced recombinantly in mammalian cells (US 5,639,640, US 5,556,957, US 4,923,805, US 4,840,896, US
5,767,251, EP 212,894 and EP 521,586, see also Howles, C.M., Huma.n Reproduction Update 2(2):172-191 (1996)).
Reproductive function in female and male inarmnals, including humans, is regulated by FSH. In females, FSH promotes the development of the follicle and consequently ovulation. In males, FSH plays a role in spermatogenesis. The synthesis of FSH by gonadotroph cells takes place witliin the anterior pituitary gland, before secretion into the general circulation. The synthesis and secretion of FSII are regulated by gonadotrophin releasing hormone, secreted by specialized neurones within the hypothalamus, and steroidal and non-steroidal products secreted from the gonads. Through high-affinity binding to its membrane receptor, FSH affects the fiulction of specific target cells in the ovaries and testes and triggers intracellular mechanisms that regulate steroidogenesis, cell replication, and the expression of specific proteins and growth factors that control gametogenesis.

Difficulties associated with exogenous administration of FSH include a short in vivo half-life requiring frequent, typically daily inj ections to achieve the desired therapeutic results. Generally such a dosing regime can result in poor patient compliance and consequently unsuccessful treatment. In addition, significant fluctuations of FSH levels in the bloodstream can cause inadequate maturation of the follicles also resulting in unsuccessful treatment.
Therefore, a need exists for FSH sustained release formulations which can lead to greater patient compliance while overcoming the difficulties which can be associated with the administration of FSH.

SUMMARY OF THE INVENTION
This invention relates to a composition, and methods of forming and using said composition, for the sustained release of FSH. The sustained release composition comprises a biocompatible polymeric matrix of a poly(lactide-co-glycolide) copolymer having a molecular weight from about 5kD to about 40kD, preferably, from about 7kD to about 20kD, such as from about 10kD to about 20kD
and having a stabilized FSH formulation, dispersed therein. The stabilized FSH
formulation comprises FSH, at least one sugar and optionally at least one buffer salt.

The concentration of FSH in the sustained release composition is between about 0.05% (w/w) and about 15% (w/w) of the total weight of the composition. In particular embodiments, the concentration of FSH is between about 0.1 1%
(w/wand about 1% (w/w). As such, the amount of stabilized FSH formulation needed to achieve this concentration of FSH in the sustained release composition can be determined based on the amount of FSH in the stabilized forinulation. The sugar can be a disaccharide, for example, sucrose, lactose or trehalose. The stabilized FSH
formulation which is incorporated into the polymer matrix comprises about 30%
(w/w) to about 99% (w/w) sugar based on the total dry weight of the stabilized formulation, such as about 50% (w/w) to about 99% (w/w) sugar based on the total dry weight of the stabilized formulation, about 1% to about 70% (w/w) FSH
based on the total dry weight of the stabilized formulation, for example, about 1%
to about 50% (w/w) FSH, such as about 1% to about 30% FSH, and about 0% to about 25%
(w/w) buffer salt based on the total dry weight of the stabilized formulation.
In preferred embodiments, the stabilized FSH formulation which is incorporated comprises 70% to 97% (w/w) sugar, 3% to 30% (w/w) FSH and 0% to 10% (w/w) buffer. The polymer can be a poly(lactide-co-glycolide) copolyiner with a terminal methyl ester, a terminal acid group or a blend of the copolymers.
The blend can be at a ratio of acid terminal end: ester terminal end for example, 1:3 one acid end to tliree ester ends. In particular embodiments, the sustained release composition releases FSH in humans over a period of at least five days, preferably the FSH is released for a period of about five days to about tliirty days. In preferred embodiments, FSH is released over a period of about five to fourteen days. In other embodiments, the sustained release composition releases FSH in humans for a period of at least 30 days. In preferred embodiments, the composition is in the forin of microparticles.
The method of the invention, for forming a composition for the sustained release of FSH includes dissolving a poly(lactide-co-glycolide) copolymer having a molecular weight from about 5kD to about 40kD such as from about 7kD to about 20kD, for example, from about l OkD to about 20kD in a polymer solvent to forin a polymer solution, adding the stabilized FSH formulation comprising FSH and at least one sugar to the polymer solution to achieve a polymer/stabilized FSH
formulation mixture with a FSH final concentration of between about 0.05%
(w/w) and about 15% (w/w) of the dry weight of the coinposition, removing the polymer solvent thereby forming a solid polymer matrix containing the FSH dispersed therein.

Typically, the stabilized FSH formulation will be added to the polymer solution in the solid form. However, the FSH can be soluble in the polymer solution. That is, the stabilized FSH forinulation can be soluble in the polymer solvent or predissolved in an FSH solvent prior to addition to the polymer solution.
When an FSH solvent is used and is different from the polymer solvent, all solvents (FSH and polymer) can be removed to forin the solid polymer matrix containing the stabilized FSH dispersed therein. When the solvent removed is a combination of the stabilized FSH formulation solvent and polymer solvent, the total solvent is referred to as the solvent phase. When the stabilized FSH formulation is dissolved in a stabilized FSH formulation solvent prior to addition to the polymer solvent the stabilized FSH formulation solvent must be miscible with the polymer solvent, not cause substantial precipitation of the polymer and not be deleterious to the FSH.

Suitable stabilized FSH fonnulation solvents include, for example: ethanol, methanol, water, acetonitrile, dimethylformamide, dimethylsulfoxide, and combinations thereof.
Suitable solvents for poly (lactide-co-glycolide) include: dimethysulfoxide, ethyl acetate, methylacetate, methylene chloride, chloroform, hexafluoroisopropanol, acetone, and combinations thereof.
The method can further comprise the step of forming droplets of the polymer/ stabilized FSH formulation mixture prior to removal of the solvent or solvent phase. Further, the method can comprise freezing the droplets prior to removal of the solvent of solvent phase. According to the method of the invention, the droplets can be microdroplets. In a specific embodiment wherein droplets are formed and then frozen, the polymer solvent or solvent phase can be removed by an extraction process. Alternatively, the polymer solvent or solvent phase can be reinoved by an evaporation process or a coinbination of an evaporation and extraction process.
The term "microdroplet" as used herein, refers to a droplet of any morphology which has a dimension less than or equal to about 1,000 microns.
In one embodiment, the method of the invention for using the sustained release composition of FSH, as described herein, coinprises administering to a patient in need of treatment a therapeutically effective amount of a composition for the sustained release of FSH, comprising a poly(lactide-co-glycolide) copolymer having a molecular weight from about 5kD to about 40kD, preferably, about 7 kD
to about 20 kD, such as from about 10 kD to about 20 kD and a stabilized FSH
formulation dispersed therein. The stabilized FSH formulation comprises FSH
and at least one sugar. The stabilized FSH formulation can optionally include at least one salt, such as a buffer salt. In particular embodiments, the concentration of FSH in the sustained release composition is between about 0.05% (w/w) and 15% (w/w) of the dry weight of the composition. As such, the amount of the stabilized FSH
formulation needed to achieve this concentration of FSH in the sustained release composition can be determined based on the amount of FSH in the stabilized formulation.
The amount of FSH present in the stabilized FSH formulation can be from about 1% (w/w) to about 70% (w/w), for exainple, from about 1% to about 50%
(w/w), such as from about 1% to about 30% (w/w). In a particular embodiment, the amount of FSH present in the stabilized FSH formulation can be from about 3%

(w/w) to about 30% (w/w) based on the total dry weight of the stabilized forinulation.
The concentration of the sugar in the stabilized formulation is about 30%
(w/w) to about 99% (w/w) based on the total dry weight of the stabilized fonnulation, such as from about 50% (w/w) to about 99% (w/w). In particular embodiments, the sugar is present from about 70% (w/w) to about 97% (w/w) based on the total dry weight of the stabilized formulation. In preferred embodiments, the sugar is a disaccharide, such as, lactose, sucrose and trehalose.
The buffer salt is present in the stabilized formulation from about 0% (w/w) to about 25% (w/w) of the total dry weight of the formulation. In a particular embodiment, the buffer salt is present from about 1% (w/w) to about 10% of the total dry weight of the stabilized formulation.
hi another embodiment, the invention is a method of promoting or stimulating the maturation of follicles in the ovaries of a patient comprising administering to a patient in need of treatment a therapeutically effective amount of a sustained release composition comprising a poly(lactide-co-glycolide) copolymer having a molecular weight from about 5kD to about 40kD, preferably, about 7 kD
to about 20 kD, such as from about 101cD to about 20 kD and a stabilized FSH
formulation dispersed therein. The stabilized FSH formulation comprises FSH
and at least one sugar. The stabilized FSH formulation can optionally include at least one salt, such as a buffer salt. In particular embodiments, the concentration of FSH in the sustained release composition is between about 0.05% (w/w) and 15% (w/w) of the dry weight of the composition. As such, the amount of the stabilized FSH
formulation needed to achieve this concentration of FSH in the sustained release composition can be determined based on the amount of FSH in the stabilized formulation.
The amount of FSH present in the stabilized FSH formulation can be from about 1% (w/w) to about 70% (w/w), for example, from about 1% to about 50%
(w/w), such as from about 1% to about 30% (w/w). In a particular embodiment, the amount of FSH present in the stabilized FSH formulation can be from about 3%

(w/w) to about 30% (w/w) based on the total dry weight of the stabilized formulation.
The concentration of the sugar in the stabilized formulation is about 30%
(w/w) to about 99% (w/w) based on the total dry weight of the stabilized formulation, such as from about 50% (w/w) to about 99% (w/w). In particular embodiments, the sugar is present from about 70% (w/w) to about 97% (w/w) based on the total dry weight of the stabilized formulation. In preferred embodiments, the sugar is a disaccharide, such as, lactose, sucrose and trehalose.
The buffer salt is present in the stabilized formulation from about 0% (w/w) to about 25% (w/w) of the total dry weight of the formulation. In a particular embodiment, the buffer salt is present from about 1% (w/w) to about 10% of the total dry weight of the stabilized formulation.
In yet another embodiment, the invention is a method of promoting spermatogenesis in the testes of a patient comprising administering to a patient in need of treatment a therapeutically effective amount of a sustained release composition comprising a poly(lactide-co-glycolide) copolymer having a molecular weight from about 51cD to about 40kD, preferably, about 7 kD to about 20 kD, such as from about 10 kD to about 20 kD and a stabilized FSH formulation dispersed therein. The stabilized FSH formulation comprises FSH and at least one sugar.
The stabilized FSH formulation can optionally include at least one salt, such as a buffer salt. In particular embodiments, the concentration of FSH in the sustained release composition is between about 0.05% (w/w) and 15% (w/w) of the dry weight of the composition. As such, the amount of the stabilized FSH formulation needed to achieve this concentration of FSH in the sustained release composition can be determined based on the amount of FSH in the stabilized formulation.

The amount of FSH present in the stabilized FSH formulation can be from about 1% (w/w) to about 70% (w/w), for example, from about 1% to about 50%
(w/w), such as from about 1% to about 30% (w/w). In a particular embodiment, the ainount of FSH present in the stabilized FSH formulation can be from about 3%
(w/w) to about 30% (w/w) based on the total dry weight of the stabilized formulation.

The concentration of the sugar in the stabilized formulation is about 30%
(w/w) to about 99% (w/w) based on the total dry weight of the stabilized formulation, such as from about 50% (w/w) to about 99% (w/w). In particular embodiments, the sugar is present from about 70% (w/w) to about 97% (w/w) based on the total dry weight of the stabilized formulation. In preferred embodiments, the sugar is a disaccharide, such as, lactose, sucrose and trehalose.

The buffer salt is present in the stabilized formulation from about 0% (w/w) to about 25% (w/w) of the total dry weight of the formulation. In a particular embodiment, the buffer salt is present from about 1% (w/w) to about 10% of the total dry weight of the stabilized formulation.
In another embodiment, the invention relates to a method of treating fertility disorders. The method comprises administering to a patient in need of treatment a therapeutically effective amount of a sustained release composition comprising a poly(lactide-co-glycolide) copolymer having a molecular weight from about 5kD
to about 40kD, preferably, about 7 kD to about 20 kD, such as from about 1010 to about 20 kD and a stabilized FSH formulation dispersed therein. The stabilized FSH
formulation comprises FSH and at least one sugar. The stabilized FSH
formulation can optionally include at least one salt, such as a buffer salt. In particular embodiments, the concentration of FSH in the sustained release composition is between about 0.05% (w/w) and 15% (w/w) of the dry weight of the composition.
As such, the ainount of the stabilized FSH formulation needed to achieve this concentration of FSH in the sustained release composition can be determined based on the amount of FSH in the stabilized formulation.

The amount of FSH present in the stabilized FSH formulation can be from about 1% (w/w) to about 70% (w/w), for example, from about 1% to about 50%
(w/w), such as from about 1% to about 30% (w/w). In a particular embodiment, the amount of FSH present in the stabilized FSH formulation can be from about 3%
(w/w) to about 30% (w/w) based on the total dry weight of the stabilized formulation.
The concentration of the sugar in the stabilized formulation is about 30%
(w/w) to about 99% (w/w) based on the total dry weight of the stabilized formulation, such as from about 50% (w/w) to about 99% (w/w). In particular embodiments, the sugar is present from about 70% (w/w) to about 97% (w/w) based on the total dry weight of the stabilized formulation. In preferred embodiments, the sugar is a disaccharide, such as, lactose, sucrose and trehalose.
The buffer salt is present in the stabilized formulation from about 0% (w/w) to about 25% (w/w) of the total dry weight of the fonnulation. In a particular einbodiment, the buffer salt is present from about 1% (w/w) to about 10% of the total dry weight of the stabilized formulation.

BRIEF DESCRIPTION OF THE DRAWINGS
The foregoing and other objects, features and advantages of the invention will be apparent from the following more particular description of preferred embodiments of the invention, as illustrated in the accompanying drawings FIG. 1 is a plot of FSH serum concentrations in rats versus time in days following administration of the indicated FSH sustained release compositions.
FIG. 2 is a plot of FSH serum concentrations in rats versus time in days following administration of different doses of the indicated FSH sustained release compositions (dose normalized).
FIG. 3 is a plot of FSH serum concentrations in rats versus time in days following adininistration of different doses of the indicated FSH sustained release coinpositions (not dose normalized).

FIG. 4 is a plot of FSH serum concentrations in rats versus time in days following administration of the indicated sustained release compositions.

FIG. 5 is a plot of FSH serum concentrations in rats versus time in days following administration of the indicated sustained release compositions.

FIG. 6 is a plot of FSH serum concentrations in rats versus time in days following administration of the indicated sustained release compositions.

FIG. 7a is a plot of FSH serum concentrations in rats versus time in days following administration of the indicated sustained release compositions.

FIG. 7b is a plot of FSH serum concentrations in humans versus time in days following administration of the indicated FSH sustained release compositions.
DETAILED DESCRIPTION OF THE INVENTION

A description of preferred embodiments of the invention follows.
The present invention is based on the unexpected discovery that a composition comprising a poly (lactide-co-glycolide) copolymer with a molecular weight from about 5kD to about 40kD, preferably, about 7kD to about 20kD such as from about lOkD to about 20kD and having a stabilized FSH formulation dispersed therein can be used to deliver FSH in a sustained manner. In general, it is desirable that the sustained release of FSH occurs for a period of at least five days.

The sustained release compositions of the invention comprise a biocompatible polymer matrix of a poly(lactide-co-glycolide) copolymer having a molecular weight from about 5kD to about 40kD, preferably, about 7kD to about 20kD such as from about 10kD to about 20kD and having a stabilized FSH

formulation dispersed therein. The stabilized FSH formulation comprises FSH
and at least one sugar. The sugar can be a disaccharide. The FSH formulation can optionally contain at least one buffer salt. The concentration of FSH in the sustained release composition is between about 0.05% (w/w) and about 15% (w/w) of the total dry weight of the sustained release composition. As such, the amount of stabilized FSH formulation needed to achieve this concentration of FSH in the sustained release composition can be determined based on the amount of FSH in the stabilized formulation.

Typically, the FSH is present in the stabilized formulation from about 1%
(w/w) to about 70% (w/w), for example, from about 1% to about 50% (w/w), such as from about 1% to about 30% (w/w). In a particular einbodiment, the amount of FSH
present in the stabilized FSH formulation can be from about 3% (w/w) to about 30%
(w/w) based on the total dry weight of the stabilized formulation. The sugar can be present in the stabilized formulation from about 30% (w/w) to about 99% (w/w) based on the total dry weight of the stabilized formulation, such as from about 50%
(w/w) to about 99% (w/w). In particular embodiments, the sugar is present from about 70% (w/w) to about 97% (w/w) based on the total dry weight of the stabilized forinulation. The buffer salt can be present in the stabilized FSH formulation from about 0% (w/w) to about 25% (w/w), such as from about 1% (w/w) to about 10%
(w/w) of the total dry weight of the stabilized formulation.

The poly(lactide-co-glycolide) (hereinafter "PLG") can have a lactide:glycolide ratio, for example, of about 10:90, 25:75, 50:50, 75:25 or 90:10. In a preferred embodiment of the invention, the lactide:glycolide ratio of the poly(lactide-co-glycolide) copolymer is 50:50. In certain embodiment, the end groups of the poly (lactide-co-glycolide) are in the methyl ester form. In other embodiments, the end groups of the poly(lactide-co-glycolide) polymer are in the acid form. In further embodiments, the ester form and acid form of the poly(lactide-co-glycolide) can be blended at a suitable ratio. For example, from about 10%
of either the ester form or acid form to about 90% of the acid form or ester form, respectively. Preferably, the sustained release coinposition releases FSH over a period of at least 5 days in humans.

The composition of the present invention as described herein provides a means for eliciting a therapeutic effect in a patient in need thereof by administering a composition comprising poly(lactide-co-glycolide) copolymer and a stabilized FSH
formulation dispersed within.
The "stabilized FSH formulation" as defined herein, comprises FSH and at least one sugar. The FSH formulation can optionally contain at least one buffer salt.
The stabilized FSH formulation can decrease degradation, aggregation, loss of potency and/or loss of biological activity of the FSH, all of which can occur during formulation of the sustained release composition, and prior to and/or during in vivo release.
A "sugar" as defined herein, is a mono, di or trisaccharide or polyol such as a polysaccharide. Suitable monosaccharides include, but are not limited to glucose, fructose and mannose. A"disaccharide" as defined herein is a compound which upon liydrolysis yields two molecules of a monosaccharide. Suitable disaccharides include, but are not limited to sucrose, lactose and trehalose. Suitable trisaccharides include but are not limited to raffinose and acarbose. It is preferred that the sugar is a non-reducing disaccharide. The amount of sugar present in the stabilized FSH
formulation can range from about 30% (w/w) to about 99% (w/w), such as from about 50% (w/w) to about 99% (w/w) of the total dry weight of the stabilized formulation. In particular embodiments, the sugar is present from about 70%
(w/w) to about 97% (w/w).
"Buffer salt" as defined herein is the salt remaining following removal of solvent from a buffer. Buffers are solutions containing either a wealc acid and a related salt of the acid, or a wealc base and a salt of the base. Buffers can maintain a desired pH to stabilize the formulation. For example, the buffer can be monobasic phosphate salt or dibasic phosphate salt or combinations thereof or a volatile buffer such as ammonium bicarbonate. Other buffers include but are not limited to acetate, citrate, succinate and amino acids such as glycine, arginine and histidine.
The buffer can be present in the stabilized formulation from about 0% to about 10%
of the total weight of the stabilized formulation. In a preferred embodiment, the buffer salt is a sodium phosphate salt.
"Surfactants" as the term is used herein refers to any substance which can reduce the surface tension between immiscible liquids. Suitable surfactants which can be added to the sustained release coinposition include polymer surfactants, such as nonionic polymer surfactants, for example, poloxamers, polysorbates, polyethylene glycols (PEGs), polyoxyethylene fatty acid esters, polyvinylpyrrolidone and combinations thereof. Examples of poloxamers suitable for use in the invention include poloxamer 407 sold under the trademark PLURONIC F 127, and poloxamer 188 sold under the trademark PLURONIC F68, both available from BASF
Wyandotte. Examples of polysorbates suitable for use in the invention include polysorbate 20 sold under the trademark TWEEN 20 and polysorbate 80 sold under the trademark TWEEN 80. Cationic surfactants, for example, benzallconium chloride, are also suitable for use in the invention. In addition, bile salts, such as deoxycholate and glycocholate are suitable as surfactants based on their highly effective nature as detergents. The surfactant can be present in the polymer phase or present in the stabilized FSH formulation. The surfactant can act to modify release of the FSH from the polymer matrix, can act to stabilize the FSH or a combination thereof.
In addition, other excipients can be added to the polyiner phase to modify the release of the FSH from the sustained release composition. Such excipients include salts, such as sodium chloride.
"Antioxidants" can also be added to the sustained release coinposition.
Suitable antioxidants can include, but are not limited to, methionine, vitamin C, vitamin E and maleic acid. The antioxidant can be present in the stabilized FSH
formulation or added in the polymer phase. In a particular embodiment, methionine can be added to reduce the oxidation of the disulfides and methionine residues in FSH.
The method of the present invention comprises administering the FSH
sustained release compositions described herein to provide a therapeutic or diagnostic effect in a patient in need of such treatment. In preferred embodiments, the sustained release composition can be administered by injection or implantation.
The therapeutic or diagnostic effect can be, for example, the stimulation of ovarian follicular development. Such effects can be useful in women undergoing assisted reproductive technology, including in vitro fertilization (IVF), embryo transfer (EF) and anovulatory infertile women, such as those with polycystic ovary disease. As such, the sustained release composition of the invention comprising a poly (lactide-co-glycolide) copolymer and a stabilized FSH formulation dispersed therein can be used for the treatment of infertility. For example, the sustained release composition having a biocompatible polymer with a stabilized FSH
formulation incoiporated therein can be suitable for use as a treatment in women with infertility problems related to follicle maturation. To effect final maturation of the follicle and ovulation in the absence of an endogenous LH surge, human chorionic gonadotropin (hCG) can be given simultaneously, following the administration of the sustained release composition, or co-encapsulated with stabilized FSH in the sustained release composition, when monitoring of the patient indicates that sufficient follicular development has occurred.

In another embodiment, the sustained release composition can be administered to the patient to stimulate spermatogenesis in the testes for the treatment of infertility. The release profile for male infertility can be for longer time periods, such as at least 30 days, preferably, for about 60 or about 90 days.
In males, FSH blocks inhibition of spermatogenesis by sertolli cells.

Currently, FSH is administered to stimulate ovarian follicular development by daily injection. Daily injections typically results in poor patient compliance. An example of a current daily injection protocol for female infertility is deinonstrated in Santibrink and Fauser (Santibrink, E. and Fauser, B. "Urinary Follicle-Stimulating Hormone for Normogonadotropic Clomiphene-Resistant Anovulatory Infertility:
Prospective, Randomized Comparison Between Low Dose Step-Up and Step-Down Dose Regimens," J. Clin. Endocrinology Metab. 52:3597-3602, 1997). As described herein, the FSH sustained release composition can release FSH in a sustained manner for an advantageous period of time.
Further, the sustained release composition can minimize some of the side effects seen with daily administration of FSH, such as Ovarian Hyperstimulation Syndrome which can occur with or without pulmonary or vascular complications.
Additionally, the sustained release composition results in a lower initial burst of the FSH when compared to administering FSH alone.

The invention described herein also relates to pharmaceutical compositions suitable for use in the invention. In one embodiment, the pharmaceutical composition comprises a sustained release composition comprising a biocompatible polymeric matrix of poly(lactide-co-glycolide) copolymer having a molecular weight from about 5kD to about 40kD, preferably, about 7kD to about 20kD such as from about lOkD to about 20kD and a stabilized FSH formulation dispersed therein.
The concentration of FSH in the sustained release composition is from about 0.05%
(w/w) to about 15% w/w. In certain embodiments, the composition can have a period of sustained release in vivo in humans for at least 5 days such as from about 5 days to about 30 days. In other embodiments, such as for women, FSH is released for up to 14 days. In men, FSH can be released for periods of at least 30 days and up to about 90 days, such as about 60 days.
The invention also relates to the use of a sustained release device comprising a poly(lactide-co-glycolide) copolymer having a molecular weight from about 5kD
to about 40kD and a stabilized FSH formulation comprising FSH and at least one sugar wherein the stabilized FSH formulation is dispersed within the polymer for the manufacture of a medicament for use in therapy.

Sustained release of biologically active FSH formulation is a release of the active FSH formulation from a sustained release composition, which occurs over a period of time which is longer than that time period during which a therapeutically significant amount of the biologically active FSH would be available following direct administration of a solution of the biologically active FSH. The resulting in vivo PK profile of FSH from a sustained release composition is also much more consistent (maintained in a desired therapeutic window) than the profile observed following administration of FSH in solution.
Methods of monitoring pharmacokinetics (PK) can be accomplished using widely available techniques such as IRMA analysis of FSH in biological fluids as is described in the Exemplification.

Follicle Stimulating Hormone (FSH), as defined herein, includes all forms of FSH and can be derived from animal, preferably human sources, or recombinantly produced. FSH as defined herein, refers to a compound having the primary, secondary and/or tertiary molecular structure of native FSH, and which has at least one FSH pharmacodynamic effect as measured in standard FSH bioassays. FSH
includes analogs, deglycosylated forms, unglycosylated forms and modified glycosylated forms. The most preferred form is produced by recombinant DNA
teclmiques. For Example, GONAL-F (Ares-Serono) is an example of a currently available FSH formulation given by injection.

"Patient" as that term is used herein, refers to the recipient of the treatment.
Mammalian and non-mammalian patients are included. In a specific embodiment, the patient is a mammal, such as a human, canine, murine, feline, bovine, ovine, swine or caprine. In a preferred embodiment, the patient is a human. In a most preferred embodiment, the patient is a female human.

The term "sustained release composition", as defined herein, comprises poly(lactide-co-glycolide) copolymer having a molecular weight of about 5 kD
to about 40 kD, preferably, about 7kD to about 20 kD, such as about 10 kD to about 20 kD and a stabilized FSH formulation dispersed therein. In a preferred embodiment, the amount of FSH present in the sustained release composition is about 0.05%
(w/w) to about 15% (w/w) of the total dry weight composition. As such, the amount of stabilized FSH formulation needed to achieve this concentration of FSH in the sustained release composition can be determined based onthe amount of FSH in the stabilized formulation. The amount of FSH administered will vary depending upon the desired effect, patient evaluation, the planned release levels, and the time span over which the FSH will be released.

In alternative compositions, the sustained release composition can contain greater than 15% (w/w) FSH (total dry weight of composition). For example, the alternative coinpositions can contain 20% to 50% FSH based on the total dry weight of the sustained release coinposition.
In one embodiment, the sustained release composition is not a sustained release composition comprising a biocoinpaible polymer which is a 75/25 blend of 50:50 2M/50:50 2A PLG as described herein and a stabilized FSH formulation comprising 10% FSH, 10% sodium phosphate and 80% sucrose with a target load of about 0.5% (w/w) of the total dry weight of the sustained release composition.

The sustained release compositions of this invention can be fonned into many shapes such as a film, a pellet, a rod, a filament, a cylinder, a disc, a wafer, a gel, or a microparticle. A microparticle is preferred. A "microparticle", as defined herein, comprises a polymer component as described having a diameter of less than about one millimeter and having a stabilized FSH formulation dispersed therein. A
microparticle can have a spherical, non'spherical or irregular shape.
Typically, the microparticle will be of a size suitable for injection. A preferred size range for microparticles is from about one to about 180 microns in diameter.
The composition of this invention can be administered in vivo, for example, to a human, or to an animal, orally, or parenterally such as by injection, implantation (e.g., subcutaneously, intramuscularly, intraperitoneally, intracranially, and intradermally), administration to mucosal ineinbranes (e.g., intranasally, intravaginally, intrapuhnonary, buccally or by means of a suppository), or in situ delivery (e.g., by enema or aerosol spray) to provide the desired dosage of FSH
based on the known parameters for treatment with FSH of particular medical conditions.
In preferred embodiments, the sustained release composition, as described herein, is administered by injection. "Injection" as that term is used herein, includes administration through a delivery port alone or in combination with a surgical scope such as a laparoscope, endoscope, laryngoscope, cystoscope, protoscope or thoracoscope. The delivery port can be, for example, a surgical tube such as a catheter with an appropriately sized bore, or a needle or needle-like port. As such, delivery can include a minor incision in the patient to permit entry of a delivery port, such as a needle or catheter, or a combination of a delivery port and a surgical scope.
Advantageously, injection of the composition avoids the need for an open surgical procedure to expose the delivery site.

The sustained release of FSH is release of FSH from a polyiner matrix comprising a poly(lactide-co-glycolide) polymer having a molecular weight from about 51cD to about 40kD, preferably, 7kD to about 201cD such as from about lOkD
to about 20kD which occurs over a period which is longer than that period during which a biologically significant amount of FSH would be available following direct adininistration of a solution of FSH. In certain embodiments of the present invention, the period of sustained release of FSH occurs over a period of at least 5 days, such as from about 5 days to about 30 days. In other embodiments, the period of sustained release is from about 7 days to about 14 days. In further embodiments, the period of sustained release can be greater than about 30 days such as about 60 days or about 90 days. A sustained release of biologically active FSH, from a sustained release coinposition can be a continuous or a discontinuous release, with relatively constant or varying rates of release. The continuity of release and level of release can be affected by the polymer composition used (e.g., molecular weight and polymer chemistry, such as choice of end groups and the addition of other excipients which modify release such as surfactants and salts) and FSH loading.
A polymer is biocompatible if the polyiner and any degradation products of the polymer are non-toxic to the recipient and also possess no significant deleterious or untoward effects on the recipient's body, such as an iminunological reaction at the injection site.
"Biodegradable", as defined herein, means the composition will degrade or erode in vivo to form smaller chemical species. Degradation can result, for example, by enzymatic, chemical and physical processes.
Suitable biocompatible, biodegradable polyiners for alternative embodiments include, for example, poly(lactide), poly(glycolide), poly(lactide-co-glycolide) at molecular weights less than 5kD or greater than 40kD, poly(lactic acid)s, poly(glycolic acid), polycarbonates, polyesteramides, polyanhydrides, poly(amino acids), polyorthoesters, poly(dioxanone)s, poly(alkylene alkylate)s, polyetheresters, polyphosphoesters, biodegradable polyurethane, blends thereof, and copolymers thereof.
In further alternative embodiments, suitable biocompatible, non-biodegradable polymers include non-biodegradable polymers selected from the group consisting of polyacrylates, polymers of ethylene-vinyl acetates and other acyl substituted cellulose acetates, non-degradable polyurethanes, polystyrenes, polyvinylchloride, polyvinyl fluoride, poly(vinyl imidazole), chlorosulphonate polyolefins, polyethylene oxide, poly ethylene glycol, poloxamers, polypropylene oxide, blends thereof, and copolymers thereof.
In alternative embodiments, acceptable molecular weights for polymers can be determined by a person of ordinary skill in the art talcing into consideration factors such as the desired polymer degradation rate, physical properties such as mechanical strength, and rate of dissolution of polymer in solvent. Typically, an acceptable range of molecular weight is less than 5,000 Daltons or between about 40,000 Daltons to about 2,000,000 Daltons.
The stabilized FSH formulation can be prepared by methods known in the art such as freeze drying, spray-freeze drying, spray drying and those described in U.S
Patent No. 6,284,283 by Costantino et al.
For example, the stabilized FSH formulation can be prepared by atomizing, using multifluid atomization, a fluid comprising the FSH at least one sugar and at least one solvent at a mass flow ratio of about 0.3 or greater to produce droplets, freezing the droplets to produce frozen droplets, removing the solvent from the frozen droplets to produce friable microstructures, forming a dispersion of the friable microstructures in at least one non-solvent for the FSH and fragmenting the dispersed friable microstructures to produce particles of stabilized FSH formulation.
A number of methods are known by which sustained release compositions (polymer/active agent matrices) can be formed. At a single stage of the process, solvent is removed from the microparticles and thereafter the microparticle product is obtained. Methods for forming a composition for the sustained release of biologically active agent are described in U.S. Patent No. 5,019,400, issued to Gombotz et al., and issued U.S. Patent No. 5,922,253, issued to Herbert et al.

In this method, a mixture comprising a biologically active agent, a biocompatible polymer such as poly (lactide-co-glycolide) and a polymer solvent such as methylene chloride is processed to create droplets, wherein at least a significant portion of the droplets contains polymer, polymer solvent and the active agent. These droplets are then frozen by a suitable means. Examples of means for processing the mixture to form droplets include directing the dispersion through an ultrasonic nozzle, pressure nozzle, Rayleigh j et, or by other known means for creating droplets from a solution. I
Means suitable for freezing droplets include directing the droplets into or near a liquified gas, such as liquid argon or liquid nitrogen to form frozen microdroplets which are then separated from the liquid gas. The frozen microdroplets are then exposed to a liquid or solid non-solvent, such as ethanol, hexane, ethanol mixed with hexane, heptane, ethanol mixed with heptane, pentane or oil.
The solvent in the frozen microdroplets is extracted as a solid and/or liquid into the non-solvent to form a polymer/active agent matrix comprising a biocompatible polymer and a biologically active agent. Mixing ethanol with other non-solvents, such as hexane, heptane or pentane, can increase the rate of solvent extraction, above that achieved by ethanol alone, from certain polymers, such as poly(lactide-co-glycolide) polymers.
A wide range of sizes of sustained release compositions can be made by varying the droplet size, for example, by changing the ultrasonic nozzle diameter. If the sustained release composition is in the form of microparticles, and very large microparticles are desired, the microparticles can be extruded, for example, through a syringe directly into the cold liquid. Increasing the viscosity of the polymer solution can also increase microparticle size. The size of the microparticles which can be produced by this process ranges, for example, from greater than about 1000 to about 1 micrometers in diameter.
When sterile product is desired, the environment in which the process steps are performed can be aseptic. Generally, terminal sterilization of the protein powder or polymer is not recommended. However, the use of Barrier (or Isolator) Technology can provide an aseptic environment. For example, Barrier Technology (Work Station Isolator, LaCalhene, Inc..) can be used to provide an aseptic environment for the manipulation, production and harvesting processes as follows:
1) The Isolator's internal environment (including the surface of any equipment or material packages present) can undergo a decontamination procedure, using vaporized hydrogen peroxide (VHP), a strong oxidizing agent.

2) The decontamination treatment, along with the unit's air filtration system, can ensure the Isolator's internal environment meets and/or exceeds class 100.
3) The operators can be completely segregated from the internal isolator environment through the use of Half Suits and/or glove extensions of the isolator(s).
4) Product contact materials can be sterilized prior to their entry into the isolator via filtration, steam, or dry heat.
5) Materials that are not sterilized during isolator decontamination (See 1, above) can be sterilized prior to their entry into the isolator.
-All process gasses and solutions can enter into the isolator via a 0.2 in sterile filters.

-Materials and equipment can be first sterilized in a dry heat oven or autoclave.
-If the aforementioned material or equipment is not sterilized as indicated above, the surfaces of these items (or packages of items) can be decontaminated using VHP within a transfer isolator (i.e., small isolator for transferring iteins to the Work Station Isolator).
The Workstation Isolator can be a flexible walled two half-suit Isolator that can be made of PVC and Divetex. It can have an inlet and outlet HEPA
filtration, ventilation/recirculation system and multiple self-propelled fans, which can provide the chamber's turbulent airflow pattern. There can be one transfer port that connects to a VHP-sanitizable freeze dryer. The Isolator can use vapor hydrogen peroxide, which can be generated from a VHP Generator such as those available from Amsco, Inc. using preprogrammed validated cycles, to decontaminate the internal environment. All product manipulation, product transfers and harvest procedures can be performed within the confines of the Isolator.
Yet another method of forming a sustained release composition, from a suspension comprising a poly(lactide-co-glycolide) copolymer and a stabilized FSH
formulation, includes fihn casting, such as in a mold, to form a film or a shape. For instance, after putting the suspension into a mold, the polymer solvent is then removed by means known in the art, preferably at a lowered temperature until a film or shape, with a consistent dry weight, is obtained.
A further example of a conventional microencapsulation process and microparticles produced thereby is disclosed in U.S. Pat. No. 3,737,337 wherein a solution of a wall or shell forming polymeric material in a solvent is prepared. The solvent is only partially miscible in water. A solid or core material is dissolved or dispersed in the polymer-containing mixture and, thereafter, the core material-containing mixture is dispersed in an aqueous liquid that is immiscible in the organic solvent in order to remove solvent from the microparticles.
Another example of a process in which solvent is removed from microparticles contain.ing a substance is disclosed in U.S. Pat. No.
3,523,906, incorporated herein by reference in its entirety. In this process a material to be encapsulated is emulsified in a solution of a polymeric material in a solvent that is immiscible in water and then the emulsion is emulsified in an aqueous solution containing a hydrophilic colloid. Solvent removal from the microparticles is then accomplished by evaporation and the product is obtained.
In still another process as shown in U.S. Pat. No. 3,691,090 organic solvent is evaporated from a dispersion of microparticles in an aqueous medium, preferably under reduced pressure.

Similarly, the disclosure of U.S. Pat. No. 3,891,570 shows a method in which solvent from a dispersion of microparticles in a polyhydric alcohol medium is evaporated from the microparticles by the application of heat or by subjecting the microparticles to reduced pressure.

Another example of a solvent removal process is shown in U.S. Pat. No.
3,960,757.
Tice et al., in U.S. Pat. No. 4,389,330, describe the preparation of microparticles containing an active agent by a method comprising: (a) dissolving or dispersing an active agent in a solvent and dissolving a wall forming material in that solvent; (b) dispersing the solvent containing the active agent and wall forming material in a continuous-phase processing medium; (c) evaporating a portion of the solvent from the dispersion of step (b), thereby forming microparticles containing the active agent in the suspension; and (d) extracting the remainder of the solvent from the microparticles.
Without being bound by a particular theory, it is believed that the release of the FSH can occur by two different mechanisms. First, FSH can be released by diffusion through aqueous filled channels generated in the polymer matrix, such as by the dissolution of the FSH, or by voids created by the removal of the polymer solvent during the preparation of the sustained release composition. A second mechanism is the release of the biologically active agent, due to degradation of the polymer. The rate of degradation can be controlled by changing polymer properties that influence the rate of hydration of the polymer. These properties include, for instance, the ratio of different monomers, such as lactide and glycolide, comprising a polymer; the use of the L-isomer of a monomer instead of a racemic mixture;
and the molecular weight of the polymer and the end group chemistry of the polymer (i.e., acid vs. ester). These properties can affect hydrophilicity and crystallinity, which control the rate of hydration of the polymer.
By altering the properties of the polymer, the contributions of diffusion and/or polymer degradation to FSH release can be controlled. For example, increasing the glycolide content of a poly(lactide-co-glycolide) polymer and decreasing the molecular weiglit of the polymer can enhance the hydrolysis of the polymer and thus, provides an increased biologically active agent release from polymer erosion.

EXEMPLIFICATION
The following methods were employed to analyze samples produced during the production and characterization of the FSH containing sustained release compositions.
METHODS OF ANALYSIS

SIZE EXCLUSION CHROMATOGRAPHY (SEC) Size Exclusion Chromatography was used to quantitate monomeric recombinant huinan FSH (rhFSH) and to determine the relative amounts of dimeric, monomeric and aggregated rhFSH in samples. A TOSO HAAS column G2000SWx17.8 X 300, 5 was utilized. The flow rate was 0.5 mL/min. The mobile phase was 0.1 M Sodiuin phosphate buffer pH 6.7, 0.1 M sodium sulfate.
REVERSE-PHASE HPLC (RPHPLC) Reverse-phase HPLC was used to determine the purity of FSH samples, specifically the amounts of oxidized and native rhFSH. A VYDAC column C4, 4.6mm, 5 microns, 300A was utilized. The eluent system consisted of mobile phase A, 0.1M triethylamine phosphate, mobile phase B, 100% acetonitrile and mobile phase C, 30% acetonitrile and 0.1% TFA employing the following gradient. The flow rate was 1 mL/min.

fflinutes) % Mobile Phase A %Mobile Phase B % Mobile Phase C
SDS-PAGE
SDS-PAGE methods, performed under non-reducing conditions, measured the degree of disassociation of subunits of FSH. Samples were dissolved with sample buffer. The sample was prepared and loaded on Excel gel SDS Gradient 8-18%. The resulting banding pattern was detected by staining with Silver Stain Plus.

The purity of the protein was estimated by densitometry and coinparison to a standard curve of subunit concentrations. The results were compared to bulk drug carried through the same SDS-PAGE sample preparation procedure.

PARTICLE SIZE ANALYSIS
The particle size of the microparticles was determined. The mean particle size was detennined using a Coulter Multisizer IIe. Briefly, approximately 10 mg of microparticles was added to IsotonOO (commercially available buffered saline solution) for particle size analysis. The suspension was then analyzed using a micron aperture tube to determine the particle size distribution. NIST
certified polystyrene beads were used as a calibration standard. The results were reported as a volume-weighted median diaineter.

POLYMER IDENTITY/ GEL PERMEATION CHROMATOGRAPHY
Determination of the average molecular weight of the polymer used in the sustained release composition was performed using Gel Permeation Chromatography (GPC). Samples were dissolved in chloroform to approximately lmg/mL and resolved on a Polymer Laboratories (Amherst, Massachusetts) PLgel Mixed-E column (300 mm x 7.5 mm, 3 m particles) using chloroform as the mobile phase. Peaks were detected by a refractive index detector. Polystyrene standards were used.

LOAD DETERMINATION
FSH load in the sustained release compositions was determined by nitrogen analysis using a standard carbon/hydrogen/nitrogen (CHN) elemental analyzer.
Samples (approximately 1-5 mg) were combusted at 980 C in an oxygen atmosphere to produce nitrogen and nitrogen oxides. The gas stream was reduced over copper metal at 700 C to produce eleinental nitrogen that was quantified by a thermal conductivity detector. NIST traceable acetanilide was used as a standard.
MAIACLONE IRMA ASSAY
I The amount of FSH in biological fluids was determined by using a MAIACLONE Kit (BioChem. Immunosystems Italia S.PA.) according to manufacturer's instructions. Briefly, samples, standard and controls are reacted with a mixture of two hig11 affinity monoclonal antibodies; one labeled with radioactive iodine and the other liiiked to fluorescein. An anti-fluorescein antibody coupled to a magnetic particle is added in excess. This antibody specifically binds to the FSH
monoclonal complex and is sedimented in a inagnetic field. The concentration of antigen is directly proportional to the radioactivity bound to the magnetic particles.

The bound fraction of each sample is measured in a gamma counter calibrated to detect Ilzs-Iodine. Using a four parameter fit standard curve, the concentration of FSH in the samples can be determined. The limit of detection is 0.5 mIU/mL and the limit of quantitation is 1.5 mIU/mL for the assay.

EXTRACTION
FSH was extracted from the sustained release compositions utilizing a filter method. The method was used to determine protein integrity and protein content.
Briefly, 60 mg of microspheres were suspended in 1 mL of methylene chloride to dissolve the polymer. The suspended protein-containing particles were collected on a 0.65 micron pore size filter. After the filter was allowed to dry, FSH was reconstituted in 1 mL of aqueous media. Following reconstitution, protein integrity and content were determined using SEC, RPHPLC and SDS-PAGE methods already described.
Alternatively, the suspended protein-containing particles were collected by centrifugation, and the supeniatant was removed. The pellet was allowed to dry, then reconstituted in 1 mL of aqueous media, and then analyzed for protein integrity and content, as described for the filter method.
FORMULATION
Formation of a stabilized FSH formulation by lyophilization of a mixture of FSH, sugar and optionally buffers. Freeze drying of the formulation was performed on droplets of the mixture or bulk freeze drying in Lyoguard trays (W.L. Gore and Associates, Delaware). Further details are described herein in Example 5.
When aseptic procedures are desired, the Workstation Isolator and Transfer Isolators, as described above for formation of the microparticles, can be used and can be decontaminated using for example, VHP. The hydrogen peroxide level can be verified as below target level prior to use. The buffers and solutions can be prepared using Water for Injection (WFI). The solution containing FSH and any other desired components can be sterile filtered by, for example, passage through a 0.2 m filter into the Workatation Isolator. The formulated FSH solution can be sprayed using an atomizing nozzle into a bed of liquid nitrogen. The frozen FSH
can then be freeze-dried using a suitable drying cycle. The lyophilized FSH

containing powder can then be collected and stored at about -80 C.

Polymers employed were purchased from Alkermes, Inc. of Cincinnati, OH.
Polymer 2A (Allcermes, 5050 2A) Poly (lactide-co-glycolide); 50:50 lactide:
glycolide ratio; 10kD Mol. Wt.; carboxylic acid end group.

Polymer 2M (Allcermes, 5050 2M) Poly (lactide-co-glycolide); 50:501actide:
glycolide ratio; 18kD Mol. Wt.; methyl ester end group.

Polymer 2A:2M 75% (w/w) 2M and 25% 2A w/w.

Polymer 1A :Alkermes, 5050 1A Poly (lactide-co-glycolide); 50:50 lactide:glycolide ratio, Mol Wt. 5k0, carboxylic end group Polymer 1A 45:55 (Alkermes, 45/55 lA) Poly (lactide-co-glycolide); 45:55 lactide:glycolide ratio, Mol Wt. 5kD, carboxylic end group PROCESS FOR PREPARING MICROPARTICLES

Fonnation of a polymer solution by dissolving polymer in a suitable polymer solvent.

Addition of the stabilized FSH formulation lyophilizate to the polymer solution to form a polymer/protein mixture.

Optional homogenization of the polymer/protein mixture.

Atomization of the polymer/protein mixture by sonication or other means of droplet formation, and freezing of the droplets by contact with liquid nitrogen.

Extraction of the polymer solvent from the polymer/protein droplets into an extraction solvent (e.g., -80 C ethanol), thereby forming particles comprising a polymer/stabilized protein matrix.
Isolation of the particles from the extraction solvent by filtration.
Removal of remaining solvent by evaporation.

Sieving of particles by passage through an appropriately sized mesh so as to produce an injectable product.

ASEPTIC PROCESSING:

The Workstation Isolator and Transfer Isolators were decontaminated as described above. The level of hydrogen peroxide level in the isolators was determined. All process liquids were passed through a sterile 0.2 m filter into the Worlcstation Isolator. The polymer soltion was prepared by dissolving the polymer in dichloromethan and then filtering it into the Workstation Isolator. The following steps were conducted in the Workstation Isolator: a) The required mass of FSH-containing lyophilized powder was weight out and the required volume of the polymer solution was added to the FSH-containing lyophilized powder. The protein/polymer suspension was sonicated to reduce the particle size of the FSH-containing powder. The proteinlpolymer suspension was atomized into liquid iiitrogen on top of a frozen ethanol bend. The microparticles were cured in the ethanol using a freeze dryer attached to the isolator as a freezer. The slurry of microparticles and ethanol was filtered and the collected microparticles were dried in a sterilized freeze-dryer. The microparticles were sieved inside the Worlcsation Isolator through a stainless steel sieve.
EXAMPLE 1: EFFECT OF PLG CHEMISTRY
The effects of PLG chemistry (end group, molecular weight and lactide-to-glycolide ratio) on recombinant FSH (rhFSH) integrity and release from sustained release compositions were assessed.

Table I PLG Nomenclature Formulation Name Lactide:glycolide End Group Nominal MW
Type (kD) 1 2M 50:50 methyl ester 18 2 2A 50:50 acid 10 3 1A 50:50 acid 5 10, 4 45:55 1A 45:55 acid 5 5 2A:2M 50:50 acid/ester 16 (1:3 blend) blend A number of polymers were tested (all at 1.0% rhFSH nominal load), namely, 2M, 2A and 1A, a type 1A polymer with a 45:55 lactide:glycolide ratio and a 2A:2M, 1:3 blend were also investigated.

Table II Effect of PLG type on post-encapsulation protein integrity and release profile Formulation % rhFSH PLG For the extracted protein: PK Results:
(nitrogen) Type Oxidation by Subunits by Cmax Duration RPHPLC SDSPAGE (mIU/mL) (day) (%) (%) 1-1 1.04 2M 1.4 1.2 680 70 up to 36 2-1 1.24 2A 2.0 0.94 660 30 17 3-1 0.96 lA 5.1 3.2 1000 20 14~--4 4-1 0.96 45:55 12.1 3.4 1300 20 10 The stabilized FSH formulation was made with 10%FSH, 80% sucrose and 10% phosphate salts. Several different polymer types were screened, as is summarized in Table H. The injected dose was nominally 10 mg of microparticles loaded with 1% rhFSH, or 100 g protein. PK data are expressed as the data dose-normalized to 200 g rhFSH/kg rat (FIG.1). C,,,zx is the experimentally determined as the higl7est rhFSH concentration that was observed during the study.
Duration is defined as the first time point in two consecutive time points with levels below the limit of quantitation, or if there are not two consecutive time points with levels below the limit of quantitation then it is the last time point with a measurable value.

One animal injected with Formulation 1-1 has measurable serum rhFSH
levels up to 36 days post-injection (last time point taken) whereas another animal given the same formulation had measurable levels on day 10 and day 31, but not in between, suggesting that in the latter case rhFSH was present in the serum at the end of the study, albeit near the limit of quantitation of the IRMA assay (FIG.
1).

In an additional study, the FSH serum concentration vs. time profile was repeated for Formulation 3-1. This formulation had a C,,,a,, of about 825 m1U/mL
and a Tma,, of about 15 hours.

EXAMPLE 2: EFFECT OF EXCIPIENTS
The effect of various excipients co-encapsulated with the lyophilizate during sustained release composition formulation were tested (Table III). Some formulations listed in Table III were made using a sucrose lyophilizate formulation (80:10:10, sucrose:FSH:sodium phosphate salts), as earlier described, and the others were made using a trehalose-containing lyophilizate (trehalose substituted for sucrose).
Various co-encapsulated excipients were tested for their effect on modulating rhFSH release from the sustained release profile of the final composition.
Excipients can modulate protein release via various mechanisms, for instance, by enhancing the porosity of the sustained release composition. For example, excipients that have an affinity for water can enhance water sorption into the sustained release composition, and upon dissolution can create additional porosity for protein to be released from the composition. As shown below, the potential release modifiers tested included the salt sodium chloride, and surfactants such as the poloxamer Pluronic F-127, and poly(ethylene glycol), (PEG) 8000 kD MW.
These compounds have an affinity for water and cover the range from the highly soluble, low-molecular-weight salt that diffuses rapidly from the microparticles to a soluble polymer (PEG) which should diffixse more slowly out of the microparticles.
The in vivo release profiles of FSH sustained release compositions were analyzed. In a typical PK experiment, three male Sprague-Dawley rats (450 50g) were injected subcutaneously with 10 mg of microparticles suspended in 0.75 mL
of TM
diluent having 3% carboxymethyl cellulose, 0.9% NaCl, and 0.1% TWEEN-20 in water for injection. Serum samples were collected for several time points for the first day, and approximately once per day for up to 35 days, depending on expected duration of the formulation tested. FSH was quantitated from serum samples using the MAIAclone (IRMA) provided by BioChem. Immunosystems Italia S.P.A. Data presented are typically dose-normalized to 200 micrograms FSH/kg.
The rhFSH release profiles demonstrated that the addition of 10% F127, formulation 3-3, or 5% F127 and 5% NaCl, formulation 3-4, to the 1A polymer did not substantially alter the release profile compared to the case of the 1A
polymer without the modifier. In all these cases the Cm. and duration were similar. At higher concentrations of release modifiers, for example, 10% NaCl in the lA
polymer phase (3-5), and a combination of 5% PEG and 10% NaCI in the lA
polymer (3-7), the duration was reduced by the presence of release modifiers to 14 days and 10 days, respectively. These results can be compared to formulation 3-3, where 10% F127 was added and the sustained release duration was 17 days. A
similar result was obtained for release modifiers in the 2A polymer. In the case where 10% NaCl (2-3) or 5% PEG and 10% NaCl, formulation 2-4 were added to 2A polymer microparticles, the last day of measurable rhFSH levels was 17 days post-injection, whereas for formulation 2-2 with 5% F127 and 5% NaCl were added, the duration was 21 days. There were no trends in Cn,a,, as a result of the addition of release modifiers. The 1A polymer samples containing release modifiers had a Cm"' ranging from about 800 to 1100 mIU/mL (Table III), comparable to the release modifier-free sample (Table IT). A similar result was seen for the 2A polymer sustained release composition samples.
Table III also presents data for the percent oxidation and percent subunit formation of the protein following extraction from the microparticles using the metllylene chloride centrif-ugation method. The formation of subunits was very low, 1% or less, in all formulations. The SEC data, not shown, demonstrated about 100%
monomeric protein for all samples. The oxidation for all formulations was 3-5%
for the formulations containing release modifiers.

Table III Effect of release modifiers on post-encapsulation protein integrity and release profile Form % PLG Stabilized Co- For the extracted PK Analyses:
ula- Rh- Type FSH sprayed protein:
tion FSH Formulation Additive Oxi- Sub C' ' Dura dation units (mIU/n-L) tion by RP- by (Day) HDLC SDS-% PAGE

2-2 1.0 2A 80:10:10 5% 4.9 0.8 620 70 21 sucrose:FSH: F127/5%
sod. phos. salt NaC1 2-3 1.1 2A 80:10:10 10% 4.9 1.0 900 20 17 sucrose:FSH: NaC1 sod. phos. salt 3-2 1.4 2A 80:10:10 5%a PEG 4.5 0.4 710 100 17 trehalose:FSH: 10%
sod. phos. salt NaC1 3-3 1.0 1A 80:10:10 10% 3.2 0.4 1100 50 17 trehalose:FSH: F127 sod. phos. salt 3-3 1.2 1A 80:10:10 5% 5.2 0.8 11001150 17 sucrose:FSH: F127/5%
sod. phos. salt NaCl 3-4 1.2 1A 80:10:10 10% 4.5 1.3 1000 100 14 sucrose:FSH: NaC1 sod. phos. salt 3-5 1.1 lA 80:10:10 5 /a PEG 5.0 0.5 1100+100 16+7 trehalose:FSH:
sod. phos. salt 3-6 1.4 lA 80:10:10 5% PEG 4.6 0.4 800 60 10 trehalose:FSH: 10%
sod. phos. salt NaCl EXAMPLE 3: EFFECTS OF PROTEIN LOAD, DOSE AND SUCROSE VS.
TREHALOSE AS STABILIZING EXCIPIENT
The effect of protein loading on protein integrity and release was assessed.
In addition to the load of 1% discussed above, lower FSH loads of 0.5% and 0.25%
were also tested. PK studies were conducted for the same 100 g dose of rhFSH
per rat, corresponding to administration of 10, 20 and 40 mg of the sustained release composition, respectively. This study utilized a sucrose-containing lyophilizate formulation for the 2A and 2M polymer types. In addition, the same load-ranging series of polymer types was produced witli the alternate trehalose lyophilizate. Data are presented in Table IV.

Table IV Effect of load and stabilizer on post-encapsulation integrity and release profile Formulation % PLG Stabilized For the extracted PK analysis:
Load Type FSH Form- protein:
rh ulation FSH Oxidation Subunits Cmax by RP- by SDS- (mIU/mL) HPLC % PAGE %

1-1 1.04 2M sucrose 1.4 1.2 680 70 1-2 0.48 2M sucrose 1.6 1.4 400 10 1-3 0.27 2M sucrose 2.3 2.2 360 30 1-4 0.92 2M trehalose 2.3 0.7 690 10 1-5 0.48 2M trehalose 1.6 0.7 220 30 1-6 0.25 2M trehalose 1.8 1.1 220 20 2-1 1.24 2A sucrose 2.0 0.9 66W:30 2-5 0.64 2A sucrose 2.2 1.6 3201150 280 3b 2-6 0.32 2A sucrose 3.2 3.6 300-00 2-7 0.75 2A trehalose 4.2 1.4 900 60 2-7 0.53 2A trehalose 3.4 2.0 340 20 320 20b 2-7 0.25 2A trehalose 4.2 2.1 290 30 a Injected dose was nominally 100 g protein except as noted.

b Injected dose was nominally 50 g protein.
All samples in Table IV contained about 100% monomeric protein by SEC
and up to about 4% oxidized and subunit species. No trends were observed between the protein stability and load or choice of sugar, for example, the sugar can be a disaccharide such as sucrose or trehalose. The release profiles were similar.
Therefore, both sucrose and trehalose are suitable stabilizers for rhFSH upon lyophilization and subsequent encapsulation in PLG sustained release compositions.
Modification of the dose of encapsulated rhFSH was also studied. For the sucrose-containing lyophilizate loaded at 0.5% rhFSH in 2A polymer, administering sustained release coinposition doses (nominally) of 15 mg, 5 mg and 2 mg were tested. The serum profiles out to 21 days are shown in FIG. 2. Upon normalization, all doses for the 2A polymer sustained release compositions from 2-15 mg (10-75 g rhFSH/rat) yielded similar release profiles over 7 days. 16 mg and 8 mg of the 2A:2M polymer blend formulation (Type 5) using the same lyophilizate and load were also administered. Serum data for these doses of the blend are shown in FIG.
2. Upon nonnalization to the saine dose, the rhFSH levels were comparable for the 16 mg and 8 mg doses. In addition, the release profiles for the 2A:2M polymer blend sustained release compositions were distinctly different than the 2A
polymer sustained release compositions, regardless of the dose. These same data are also presented in non dose-normalized form in FIG. 3. Data beyond 17 days for all doses were below the assay quantitation limit.

EXAMPLE 4: CHARACTERIZATION OF rhFSH SUSTAINED RELEASE
COMPOSITION
Table V presents data for a number of batches of tllree formulations of rhFSH sustained release compositions (all at 0.5% protein load, three different polymers, namely, 2M, 2A and a 1:3 (w/w) blend of 2A:2M, formulations 1, 2 and respectively). These sustained release compositions batches were made using the sucrose-containing lyophilizate fonnulation ( 80:10:10; sucrose, FSH, phosphate salts). The data for extracted protein were generated using the filter extraction method (except where so noted in Table V). In addition to integrity data for the extracted protein (using the filter method), the Table also presents sustained release composition characterization data: median particle size (D,.50) and moisture content.
For comparison, Table V also contains data from Example 1 of 2A (formulation 2-5), 2M (formulation 1-2) and the 2A:2M blend formulation (5-1). The data show that stability of rIiFSH towards encapsulation was similar to that observed in earlier batches of the same fonnulation. For all batches, the median microparticle size was about 40-60 m. The range of water content for all polymer batches was between about 0.5 and 1.0%. Release profiles for various batches of the lead rhFSH
microparticle formulations are shown in FIG. 4 (2A polymer fonnulation), FIG.

(2M polymer formulation) and FIG.6 (2A:2M polymer blend formulation). FIGS. 4, 5 and 6 and Table V show that the in vitro and in vivo results are similar between the batches of each formulation.
Table V Characterization of microparticles containing a load of 0.5% protein formulated as 80:10:10 sucrose: protein: sodium phosphate Formulation % Polymer Dv.50 HZ0 For the extracted protein:
rh Type (gm) (%) FSH Monomer Oxidation Sub-by SEC by RP- units by (%) HPLC (%) SDS-PAGE
(%) 1-2 0.48 2M 58.7 0.52 100a 1.6a 1.4a 1-7 0.59 2M 41.7 0.57 99.5 2.4 -<0.25 1-8 0.51 2M 51.7 1.54 na na na 1-9 0.61 2M na 0.70 99.6 3.1 ' -<0.25 1-10 0.61 2M 51.8 0.65 99.7 2.8 -<0.25 56.0 2-4 0.64 2A 67.7 0.84 100a 2.2a 1.6a 2-9 0.51 2A 59.5 0.73 99.7 3.1 -<0.25 2-10 0.59 2A 39.9 0.90 99.7 6.6 -<0.25 2-11 0.48 2A 54.0 0.84 99.7 4.3 -<0.25 46.9 2-12 0.61 2A na na 99.8 6.9 -<0.25 2-13 0.48 2A 64.0 0.68 99.7 5.5 -<0.25 1-10 0.61 2M 51.8 0.65 99.7 2.8 -<0.25 56.0 5-1 0.61 2A:2M na 0.37 99.7a 1.2a 0.8a 1:3 5-2 0.56 2A:2M 54.2 0.85 99.6 2.6 {0.25 1:3 5-3 0.61 2A:2M na na 99.8 2.8 <0.25 1:3 5-4 0.59 2A:2M 42.0 0.59 99.7 2.8 {0.25 1:3 a Data from a centrifuge-extracted sample b na=not available EXAMPLE 5: STABILIZED FSH FORMULATION
The stabilized lyophilizate used in the sustained release composition described above were identified by their ability to minimize degradation, aggregation, loss of potency and/or loss of the FSH, all of which can occur during formulation of the sustained release composition. Stable FSH-containing lyophilizates which were subsequently encapsulated in polymer were produced.
Lyophilizate formulation studies were conducted to identify additives that stabilize FSH through the spray-freeze drying processing step and to assess the stability of the lyophilizates after being exposed to moisture at a physiologic temperature, a condition mimicking the early stages of protein dissolution and release from microparticles.
Seven FSH-containing lyophilizates were produced to identify salts and additives that stabilized FSH formulation through lyophilization and after exposure to moisture. Each lyophilizate contained 10% FSH, 10% salt, and the remainder (80%) a stabilizing additive. The lyophilizate composition was determined based on an estimated total load of 100 g FSH to be delivered in one weekly injection and a target lyophilizate load (FSH mass+salt+stabilizer) of 10% in 10 mg of microparticles. The formulations are summarized in Table VI.
Table VI Components of Initial Lyophilizate Formulations Formulas Protein Salt Additive L-1 FSH Sodium Phosphate Sucrose L-2 FSH Sodium Bicarbonate Sucrose L-3 FSH Sodium Phosphate Trehalose L-4 FSH Sodiuin Bicarbonate Trehalose L-5 FSH Sodium Phosphate Ammonium Sulfate L-6 FSH Sodium Bicarbonate Ammonium Sulfate L-7 FSH Sodium Bicarbonate Zinc Acetate The formulations were prepared by adding solutions of the additive and salt to the bulk drug. Each formulated solution was then spray-freeze dried to produce a lyophilized powder. A sample of each powder was dissolved in DI water and evaluated by SEC to assess post-lyophilization stability. In addition, each powder was exposed to 100% relative humidity for 24 hours in 37 C and subsequently evaluated by SEC to assess post-humidification stability. The results from these experiments are given in Tables VII and VIII, respectively.
Table VII. Effect of Formulation Components on FSH Stability After Lyophilization Formulatio Nominal Measured % % Monomer n Concentration Concentration Recovered ( g/mL) (SEC) (Measured/
( g/mL) Nominal x 100) peak tailing The data in Table VII show that FSH is stable through lyophilization as assessed by SEC for 6 of 7 formulations. The low recovery and peak tailing observed in the chromatogram for sample L-7 ()Wzinc acetate) may be indicative of degradation.
Table VIII. Effect of Formulation Components on FSH Lyophilizate Stability After Exposure to Moisture at 37 C

Formulation Nominal Measured % Recovered %
Concentration Concentration (Measured/ Monomer ( g/mL) (SEC) Nominal x ( g/mL) 100) Table VIII shows that the salt and additive have significant effects on the stability of FSH after humidification. The data suggest that these formulations are stable through lyophilization and humidification. Both the sucrose and trehalose formulations made with bicarbonate showed a small peak at 13.7 minutes by SEC
suggesting the presence of aggregates. Formulations L-5, L-6 and L-7 had aggregates and/or subunits. Ammonium sulfate appears to be a strongly destabilizing additive for FSH.
Based on these results, trehalose/phosphate and sucrose/phosphate lyophilizate formulations have been identified as stable lyophilizates.
The stabilized FSH formulation is prepared from a formulated aqueous solution containing FSH, a stabilizing excipient (e.g., sugar) and possibly at least one buffer salt The formulated aqueous solution can be dried into a friable form suitable for processing to produce sustained release compositions by a variety of pharmaceutical processing methods such as bulk freeze drying, spray drying, spray-freeze drying, rotary evaporation vacuum drying, and supercritical fluid drying.
Spray-freeze drying in particular is suitable for production of highly friable dried solids that, according to the processing conditions, can yield micron down to sub-micron powders (Costantino et al., U.S. Patent No. 6,284,283.
Somewhat less friable powders can be achieved by bulk freeze drying. In a preferred embodiment, the formulated aqueous solution can be poured into a container, for example a LYOGUARDTM tray (W.L. Gore & Associates, Elkton,lVID), frozen on the lyophilizer shelf, and dried in a lyophilizer. In another preferred embodiment, the formulated aqueous solution in sprayed into a freezing medium (e.g., liquid nitrogen) using an atomization technique (e.g., single fluid, high pressure nozzle) and the liquid nitrogen slurry is poured into the container, and the frozen material dried by lyophilization in a lyophilizer. The latter embodiment allows for production of powders with larger particle size compared to those generated by spray-freeze drying.
Comparison of the particle sizes generated using these different approaches for the 10% FSH, 80% sucrose and 10% phosphate salt stabilized FSH formulation is presented in the following table:
Table IX

Liquid Processing Method Particle Size (D,50 in microns) Two fluid atomization (1) 0.41 Two fluid atomization (2) 0.47 Bulk frozen in LYOGUARD tray (1) 7.7-10.4 Bulk frozen in LYOGUARD tray (2) 9.6 Bulk frozen in LYOGUARD tray (3) 7.3-9.0 Flash frozen (single fluid atomization) 0.7-2.0 EXAMPLE 6: PHARMACOKINETIC STUDIES OF FSH UNENCAPSULATED
The pharmacokinetics (PK) of recombinant huinan Follicle Stimulating Hormone (rhFSH) (Serono) bulk doses were evaluated in male Sprague-Dawley rats weighing about 400g:L50g (SD) to demonstrate the ability to detect FSH in serum.
For rhFSH PK characterization the doses were:
A) 0.5 g/0.5mL as an intravenous (IV) bolus B) 10 g/0.5mL, 5 g/0.5mL and 1 4g/0.5mL as a subcutaneous bolus (SC) C) 0.5 g/hr (0.5ug/ L), 0.25 g/hr (0.25 g/ L) and 0.05 g/hr (0.05 g/ L) as a continuous SC delivery from an ALZET osmotic pump (Mode12001, 1.0 1/hr, 1 week duration). The vehicle for the IV and SC bolus studies, and the SC
osmotic pump study was 0.9% saline.
Blood samples were obtained at various time points and serum was separated for assay. Samples were analyzed according to the immunoradiometric assay (IRMA) method. The following parameters for rhFSH were estimated:
distribution and elimination phase half-lives (alpha ald beta HL), area under the serum concentration-time curve (AUC) from time zero to infinity, maximum observed concentration (Cmax) time for the maximum observed concentration (Tmax) and average steady-state concentration from 2 days to 7 days following SC pump iinplantation (Css). The following studies were completed, rhFSH 01 - rhFSH
03.
The single dose pharmacolcinetics of rhFSH in rats following a single IV

dose of 0.5 g/0.5mL (actual dose: 0.455 g/0.5mL) were determined: mean AUC
o_ ,ast and AUCo_;,,fn;h,were 3535 and 3852mIU/hr/mL, respectively, and alpha and beta half-lives for the distribution and elimination phases were 0.46+0.19 and 4.02:L1.04hr, respectively, with a clearance of 36.43mL/hr/kg Three doses of rhFSH were given as a SC single bolus. The rhFSH was absorbed in a dose proportional manner witll maximum mean serum concentration at approximately 9.4 to10 hours. A comparison of the mean PK parameters from the three dose groups A, B, and C of the SC bolus treatment show alpha and beta half-lives of 5.5 0.97 and 8.0 1.6 hr for the l0ug dose group (actual dose:
9:02 g);
and 4.8:L1.6 and 8.6 1.3 hr for the 5 g dose group (actual dose: 4.55 g);
and 5.4 ~:0.98 and 10.3 5.0 hr for the 1 g dose group (actual dose:l g) the Tmax values were 9.5 0.4, 9.4 ::L0.6 and 10.0 0.9 hr per group, respectively. The mean Cmax values 23 5 27 mIU/mL for the 10 g group, 90 9.3 mIU/mL for the 5 g group, and 16.1 2.0, for the 1 g group. The mean bioavailibilities of the SC bolus groups were 82.2 6.7, and 63.7 18.7 percent, respectively. Cmax, AUC, and relative bioavailability all increased with increasing dose in a slightly non-proportional manner.
In an osmotic pump study, the pharmacokinetics of rhFSH in non-immunosuppressed rat model versus an iiumunosuppressed rat model were first compared. Two groups were compared each received 0.25 g of rhFSH infused per hour (actual dose: 0.273 g/hr) with one group receiving Sandiinmune Cyclosporine intra-peritoneal (IP). No significant differences between groups B and D were noticed.
In addition, sustained release levels from the osmotic pump were assess for all doses and the 0.25 g/hr immunosuppressed group. A mean Css (from day 2 to day 7) of 149 5.2 mICT/mL for the 0.5 g/hr group, 70 4.1 mIU/mL for the 0.25ug/hr group, 13 1.8 mlU/inL for the 0.05 g/hr group, and 79 7.1 mIU/mL
for the 0.25 g/hr in the immunosuppressed group. The absolute bioavailability for the four groups ranged from 37% to 42%; and relative bioavailability based upon the SC bolus doses ranged from 47% to 57%. Linearity was observed for group C.
values throughout the study.

EXAMPLE 7: Pharmacokinetic Profiles in Rats of Lots 2-14, 1-11, and 5-5, and comparison with Human data.

Table X

Formulation PLG Mol. Sugar salt %FSH
wt.

1-11 2M 18 sucrose sodium 0.51 (methyl phosphate ester) 2-14 2A 10 sucrose sodium 0.54 (acid) phosphate 5-5 2A:2M 16 sucrose sodium 0.55 (1:3 blend) phosphate Each formulation from Table X was injected into 3 male Sprague-Dawley rats at a dose of 200 glkg of sustained release composition. Serum samples were collected periodically and the rhFSH serum concentration was determined using the MAIAclone IItMA assay. FIG. 7a shows the serum concentration of rhFSH versus time in days following administration of the indicated fornlulations. FIG. 7b shows the human data of serum concentration of rhFSH versus time in days following subcutaneous administration of the same formulations at a dose of 7 g/kg.

While this invention has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims.

Claims (20)

What is claimed is:
1. A method for forming a composition for the sustained release of FSH
comprising:

a) dissolving a poly(lactide-co-glycolide) copolymer having a molecular weight from about 5kD to about 40kD in a polymer solvent to form a polymer solution;

b) adding a stabilized FSH formulation comprising, by weight of the total dry weight of the stabilized FSH formulation, 10% FSH, 10% of a phosphate buffer salt and 80%
trehalose;

c) removing the solvent from the polymer/stabilized FSH mixture; and d) solidifying the polymer to form a polymer matrix containing the stabilized FSH
formulation dispersed therein.
2. The method of Claim 1 further comprising the steps of:

a) forming droplets of the polymer/stabilized FSH formulation mixture;

b) freezing the droplets of the polymer/stabilized FSH formulation mixture wherein said forming and freezing steps are performed prior to removal of the solvent.
3. The method of Claim 1 wherein the solvent is removed by extraction with an extraction solvent.
4. The method of Claim 1 wherein the droplets are microdroplets.
5. The method of Claim 1 wherein the extraction solvent is ethanol.
6. The method of Claim 1 wherein the sustained release composition is in the form of microparticles.
7. A composition for the sustained release of FSH prepared by a method comprising:

a) dissolving a poly(lactide-co-glycolide) copolymer having a molecular weight from about 5kD to about 40kD in a polymer solvent to form a polymer solution;

b) adding a stabilized FSH formulation comprising, by weight of the total dry weight of the stabilized FSH formulation, 10% FSH, 10% of a phosphate buffer salt and 80%
trehalose to achieve a mixture comprising a polymer/stabilized FSH
formulation; and c) removing the solvent from the polymer/stabilized FSH mixture, thereby forming a polymer matrix containing solid FSH dispersed therein.
8. The composition of Claim 7 wherein the method further comprising the steps of:
a) forming droplets of the polymer/stabilized FSH formulation mixture;

b) freezing the droplets of the polymer/stabilized FSH formulation mixture wherein said forming and freezing steps are preformed prior to removal of the solvent.
9. The composition of Claim 7 wherein the solvent is removed by extraction with an extraction solvent.
10. The composition of Claim 9 wherein the droplets are microdroplets.
11. The composition of Claim 7 wherein the extraction solvent is ethanol.
12. The composition of Claim 7 which is in the form of microparticles.
13. A composition for the sustained release of FSH comprising:

a) a poly(lactide-co-glycolide) copolymer having a molecular weight from about 5kD
to about 40kD; and b) a stabilized FSH formulation comprising, by weight of the total dry weight of the stabilized FSH formulation, 10% FSH, 10% of a phosphate buffer salt and 80%
trehalose wherein the stabilized FSH formulation is dispersed within the polymer.
14. The composition of Claim 13, wherein the FSH is released for at least 5 days.
15. The composition of Claim 13, wherein the FSH is released for at least 30 days.
16. The composition of Claim 13, wherein the poly(lactide-co-glycolide) copolymer has a molecular weight from about 10kD to about 20kD.
17. The composition of Claim 13, wherein the poly(lactide-co-glycolide) copolymer has an acid terminal group.
18. The composition of Claim 13, wherein the poly(lactide-co-glycolide) copolymer has a methyl ester terminal group.
19. The composition of Claim 13, wherein the poly(lactide-co-glycolide) copolymer is a blend comprising at least one acid terminal end group poly(lactide-co-glycolide) and at least one methyl ester terminal poly(lactide-co-glycolide).
20. The composition of Claim 19, wherein the blend of copolymers is a ratio of one acid terminal end group to 3 ester terminal end groups.
CA002474698A 2002-02-08 2003-02-07 Polymer-based compositions for sustained release Expired - Fee Related CA2474698C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US35515902P 2002-02-08 2002-02-08
US60/355,159 2002-02-08
PCT/US2003/003981 WO2003066585A2 (en) 2002-02-08 2003-02-07 Polymer-based compositions for sustained release

Publications (2)

Publication Number Publication Date
CA2474698A1 CA2474698A1 (en) 2003-08-14
CA2474698C true CA2474698C (en) 2009-07-21

Family

ID=27734472

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002474698A Expired - Fee Related CA2474698C (en) 2002-02-08 2003-02-07 Polymer-based compositions for sustained release

Country Status (9)

Country Link
US (1) US20040028733A1 (en)
EP (1) EP1471891A4 (en)
JP (1) JP2005517012A (en)
AU (1) AU2003217367B2 (en)
CA (1) CA2474698C (en)
IL (1) IL163218A (en)
NZ (1) NZ535008A (en)
WO (1) WO2003066585A2 (en)
ZA (1) ZA200405852B (en)

Families Citing this family (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040234602A1 (en) * 2001-09-21 2004-11-25 Gina Fischer Polymer release system
US20040253310A1 (en) 2001-09-21 2004-12-16 Gina Fischer Morphine polymer release system
US20050019380A1 (en) * 2002-04-26 2005-01-27 Xylos Corporation Microbial cellulose wound dressing for treating chronic wounds
AU2003277446A1 (en) 2002-10-17 2004-05-04 Alkermes Controlled Therapeutics, Inc. Ii Microencapsulation and sustained release of biologically active polypeptides
ES2360102T3 (en) 2003-03-26 2011-05-31 Egalet A/S SYSTEM FOR CONTROLLED RELEASE OF MORPHINE.
KR101105486B1 (en) 2003-04-02 2012-01-13 아레스 트레이딩 에스.에이. Liquid pharmaceutical formulations of FSH and LH together with a non-ionic surfactant
US7090433B2 (en) * 2003-10-07 2006-08-15 Steve Searby Underground cable laying apparatus
MXPA06011924A (en) 2004-04-15 2007-06-11 Alkermes Inc Polymer-based sustained release device.
US7456254B2 (en) 2004-04-15 2008-11-25 Alkermes, Inc. Polymer-based sustained release device
NZ550926A (en) 2004-05-17 2010-01-29 Ares Trading Sa Hydrogel interferon formulations
UA92146C2 (en) 2004-06-01 2010-10-11 Эйрэс Трейдинг С.А. Stabilized interferon liquid formulations
US7238974B2 (en) * 2004-10-29 2007-07-03 Infineon Technologies Ag Semiconductor device and method of producing a semiconductor device
US11246913B2 (en) 2005-02-03 2022-02-15 Intarcia Therapeutics, Inc. Suspension formulation comprising an insulinotropic peptide
CA2611081C (en) * 2005-06-03 2016-05-31 Egalet A/S A drug delivery system for delivering active substances dispersed in a dispersion medium
KR101105871B1 (en) * 2005-09-27 2012-01-16 주식회사 엘지생명과학 hFSF Aqueous Formulation
ES2422864T3 (en) 2006-08-09 2013-09-16 Intarcia Therapeutics, Inc Osmotic release systems and piston units
AU2007234612B2 (en) * 2006-12-14 2013-06-27 Johnson & Johnson Regenerative Therapeutics, Llc Protein stabilization formulations
NZ577560A (en) * 2007-01-16 2012-01-12 Egalet Ltd Use of i) a polyglycol and ii) an active drug substance for the preparation of a pharmaceutical composition for i) mitigating the risk of alcohol induced dose dumping and/or ii) reducing the risk of drug abuse
RU2440097C2 (en) 2007-04-23 2012-01-20 Интарсия Терапьютикс, Инк. Method of treating insulin-independent diabetes and obesity, osmotic delivery system and method for making it
AU2008258596B2 (en) 2007-06-04 2013-02-14 Egalet Ltd Controlled release pharmaceutical compositions for prolonged effect
US7678764B2 (en) 2007-06-29 2010-03-16 Johnson & Johnson Regenerative Therapeutics, Llc Protein formulations for use at elevated temperatures
CA2695697A1 (en) 2007-08-07 2009-02-12 Advanced Technologies And Regenerative Medicine, Llc Protein formulations comprising gdf-5 in aqueous acidic solution
WO2009043843A1 (en) * 2007-10-01 2009-04-09 Universite Catholique De Louvain Scaffolds for follicle transplantation
US20100323441A1 (en) * 2007-12-13 2010-12-23 Biocell Center S.P.A. Method of collection and preservation of fluids and/or materials, in particular of organic fluids and/or materials containing stem cells, and device employable in such method
WO2009080699A2 (en) 2007-12-20 2009-07-02 Merck Serono S.A. Peg-interferon-beta formulations
EP2240155B1 (en) 2008-02-13 2012-06-06 Intarcia Therapeutics, Inc Devices, formulations, and methods for delivery of multiple beneficial agents
AU2009236459B2 (en) * 2008-04-14 2013-07-25 Advanced Technologies And Regenerative Medicine, Llc Liquid buffered GDF-5 formulations
CN101269215B (en) * 2008-05-15 2011-03-23 上海天伟生物制药有限公司 Glucoprotein incretion composition
NZ594207A (en) 2009-02-06 2013-03-28 Egalet Ltd Immediate release composition resistant to abuse by intake of alcohol
EP2445487A2 (en) 2009-06-24 2012-05-02 Egalet Ltd. Controlled release formulations
EP3323423B1 (en) 2009-09-28 2020-06-17 Intarcia Therapeutics, Inc Rapid establishment and/or termination of substantial steady-state drug delivery
KR101042302B1 (en) * 2010-12-27 2011-06-17 위재영 The housing for exterior imaging device of the vehicle
US20120208755A1 (en) 2011-02-16 2012-08-16 Intarcia Therapeutics, Inc. Compositions, Devices and Methods of Use Thereof for the Treatment of Cancers
IN2014DN06817A (en) * 2012-01-19 2015-05-22 Natrix Separations Inc
EP2877161A1 (en) 2012-07-06 2015-06-03 Egalet Ltd. Abuse deterrent pharmaceutical compositions for controlled release
US9889085B1 (en) 2014-09-30 2018-02-13 Intarcia Therapeutics, Inc. Therapeutic methods for the treatment of diabetes and related conditions for patients with high baseline HbA1c
EP3302354B1 (en) 2015-06-03 2023-10-04 i2o Therapeutics, Inc. Implant placement systems
WO2017200943A1 (en) 2016-05-16 2017-11-23 Intarcia Therapeutics, Inc. Glucagon-receptor selective polypeptides and methods of use thereof
USD860451S1 (en) 2016-06-02 2019-09-17 Intarcia Therapeutics, Inc. Implant removal tool
USD840030S1 (en) 2016-06-02 2019-02-05 Intarcia Therapeutics, Inc. Implant placement guide
KR20190104039A (en) 2017-01-03 2019-09-05 인타르시아 세라퓨틱스 인코포레이티드 Methods Including Continuous Administration of GLP-1 Receptor Agonists and Co-administration of Drugs
US11376220B2 (en) * 2017-06-30 2022-07-05 Therio, LLC Single-injection methods and formulations to induce and control multiple ovarian follicles in bovine, caprine, ovine, camelid and other female animals

Family Cites Families (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE634668A (en) * 1962-07-11
BE744162A (en) * 1969-01-16 1970-06-15 Fuji Photo Film Co Ltd ENCAPSULATION PROCESS
DE2010115A1 (en) * 1970-03-04 1971-09-16 Farbenfabriken Bayer Ag, 5090 Leverkusen Process for the production of micro-granules
JPS523342B2 (en) * 1972-01-26 1977-01-27
GB1413186A (en) * 1973-06-27 1975-11-12 Toyo Jozo Kk Process for encapsulation of medicaments
US4389330A (en) * 1980-10-06 1983-06-21 Stolle Research And Development Corporation Microencapsulation process
CH661206A5 (en) * 1983-09-23 1987-07-15 Debiopharm Sa PROCESS FOR THE PREPARATION OF A MEDICINAL PRODUCT FOR THE TREATMENT OF HORMONDEPENDENT DISEASES.
US4923805A (en) * 1983-11-02 1990-05-08 Integrated Genetics, Inc. Fsh
US5639639A (en) * 1983-11-02 1997-06-17 Genzyme Corporation Recombinant heterodimeric human fertility hormones, and methods, cells, vectors and DNA for the production thereof
US4840896A (en) * 1983-11-02 1989-06-20 Integrated Genetics, Inc. Heteropolymeric protein
US6217911B1 (en) * 1995-05-22 2001-04-17 The United States Of America As Represented By The Secretary Of The Army sustained release non-steroidal, anti-inflammatory and lidocaine PLGA microspheres
US5019400A (en) * 1989-05-01 1991-05-28 Enzytech, Inc. Very low temperature casting of controlled release microspheres
MY107937A (en) * 1990-02-13 1996-06-29 Takeda Chemical Industries Ltd Prolonged release microcapsules.
US5384132A (en) * 1990-03-20 1995-01-24 Akzo N.V. Stabilized gonadotropin containing preparations
IT1250075B (en) * 1991-12-18 1995-03-30 Serono Cesare Ist Ricerca PHARMACEUTICAL COMPOSITIONS CONTAINING GONADOTROPINE.
EP0674506B1 (en) * 1992-12-02 2000-08-23 Alkermes Controlled Therapeutics, Inc. Controlled release growth hormone containing microspheres
US6447796B1 (en) * 1994-05-16 2002-09-10 The United States Of America As Represented By The Secretary Of The Army Sustained release hydrophobic bioactive PLGA microspheres
US6117455A (en) * 1994-09-30 2000-09-12 Takeda Chemical Industries, Ltd. Sustained-release microcapsule of amorphous water-soluble pharmaceutical active agent
US5922253A (en) * 1995-05-18 1999-07-13 Alkermes Controlled Therapeutics, Inc. Production scale method of forming microparticles
FR2744367B1 (en) * 1996-02-02 1999-07-09 Emperaire Jean Claude MEDICATIONS FOR OVULATION TRIGGERING
JP2001522812A (en) * 1997-11-07 2001-11-20 カイロン コーポレイション Methods of making IGF-1 sustained release formulations
CA2335460A1 (en) * 1998-06-18 1999-12-23 Johns Hopkins University School Of Medicine Polymers for delivery of nucleic acids
JP2003501375A (en) * 1999-06-04 2003-01-14 アルザ・コーポレーション Implantable gel composition and method of manufacture
US6284283B1 (en) * 1999-10-21 2001-09-04 Alkermes Controlled Therapeutics, Inc. Method of producing sub-micron particles of biologically active agents and uses thereof

Also Published As

Publication number Publication date
JP2005517012A (en) 2005-06-09
WO2003066585A2 (en) 2003-08-14
ZA200405852B (en) 2005-01-24
AU2003217367B2 (en) 2005-09-08
AU2003217367A1 (en) 2003-09-02
EP1471891A4 (en) 2007-04-11
IL163218A (en) 2009-11-18
US20040028733A1 (en) 2004-02-12
EP1471891A2 (en) 2004-11-03
CA2474698A1 (en) 2003-08-14
NZ535008A (en) 2005-09-30
WO2003066585A3 (en) 2004-03-11

Similar Documents

Publication Publication Date Title
CA2474698C (en) Polymer-based compositions for sustained release
JP3698721B2 (en) Excipient stabilization of polypeptides treated with organic solvents
KR100858856B1 (en) Stable non-aqueous single phase viscous vehicles and formulations utilizing such vehicles
US7655254B2 (en) Implantable device for continuous delivery of interferon
US20170319662A1 (en) Stable non-aqueous single phase viscous vehicles and formulations utilizing such vehicles
US8637077B2 (en) Sustained-release preparation
US20080226689A1 (en) Stable non-aqueous single phase viscous vehicles and formulations utilizing such vehicles
US20090142399A1 (en) Dispersant agent for sustained-release preparations
US20080112994A1 (en) Formulations having increased stability during transition from hydrophobic vehicle to hydrophilic medium
CA2154164C (en) Excipient stabilization of polypeptides treated with organic solvents

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20140207