CA2730391C - Delivery of dry formulations of octreotide - Google Patents

Delivery of dry formulations of octreotide Download PDF

Info

Publication number
CA2730391C
CA2730391C CA2730391A CA2730391A CA2730391C CA 2730391 C CA2730391 C CA 2730391C CA 2730391 A CA2730391 A CA 2730391A CA 2730391 A CA2730391 A CA 2730391A CA 2730391 C CA2730391 C CA 2730391C
Authority
CA
Canada
Prior art keywords
octreotide
subject
implantable device
months
formulation
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
CA2730391A
Other languages
French (fr)
Other versions
CA2730391A1 (en
Inventor
Petr Kuzma
Stefanie Decker
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.)
Endo Pharmaceuticals Solutions Inc
Original Assignee
Endo Pharmaceuticals Solutions 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 Endo Pharmaceuticals Solutions Inc filed Critical Endo Pharmaceuticals Solutions Inc
Publication of CA2730391A1 publication Critical patent/CA2730391A1/en
Application granted granted Critical
Publication of CA2730391C publication Critical patent/CA2730391C/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/12Cyclic peptides, e.g. bacitracins; Polymyxins; Gramicidins S, C; Tyrocidins A, B or C
    • 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
    • A61K9/0024Solid, semi-solid or solidifying implants, which are implanted or injected in body tissue
    • 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
    • A61K38/31Somatostatins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/32Macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. carbomers, poly(meth)acrylates, or polyvinyl pyrrolidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/34Macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyesters, polyamino acids, polysiloxanes, polyphosphazines, copolymers of polyalkylene glycol or poloxamers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • A61P5/06Drugs for disorders of the endocrine system of the anterior pituitary hormones, e.g. TSH, ACTH, FSH, LH, PRL, GH
    • A61P5/08Drugs for disorders of the endocrine system of the anterior pituitary hormones, e.g. TSH, ACTH, FSH, LH, PRL, GH for decreasing, blocking or antagonising the activity of the anterior pituitary hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives

Abstract

Methods and devices are described for delivering octreotide to a patient, comprising implanting a controlled release composition for delivering octreotide, wherein the composition does not require hydration prior to implantation, and wherein the composition optionally comprises a release agent.

Description

t DELIVERY OF DRY FORMULATIONS OF OCTREOTIDE
BACKGROUND
Acrornegaly is a hormonal disorder that results when the pituitary gland produces excess growth hormone (GH). It most commonly affects middle-aged adults and can result in serious illness and premature death. Once diagnosed, acromegaly is treatable in most patients, but because of its slow and often insidious onset, it frequently is not diagnosed correctly. The most serious health consequences of acromegaly are diabetes mellitus, hypertension and increased risk of cardiovascular disease. Patients with acromegaly are also at increased risk for polyps of the colon that can develop into cancer. When Gil-producing tumors occur in childhood, the disease that results is called gigantism rather than acromegaly.
Fusion of the growth plates of the long bones occurs after puberty so that development of excessive GH production in adults does not result in increased height. Prolonged exposure to excess GH before fusion of the growth plates causes increased growth of the long bones and increased height.
Acromegaly is caused by prolonged overproduction of growth hormone (OH) by the pituitary gland. The pituitary is a small gland at the base of the brain that produces several important hormones to control body functions such as growth and development, reproduction, and metabolism. GH is part of a cascade of hormones that, as the name implies, regulates the physical growth of the body.
This cascade begins in a part of the brain called the hypothalamus, which makes hormones that regulate the pituitary. One of these, growth hormone-releasing hormone (GHRH), stimulates the pituitary gland to produce GH. Another hypothalamic hormone, somatostatin, inhibits GH production and release.
Secretion of GH by the pituitary into the bloodstream causes the production of another hormone, called insulin-like growth factor 1 (IGF-1), in the liver. IGF-1 is the factor that causes the growth of bones and other tissues of the body. IGF-1, in turn, signals the pituitary to reduce GH production. GHRH, somatostatin, GH and IGF-1 levels in the body are tightly regulated by each other, and their levels are influenced by environmental stimuli such as sleep, exercise, stress, food intake and blood sugar levels. If the pituitary produces GH independent from the normal regulatory mechanisms, the level of IGF-1 would rise, leading to bone growth and organ enlargement. Excess GH also causes changes in sugar and lipid metabolism and can cause diabetes.
In over 90% of acromegaly patients, the overproduction of GH is caused by a benign tumor of the pituitary gland, called an adenoma. These tumors produce excess GH and, as they expand, compress surrounding brain tissues, such as the optic nerves. This expansion causes the headaches and visual disturbances that are often symptoms of acromegaly. In addition, compression of the surrounding normal pituitary tissue can alter production of other hormones, leading to changes in menstruation and breast discharge in women and impotence in men.
In some patients, acromegaly is caused not by pituitary tumors but by tumors of the pancreas, lungs and adrenal glands. These tumors lead to an excess of GH, either because they produce GH themselves or, more frequently, because they produce GHRH, the hormone that stimulates the pituitary to make GH. In these patients, the excess GHRH can be measured in the blood and establishes that the cause of the acromegaly is not due to a pituitary defect. When these non-pituitary tumors are surgically removed, GH levels fall and the symptoms of acromegaly improve.
Acromegaly treatment regimens include reducing GH production to normal levels to relieve the pressure that the growing pituitary tumor exerts on the surrounding brain areas, to preserve normal pituitary function, and to reverse or ameliorate the symptoms of acromegaly. Treatment options include surgical removal of the tumor, drug therapy and radiation therapy of the pituitary.
Octreotide has been demonstrated to be effective in the management of acromegaly. GH levels usually decrease within two hours following a subcutaneous octreotide injection. Octreotide results in a decrease in GH and IGF-1 levels in a majority of patients with normalization of IGF-1 levels in up to 60% of patients, indicating biochemical remission. Most patients note a marked improvement in their symptoms of acromegaly including headaches, joint pains and diaphoresis very soon after starting octreotide therapy.
Octreotide is currently available as Sandostatin LAR Depot, which is, upon reconstitution, a suspension of
-2-microspheres containing octreotide acetate. Sandostatin LAR Depot is the only medication indicated for the long-term maintenance therapy in acromegalic patients.
It is also indicated for the long-term treatment of severe diarrhea and flushing episodes associated with metastatic carcinoid tumors and profuse water diarrhea associated with VIP-secreting tumors. Sandostatin LAR Depot is administered via intramuscular injection every four weeks, following a titration period.
Octreotide acetate has also been available in an immediate-release formulation, Sandostatin Injection solution, which is required to be administered by injection three times daily.
In patients who do not have a significant reduction in GH levels in response to intermittent octreotide injections, more frequent dosing of octreotide may result in a greater clinical response. Octreotide may be administered continuously by a subcutaneous pump to patients with refractory acromegaly to prevent escape of GH
between injections.
In light of the efficacy of octreotide for treating acromegaly and lack of a controlled-release treatment method and formulation of octreotide, there is a clear need for a formulation and delivery method that can deliver octreotide over a period of time at a controlled rate to avoid the complications of a patient's having to suffer, for example, multiple periodic injections.
SUMMARY
The present invention relates generally to an octreotide pharmaceutical composition that can be used to treat individuals affected with hormonal disorders.
The present invention is preferably formulated as a controlled-release formulation.
In particular, the present invention is based on the unexpected discovery that octreotide can be released at a controlled rate using an implantable device, e.g., an implantable device that does not require priming prior to implantation.
One embodiment is directed to a method of delivering octreotide to a subject with a substantially zero-order release profile over an extended period of time, but no less than about six months, wherein the subject is a mammal that is not a dog, the method comprising subcutaneously implanting in the subject at least one implantable device, wherein the at least one implantable device comprises a composition comprising octreotide, wherein the composition is encased in a
-3-hydrophilic polymer, and wherein the implantable device is implanted in a dry state, such that the subject receives on a daily basis over a period of at least about six months dose amounts of octreotide, which are effective to treat the subject.
In a particular embodiment, the hydrophilic polymer comprises one or more polyurethane-based polymers or one or more methacrylate-based polymers. In a particular embodiment, the octreotide is in free form, salt form or in the form of a complex thereof, e.g., wherein the octreotide is octreotide acetate. In a particular embodiment, the subject is afflicted with a GH or IGF-1 hormone disorder or its symptoms, e.g., acromegaly. In a particular embodiment, the subject receives octreotide at an average rate ranging from about 75 ,ug per day to about 300 ,ug per day over a period of at least about six months. In a particular embodiment, the dose amounts of octreotide received by the subject result in octreotide serum levels ranging from about 0.5 ng/mL to about 2 ng/mL, from about from about 0.5 ng/mL

to about 1.4 ng/mL, from about 0.6 ng/mL to about 1.2 ng/mL, from about 0.8 ng/mL to about 1.2 ng/mL or from about 0.9 ng/mL to about 1.0 ng/mL. In a particular embodiment, the subject receives an effective amount of octreotide for a period of at least about twelve months. For the purposes of determining serum levels, the range can be indicated as an average over a period of time, e.g., from about 3 days to about 150 days, from about 3 days to about 120 days, from about 5 days to about 100 days, from about 10 days to about 75 days, etc. In a particular embodiment, the dose amounts of octreotide received by the subject result in Cõ,,õ
for octreotide serum levels below about 1.2 ng/mL. In a particular embodiment, the dose amounts of octreotide received by the subject result in Cmax for octreotide serum levels below about 1.0 ng/mL. In a particular embodiment, release of octreotide occurs at least three to about ten days after implantation. In a particular embodiment, the subject is afflicted with a condition selected from the group consisting of: carcinoid syndrome, VIPomas, neuroendocrine tumors, proliferative diabetic retinopathy, rosacea, pancreatitis, gastrointestinal bleeding, pancreatic and intestinal fistulas, Graves-Basedow ophthalmopathy, glaucoma, and symptoms associated with chemotherapy or AIDS.
-4-One embodiment is directed to an implantable device, comprising a controlled-release formulation comprising octreotide substantially encased by a polyurethane-based or methacrylate-based polymer, wherein the implantable device delivers octreotide to a subject with a substantially zero-order release profile over an extended period of time, but no less than about six months, wherein the subject is a mammal that is not a dog, when the implantable device is implanted in a dry state into the subject. In a particular embodiment, the hydrophilic polymer comprises one or more polyurethane-based polymers. In a particular embodiment, the octreotide is in free form, salt form or in the form of a complex thereof, e.g., octreotide acetate.
In a particular embodiment, when the implantable device is implanted into the subject, the subject receives octreotide at an average rate ranging from about 75 ,ug per day to about 300 pg per day over a period of at least about six months. In a particular embodiment, when the implantable device is implanted into the subject, the dose amounts of octreotide received by the subject result in octreotide serum levels ranging from about 0.5 ng/mL to about 2 ng/mL. In a particular embodiment, when the implantable device is implanted into the subject, the dose amounts of octreotide received by the subject result in octreotide serum levels ranging from about 0.8 ng/mL to about 1.8 ng/mL. In a particular embodiment, when the implantable device is implanted into the subject, the dose amounts of octreotide received by the subject result in Cmax for octreotide serum levels below about 1.2 ng/mL. In a particular embodiment, when the implantable device is implanted into the subject, the dose amounts of octreotide received by the subject result in C.
for octreotide serum levels below about 1.0 ng/mL. In a particular embodiment, when the implantable device is implanted into the subject, release of octreotide occurs at least three to about ten days after implantation.
One embodiment is directed to the use of octreotide for the manufacture of a medicament for treating a mammalian subject that is not a dog, wherein the medicament is a controlled-release formulation substantially encased by a polyurethane-based or methacrylate-based polymer, thereby forming an implantable device, wherein, upon implantation into the subject, the implantable device delivers octreotide to the subject with a substantially zero-order release profile over an extended period of time, but no less than about six months, wherein the subject is a
-5-
6 PCT/US2009/050215 mammal that is not a dog, when the implantable device is implanted in a dry state into the subject.
The present invention provides a therapeutically-effective amount of octreotide over an extended period of time, preferably at least about two months, more preferably about six months and up to about two years. The present invention also provides compositions that provide controlled-release of octreotide over at least about two months, preferably about six months, and up to about two years.
Embodiments of the present invention relate to a pharmaceutical composition comprising octreotide or salts, prodrugs or derivatives thereof, which can be used in the effective treatment of various diseases and conditions.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 is a graph showing the linear relationship between the equilibrium water content vs. the weight percent content of hydroxypropyl methacrylate (HPMA) units in crosslinked HEMA/HPMA polymers at their maximum state of hydration.
FIG. 2 is a graph showing the release of octreotide from an implant formulation of the present invention.
FIG. 3 is a graph showing the release of octreotide from an implant formulation of the present invention.
FIG. 4 is a graph showing the release of octreotide from six different implant formulations of the present invention.
FIG. 5 is a graph showing the release of octreotide from different implant formulations of the present invention.
FIG. 6 is a graph showing octreotide and IGF-1 serum levels in a healthy dog implanted with an octreotide formulation of the present invention.
FIG. 7 is a graph showing octreotide and IGF-1 serum levels in a group of three healthy dogs implanted with one octreotide implant formulation of the present invention over a six month period.
FIG. 8 is a graph showing octreotide and IGF-1 serum levels in a group of three healthy dogs implanted with two octreotide implant formulations of the present invention over a six month period.

FIGS. 9A and 9B are graphs depicting the IGF-1 serum level and percent change in eleven human subjects with acromegaly over six months implanted with an octreotide formulation of the present invention, respectively.
FIG. 10 is a graph depicting octreotide serum levels in eleven human subjects with acromegaly over six months implanted with an octreotide formulation of the present invention.
FIG. 11 is a graph depicting octreotide serum levels in two dogs over six months implanted with an octreotide formulation of the present invention.
FIG. 12 is a graph depicting IGF-1 serum levels in two dogs over six months io implanted with an octreotide formulation of the present invention.
FIG. 13 is a graph showing serum octreotide levels after hydrated implant delivery and dry implant delivery (see also Table 6).
FIG. 14 is a graph showing serum octreotide levels after hydrated implant delivery and dry implant delivery (see also Table 6).
FIG. 15 are graphs showing the level of growth hormone after delivery of octreotide by hydrated and dry implants (GH concentration, upper panel; % GH
decrease, bottom panel).
FIG. 16 are graphs showing the level of insulin-like growth factor 1 (IGF-1) after delivery of octreotide by hydrated and dry implants (IGF-1 concentration, upper panel; standard deviation, bottom panel).
FIG. 17 are graphs showing the level of insulin-like growth factor 1 (IGF-1) after delivery of octreotide by hydrated and dry implants (both panels show data from studies with values expressed as the percent of normal IGF-1 levels).
DETAILED DESCRIPTION
Before the present compositions and methods are described, it is to be understood that this invention is not limited to the particular molecules, compositions, methodologies or protocols described, as these may vary. It is also to be understood that the terminology used in the description is for the purpose of describing the particular versions or embodiments only, and is not intended to limit the scope of the present invention which will be limited only by the appended claims. The terms used herein have meanings recognized and known to those of
-7-skill in the art, however, for convenience and completeness, particular terms and their meanings are set forth below.
The singular forms "a", "an", and "the" include plural reference unless the context clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of embodiments of the present invention, the preferred methods, devices, and materials are now described. Nothing herein is to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue of prior invention.
As used herein, the term "about: means plus or minus 10% of the numerical value of the number with which it is being used. For example, about 50% means in the range of 45%-55%.
"Controlled-release formulation" refers to a formulation designed to consistently release a predetermined, therapeutically-effective amount of drug or other active agent such as a polypeptide or a synthetic compound over an extended period of time, with the result being a reduction in the number of treatments necessary to achieve the desired therapeutic effect. A controlled-release formulation decreases the number of treatments necessary to achieve a desired effect in terms of decreased growth hormone levels or decreased IGF-1 levels, or an improvement in symptoms associated with, for example, acromegaly including but not limited to abnormal growth, carcinoid syndrome, VIPomas (Vasoactive Intestinal Peptide Secreting Adenomas), neuroendocrine tumors (specifically treating the symptoms of flushing and diarrhea), proliferative diabetic retinopathy, rosacea, pancreatitis, gastrointestinal bleeding, pancreatic and intestinal fistulas, Graves-Basedow ophthalmopathy, glaucoma, or treating symptoms of chemotherapy and AIDS. The controlled-release formulations of the present invention achieve a desired pharmacokinetic profile in a subject, preferably commencement of the release of the active agent substantially immediately after placement in a delivery environment,
-8-followed by consistent, sustained, preferably zero-order or near zero-order release of the active agent.
As used herein, the term "controlled-release" includes the predetermined, consistent release of active agent from the dosage formulation at a rate such that a therapeutically beneficial blood level below toxic levels of the active agent is maintained over a period, for example, of at least about two months, about six months or more (e.g., up to about two years).
The terms "patient" and "subject" mean all animals including humans.
Examples of patients or subjects include humans, cows, dogs, cats, goats, sheep, and pigs.
The term "pharmaceutically acceptable salts, esters, amides, and prodrugs"
as used herein refers to those carboxylate salts, amino acid addition salts, esters, amides, and prodrugs of the compounds of the present invention that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of patients without undue toxicity, irritation, allergic response and the like.
Their use is commensurate with a reasonable benefit/risk ratio, and is effective for their intended use. Zwitterionic forms, where possible, are also useful compounds of the invention. The compounds of the present invention additionally can exist, for example, in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as, for example, water, ethanol and the like. In general, the solvated forms are considered equivalent to the unsolvated forms for the purposes of the present invention.
The term "prodrug" refers to compounds that are rapidly transformed in vivo to yield the parent compounds of the above formula, for example, by hydrolysis in blood. A thorough discussion is provided in T. Higuchi and V. Stella, "Pro-drags as Novel Delivery Systems," Vol. 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987.
The term "salts" refers to the relatively non-toxic, inorganic and organic acid addition salts of compounds of the present invention. These salts can be prepared in situ during the final isolation and purification of the compounds or by separately
-9-reacting the purified compound in its free base form with a suitable organic or inorganic acid and isolating the salt thus formed. Representative salts include the acetate, hydrobromide, hydrochloride, sulfate, bisulfate, nitrate, acetate, oxalate, valerate, oleate, palmitate, stearate, laurate, borate, benzoate, lactate, phosphate, s tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthylate mesylate, glucoheptonate, lactobionate and laurylsulphonate salts, and the like. These may include cations based on the alkali and alkaline earth metals, such as sodium, lithium, potassium, calcium, magnesium, and the like, as well as non-toxic ammonium, tetramethylarnmonium, tetraethylammonium, methyl amine, dimethylamine, trimethylamine, triethylamine, ethylamine and the like (See, for example, S. M. Barge et al., "Pharmaceutical Salts," J. Pharm. Sc., 1977, 66:1-19).
"Treatment" refers to the administration of medicine or the performance of medical procedures with respect to a patient, either for prophylaxis (prevention) or to cure the infirmity or malady in the instance where the patient is afflicted.
A "therapeutically effective amount" is an amount sufficient to decrease, prevent or ameliorate the symptoms associated with a medical condition. In the context of hormonal therapy it can also mean an amount sufficient to normalize body functions or hormone levels in disease or disorders. For example, a therapeutically effective amount of a controlled-release formulation of octreotide is a predetermined amount calculated to achieve the desired effect, e.g., to effectively decrease growth hormone or IGF-1 levels in a patient.
The present invention can be utilized to treat a variety of hormonal disorders, including, for example, acromegaly and gigantism, or other diseases, disorders or symptoms that are effectively treated with, for example, octreotide, e.g., carcinoid syndrome, VIPomas (Vasoactive Intestinal Peptide Secreting Adenomas), neuroendocrine tumors (specifically treating the symptoms of flushing and diarrhea), proliferative diabetic retinopathy, rosacea, pancreatitis, gastrointestinal bleeding, pancreatic and intestinal fistulas, Graves-Basedow ophthalmopathy, glaucoma, or so treating symptoms of chemotherapy and AIDS.
-10-Acromegaly is characterized by a number of clinical features including enlargement of the hands and feet, facial changes including frontal bossing, enlarged mandible and increased dental spacing, arthralgias, diaphoresis, sleep apnea, hypertension, diabetes mellitus and hypertrophic cardiomyopathy. Tumors that cause acromegaly frequently cause local anatomic compression, resulting in, for example, visual field deficits, headaches, hypopituitarism, and cranial nerve palsies.
There is a 2 to 5 fold increase in the mortality rate in acromegalic patients largely due to cardiovascular and cerebrovascular disease. There is also an increased rate of malignancy associated with acromegaly, with colon cancer the best characterized.
Carcinoid tumors usually appear in the appendix, bronchial tubes, colon, or small intestine and secrete chemicals that cause the dilation of blood vessels-such as serotonin. Vasodilation may be responsible for the symptoms usually observed with Carcinoid tumors¨ such as, for example, diarrhea, flushing and asthma.
Depending on the hormones and biochemicals secreted by carcinoid tumors a number of symptoms can be present. These are collectively known as "Carcinoid Syndrome".
Biochemically, people with Carcinoid tumors tend to produce more serotonin, using the amino acid tryptophan as a base- serotonin is further broken down in the body to product 5- hydroxy indole acetic acid (5-HIAA) which is seen in the urine of the majority of such patients. Diagnostic tests on blood and urine would show a patient with a Carcinoid tumor- exhibits elevated urinary 5-HIAA, low blood tryptophan, high blood chromogranin A, and serotonin. Blood tests are also used to levels of histamine, bradykinin, neurone-specific enolase, calcitonin, Substance-P, neurokinin-A, and pancreatic polypeptide.
An "OctreoScan" is a scanning test used to identify carcinoid tumors and neuroendocrine tumors. This scan utilizes a radioactive octreotide derivative called pentetreotide. Post-injection, this concentrates in tissues expressing the somatostatin receptor. Ne-uroendocrine tumors over-express the receptor and are imaged using this test.
As used herein, the term "octreotide" refers generally to all compounds comprising the structure as shown, including various salt forms. Octreotide comprises an octapeptide with the following amino acid sequence: L-cysteinamide, D-phenylalanyl-L-cysteiny-L-phenylalanyl-D-tryptophyl-L-lysyl-L-threonyl-- N-[2-
-11-hydro xy-1 -(hydroxymethyl)propyl] -,cycli c(2 4 7)-di sul fide ; [R--(R*,R*)]. The structure of octreotide is shown below.

Si 401 S
-:

H I H - H I H

H N _0- I
H N,- N 1 0 0 0 0 _ S ---- H3C 'OH H3C 'OH
N

Octreotide The chemical formula is C49H66N10010S2 and its molecular weight is 1019.3 Da. Its therapeutic category is gastric anti-secretory agent. The octreotide of the present invention can exist in, for example, a free form, a salt form or in the form of complexes thereof. Acid addition salts can be formed with, for example, organic acids, polymeric acids and inorganic acids. Acid addition salts include, for example, the hydrochloride and acetates. Complexes are formed, for example, from octreotide on addition of inorganic substances, e.g., inorganic salts or hydroxides such as Ca- and Zn-salts and/or addition of polymeric organic substances. The acetate salt is the preferred salt for formulations of the present invention.
Embodiments of the present invention provide a drug delivery device that can achieve the following objectives: a controlled-release rate (zero or about zero order release rate) to maximize therapeutic effects and minimize unwanted side effects; a convenient way to retrieve the device if it is necessary to end the treatment; and an increase in bioavailability with less variation in absorption and no first pass metabolism.
The controlled-release pharmaceutical composition comprising octreotide acetate can be part of a controlled-release hydrogel device. The composition of the present invention is capable of providing, upon administration to a patient, a release profile of octreotide extending over at least about two months, preferably at least about six months or more, e.g., up to about two years. Octreotide can be contained
-12-within the hydrogel, for example, and the formulation releases a therapeutically effective amount of octreotide over an extended period of time. The hydrogel can comprise a polymer selected from methacrylate-based polymers, polyurethane-based polymers and combinations thereof. A therapeutically effective amount is an amount of octreotide, preferably octreotide acetate, that when administered to a patient or subject, ameliorates a symptom of acromegaly. The formulation can further include pharmaceutically acceptable excipients.
When the compositions of the present invention are administered to a patient, the concentration of octreotide in the patient's plasma over time (release profile) can extend over a period of at least about two months, preferably about six months, and up to about two years. The compositions can provide a mean plasma concentration at steady state of octreotide in a human patient of from about 0.1 to about 9 ng/ml, about 5 ng/ml to about 1 ng/ml, about 1 to about 2 ng/ml, or about 1.2 to about 1.6 ng/ml. Steady state is the point at which the amount of drug administered over a dosing interval equals the amount of drug being eliminated over that same period.
The hydrophilic implant comprising the octreotide formulation can be formed from a xerogel such that it readily absorbs water. In a hydrated state, the xerogel is referred to as a hydrogel. In either form, hydrated or unhydrated, it is biocompatible and non-toxic to the host and non-biodegradable. It is water-swellable and water-insoluble. When the hydrogel attains its maximum level of hydration, the water content of the hydrogel is referred to as "equilibrium water content" (EWC). The percent water content of the hydrogel (any state of hydration) is determined as follows:
weight of hydrogel ¨ weight of dry polymer (xerogel) x 100 weight of hydrogel The hydrogel can be a homogeneous homopolymer or copolymer having a predetermined equilibrium water content (EWC) value formed by the polymerization of a mixture of ethylenically unsaturated monomer A and ethylenically unsaturated monomer B, for example, 2-hydroxyethyl methacrylate (HEMA) and hydroxypropyl methacrylate (HPMA). The predetermined EWC can be calculated by determining the EWC values of the hydrogel homopolymer of hydrophilic monomer A (homopolymer A) and the hydrogel homopolymer of
-13-hydrophilic monomer B (homopolymer B); determining the relationship of the EWC

values of the homogeneous copolymers AB versus the chemical composition of said copolymers AB; selecting the targeted EWC value and determining the chemical composition of copolymer AB having the targeted EWC value; forming a polymerizable mixture of monomer A and monomer B in amounts sufficient to yield copolymer AB having the targeted EWC value; and effect the polymerization reaction to yield copolymer AB characterized by the targeted EWC value.
As used herein, "copolymer AB" or "copolymer AB consisting essentially of monomer A units and monomer B units" means that the addition copolymerization of monomer A and monomer B has been effected through the polymerizable ethylenic bond of the monomers. By way of illustration, if monomer A is 2-hydroxyethyl methacrylate and monomer B is N-methylacrylamide, copolymer AB
contains recurring monomer A units and recurring monomer B units.
Unless the context indicates otherwise, the term "copolymer" includes polymers made by polymerizing a mixture of at least two ethylenically unsaturated monomers.
As used herein, "HEMA unit(s)" refer to a structure recurring in the polymer obtained by polymerizing hydrophilic material containing 2-hydroxyethyl methacrylate ("HEMA"). By the term "HEMA unit(s)" is meant the structure:

C=0 C2H.40H
As used herein, "HPMA unit(s)" refers to a structure obtained by polymerizing hydrophilic material containing hydroxypropyl methacrylate ("HPMA"). By the term "HPMA unit(s)" is meant the structure:
-14-C ______________________________ CH2 C = 0 Liquid polymerizable material useful in the hydrophilic products include a wide variety of polymerizable hydrophilic, ethylenically unsaturated compounds, in particular, hydrophilic monomers such as, for example, the monoester of an acrylic acid or methacrylic acid with a polyhydroxy compound having an esterifiable hydroxyl group and at least one additional hydroxyl group such as, for example, the monoalkylene and polyalkylene polyols of methacrylic acid and acrylic acid, e.g., 2-hydroxyethyl methacrylate and acrylate, diethylene glycol methacrylate and acrylate, propylene glycol methacrylate and acrylate, dipropylene glycol methacrylate and acrylate, glycidyl methacrylate and acrylate, glyceryl methacrylate o and acrylate, and the like; the 2-alkenamides, e.g., acrylamide, methacrylamide, and the like; the N-alkyl and N,N-dialkyl substituted acrylamides and methacrylamides such as N-methylmethacrylamide, N,N-dimethylmethacrylamide, and the like; N-vinylpyrrolidone; the alkyl-substituted N-vinylpyrrolidones, e.g., methyl substituted N-vinylpyrrolidone; N-vinylcaprolactam; the alkyl-substituted N-vinylcaprolactam, e.g., N-vinyl-2-methylcaprolactam, N-vinyl-3,5-dimethylcaprolactam, and the like.
Acrylic and methacrylic acid can also be useful in these formulations.
Mixtures of hydrophilic monomers are employed in the polymerization reaction. The type and proportion of monomers are selected to yield a homogeneous polymer, preferably a crosslinked homogeneous polymer, which on hydration possesses the desired EWC value for the contemplated application or use. This value can be predetermined by preparing a series of copolymers using different monomer ratios, e.g., mixtures of HEMA and HPMA of varying ratios, ascertaining the EWC values of the copolymers, and plotting the relationship of % HPMA (or %
-15-HEMA) units in the HPMA/HEMA copolymers versus weight percent EWC of the copolymers (FIG. 1).
In some instances the polymerization of certain hydrophilic monomeric mixtures results in homogeneous hydrophilic copolymers that dissolve, to a varying extent, in an aqueous medium. In such cases, a small amount, e.g., up to 3 percent, of a copolymerizable polyethylenically unsaturated crosslinking agent, can be included in the monomeric mixture to obtain homogeneous crosslinked copolymers that are water-insoluble as well as water-swellable.
Slightly crosslinked homopolymers of HEMA can have an EWC value of, for example, about 38%.
Crosslinked copolymers of HEMA and HPMA have EWC values below about 38%.
On the other hand, crosslinked copolymers of HEMA and acrylamide exhibit EWC
values above 38 % (w/v), e.g., upwards to approximately 75 %, and higher.
Therefore, depending on the useful or effective elution rate of the active compound, e.g., drug, that is required of a hydrogel delivery system for a particular application, one skilled in the art, by following the teachings disclosed herein, can tailor copolymer hydrogel membranes to elute the drug at a desired rate. Copolymers can contain, for example, about 15% to about 70 % (weight) of HEMA units and from about 85 to 30 % (weight) of units of a second ethylenic monomer and possess predetermined EWC values in the range of from about 20% to about 75%, preferably about 25%. Homogenous copolymers can include those made from hydrophilic monomeric mixtures containing from about 80 % HPMA (weight), and from about 20 % HEMA (weight). In further embodiments, the mixture can further contain a small amount of a polyethylenically unsaturated crosslinking agent, e.g., trimethylolpropane trimethacrylate ("TMPTMA").
Various aspects of the invention include homogeneous hydrophilic copolymers whose homogeneous polymer structure is formed by the polymerization of a mixture of hydrophilic monomers described previously; and the drug delivery device that utilizes the homogeneous polymer cartridges in the delivery system. The polymerization of a mixture of hydrophilic monomers and hydrophobic monomers yields heterogeneous polymers. Where hydrophobic segments are present in the polymer, the interfacial free energy increases, thus enhancing protein adsorption and mineralization after implantation in an animal. Hydrogels of poly-HEMA, for
-16-example, were measured to have interfacial free energy close to zero.
According to the interfacial free energy interpretation, hydrogels of strictly hydrophilic components would strongly appear to be biocompatible with body tissue.
Slightly crosslinked poly-HEMA is a homogeneous, hydrophilic "homopolymer"
(disregarding the relatively small quantities of polymerized crosslinking agent therein) of relatively fixed characteristics or values. Techniques for altering the "homopolymer" poly-HEMA to impart to it additional characteristics or properties are difficult, time-consuming, and oftentimes result in erratic property behavior. On the other hand, mixtures of HEMA with varying quantities of other polymerizable hydrophilic comonomer(s) can be polymerized to give predictable homogeneous hydrophilic copolymers having (predetermined) tailor-made properties.
Useful crosslinking agents that can be included in the polymerizable reaction medium include, for example, the polyethylenically unsaturated compounds having at least two polymerizable ethylenic sites, such as the di-, tri- and tetra-ethylenically unsaturated compounds, in particular, the tri-unsaturated crosslinking agents with/without the di-unsaturated crosslinking compounds, for example, divinylbenzene, ethylene glycol dimethacrylate and diacrylate, propylene glycol dimethacrylate and diacrylate; and the di-, tri- and tetra-acrylate or methacrylate esters of the following polyols: triethanolamine, glycerol, pentaerythritol, 1,1,1-trimethylolpropane and others.
The polymerization reaction can be carried out in bulk or with an inert solvent. Suitable solvents include, for example, water; organic solvents (e.g., water-soluble lower aliphatic monohydric alcohols as well as polyhydric alcohols, e.g., glycol, glycerine, dioxane, etc.; and mixtures thereof).
Compounds useful in the catalysis of the polymerizable ethylenically unsaturated compounds include the free-radical compounds and/or initiators of the type commonly used in vinyl polymerization such as the organic peroxides, percarbonates, hydrogen peroxides, and alkali metal sulfates. Illustrative examples include cumene hydroperoxide, t-butyl hydroperoxide, benzoyl peroxide, bis(4-t-butylcyclohexyl) peroxydicarbonate, hydrogen peroxide, 2,4-dichlorobenzoyl peroxide, acetyl peroxide, di-n-propyl peroxydicarbonate, di-t-butyl peroxide, di-sec-butyl peroxydicarbonate, ammonium sulfate, potassium sulfate, and sodium
-17-sulfate. A preferred catalyst is one that is effective at moderately low temperature such as, for example, at about 20-80 C (e.g., tert-butyl peroctoate, benzoyl peroxide, and di(secbutyl) peroxydicarbonate).
A conventional redox polymerization catalyst can also be employed.
Polymerization of the ethylenic compounds can be effected, for example, using radiation, e.g., ultraviolet, X-Ray, gamma radiation, microwave or other well-known forms of radiation. A preferred catalyst for ultraviolet cure is benzoin methyl ether.
Catalysts and/or initiators and/or radiation are employed in a catalytically-effective amount to optimize the polymerization reaction.
The current invention focuses on the application of polyurethane based polymers, thermoplastics or thermosets, to the creation of implantable drug devices to deliver biologically active compounds at controlled rates for prolonged period of time. Polyurethane polymers are preferably made into cylindrical hollow tubes with one or two open ends through extrusion, (reaction) injection molding, compression molding, or spin-casting (see, e.g., U.S. Pat. Nos. 5,266,325 and 5,292,515), depending on the type of polyurethane used.
Thermoplastic polyurethane can be processed through extrusion, injection molding or compression molding. Thermoset polyurethane can be processed through reaction injection molding, compression molding or spin-casting. The dimensions of the cylindrical hollow tube are determinable and can be adjusted precisely.
Polyurethane based polymers are synthesized from multi-functional polyols, isocyanates and chain extenders. The characteristics of each polyurethane can be attributed to its structure.
Thermoplastic polyurethanes are made of macrodiols, diisocyanates and difunctional chain extenders (e.g., U.S. Pat. Nos. 4,523,005 and 5,254,662).
Macrodiols make up the soft domains.
Diisocyanates and chain extenders make up the hard domains. The hard domains serve as physical crosslinlcing sites for the polymers. Varying the ratio of these two domains can alter the physical characteristics of the polyurethanes.
-18-Thermoset polyurethanes can be made of multifunctional (greater than difunctional) polyols and/or isocyanates and/or chain extenders (e.g., U.S.
Pat. Nos.
4,386,039 and 4,131,604).
Thermoset polyurethanes can also be made by introducing unsaturated bonds in the polymer chains and appropriate crosslinkers and/or initiators to do the chemical crosslinking (e.g., U.S. Pat. No. 4,751,133).
By controlling the amounts of crosslinking sites and how they are distributed, the release rates of the actives can be controlled.
Different functional groups can be introduced into the polyurethane polymer chains through the modification of the backbones of polyols depending on the properties desired. Where the device is used for the delivery of water soluble drugs, hydrophilic pendant groups such as ionic, carboxyl, ether, and hydroxy groups are incorporated into the polyols to increase the hydrophilicity of the polymer (e.g., U.S.
Pat. Nos. 4,743,673 and 5,354,835). Where the device is used for the delivery of hydrophobic drugs, hydrophobic pendant groups such as alkyl, siloxane groups are incorporated into the polyols to increase the hydrophobicity of the polymer (e.g., U.S.
Pat. No. 6,313,254). The release rates of the actives can also be controlled by the hydrophilicity/hydrophobicity of the polyurethane polymers.
Small cylindrically-shaped implants of the invention can contain within their core, octreotide, preferably octreotide acetate, and optionally, a pharmaceutically acceptable carrier. The membrane thickness (between the interior and exterior surfaces) of the implant is substantially uniform, and serves as a rate-limiting barrier for the release of the contained agent. Such implants can be plasticized or hydrated and reshaped into other geometrically shaped articles for use in various medical applications.
In the manufacture of the implantable formulation, several factors can be considered. The release profile (delay time, release rate, and duration) is determined; the hydrophilic polymeric material is identified; and the diffusivity of the active agent through it (as a rate-limiting membrane) is measured. The hydration profile of the rate-limiting membrane for a given active agent may be readily
-19-determined by preparing a film of the selected polymer and subjecting it to a diffusion study, using a two compartment vertical glass cell, as is well known in the art.
The diffusion coefficient and the water content at which diffusion begins (below which substantially no diffusion occurs- hereinafter "% Ha") are determined.
A series of membranes is prepared from various polymers. The membranes are then hydrated to their capacity and their EWCs are measured. The fully hydrated membranes are placed in the two-compartment, vertical glass cells to measure and the diffusion of the macromolecular composition through the membrane materials at the various EWCs is plotted. The equilibrium water content of the most hydrated membrane through which no diffusion is detected (e.g., none of the active agent diffuses into the receptor cell) is the % Hd for the system being tested. This can be accomplished by plotting a curve of the permeability versus EWC.
The permeability results (diffusion coefficients) are obtained according to Fick's First Law of Diffusion, by use of the equation:
del = APCd dt 1 wherein dQ/dt is the flux through the membrane material (pg/hr); it is measured as the slope of the linear part of the curve of cumulative transport versus time;
wherein A is the area of the membrane (cm2); wherein P is the membrane's permeability coefficient (cm2/hr), or D1Q, wherein D is the diffusivity of the membrane (cm2/hr), and IQ is the partition coefficient for the membrane/donor solution; wherein 1 is the membrane thickness as measured at the end of the experiment (cm); and wherein Cd is the concentration of the donor solution (j.1g/cm3).
The release delay profile can then be determined. Another series of polymeric membranes can be prepared, again varying the amounts of crosslinker and monomers. These membranes are then hydrated, but only partially, e.g., to a water content less than or equal to % Hd. The partially hydrated membranes are placed in two-compartment vertical glass cells to measure and plot the diffusion of the active compound through the membranes versus time. Buffer solutions for the donor and receptor cells can be selected to contact the partially-hydrated membranes and
-20-further hydrate them at approximately the same rate at which they will hydrate in the delivery environment. The time between commencement of the diffusion study, i.e., addition of the active agent to the donor cell, and the detection of a pharmaceutically-effective concentration of the active agent in the receptor cell is the release delay time for that combination of polymer and initial percent hydration.
To determine the physical dimensions of the cylindrically-shaped device, the total amount of active agent to be delivered is determined. This is the product of the desired daily dosage and the duration of delivery. In preferred embodiments, the duration of delivery is at least about 2 months, more preferably about 6 months, and up to about two years. The desired daily dosage is, for example, about 10 to about 1000 jig of octreotide per day, preferably about 20 to about 800 jig of octreotide per day, more preferably about 30 to about 300 jig of octreotide per day.
The volume of the cylindrical reservoir (core) of a cylindrically-shaped device is equal to nr12 h wherein r, is the radius of the reservoir and h is its height.
The formula for steady state release from a cylinder is:
[dQ/dt]=[211hDKdCd]/[In (r0/r,)]
wherein ro is the outside radius of the cylindrical device; and wherein Cd is the concentration of drug in the donor solution, i.e., the carrier. Steady state release is obtained when Cd is maintained at saturation. The thickness of the membrane zo needed for the desired sustained release is, therefore, ro -r1.
The amount of active agent employed will depend not only on the desired daily dose but also on the number of days that dose level is to be maintained.
While this amount can be calculated empirically, the actual dose delivered is a function of any interaction with materials and the carrier, if employed in the device.
Once the appropriate polyurethane polymer is chosen, the best method to fabricate the cylindrically shaped implants can be determined by one of skill in the art to achieve suitable delivery characteristics as described herein.
For thermoplastic polyurethanes, precision extrusion and injection molding are can be used to produce two open-end hollow tubes with consistent physical dimensions. The reservoir can be loaded freely with appropriate formulations containing actives and carriers or filled with pre-fabricated pellets to maximize the
-21-loading of the actives. To seal the two open ends, two pre-fabricated end plugs can be used. The sealing step can be accomplished through the application of heat or solvent or any other means to seal the ends, preferably permanently.
For thermoset polyurethanes, precision reaction injection molding or spin casting is the preferred choice depending on the curing mechanism. Reaction injection molding is used if the curing mechanism is carried out through heat and spin casting is used if the curing mechanism is carried out through light and/or heat.
Preferably, hollow tubes with one open end are made by spin casting.
Preferably, hollow tubes with two open ends are made by reaction injection molding. The reservoir can be loaded in the same way as the thermoplastic polyurethanes.
Preferably, to seal an open end, an appropriate light-initiated and/or heat-initiated thermoset polyurethane formulation is used to fill the open end and this is cured with light and/or heat. More preferably, a pre-fabricated end plug can also be used to seal the open end by applying an appropriate light-initiated and/or heat-initiated thermoset polyurethane formulation on to the interface between the pre-fabricated end plug and the open end and cured it with the light and/or heat or any other means to seal the ends, preferably permanently. The solid active agent and optional carriers can be compressed into pellet form to maximize the loading of the actives.
Prior to implantation, the implantable formulations can be optionally hydrated or "primed" for a predetermined period of time. Priming can enable the active ingredient to begin to infiltrate and saturate the walls of the hydrogel and potentially begin to leach out of the hydrogel prior to implantation depending upon the amount of time the implant is primed. A primed implant will begin to release active ingredient substantially upon implantation, and may result in a peak release of the drug shortly after implantation. In contrast, little to no priming may result in substantially no release of the active ingredient upon implantation for a period of time until the implant becomes hydrated and the active ingredient begins to be released, however these priming characteristics depend on the specific formulations being used. The invention is directed to, for example, a method of administering a controlled-release octreotide formulation comprising implanting a dehydrated octreotide formulation into a subject.
-22-Depending upon the types of active ingredient, hydrophilic or hydrophobic, the appropriate conditioning and priming media will be chosen. Water based media are preferred for hydrophilic actives and oil based media are preferred for hydrophobic actives. Alternatively, certain implants of the invention do not need to be primed prior to implantation. In some instances, priming will improve delivery of the active agent in a controlled fashion, but in other instances, priming prior to implantation in a subject will not affect delivery in a way to justify the added time and hassle required for priming the implant.
The hydrating liquid useful in the practice of the invention is typically a 1 o liquid simulating the environment in which the active compound will be released, e.g., body fluid, sterile water, tear fluid, physiological saline solution, phosphate buffer solution and the like. While liquids other than water are useful as the hydrating liquid, the degree to which a hydrophilic membrane is hydrated is referred to as its "water content."
The priming and conditioning of the drug delivery devices involves the loading of the actives (drug) into the polymer that surrounds the reservoir, and thus prevent loss of the active before the actual use of the implant. The conditions used for the conditioning and priming step depend on the active agent, the temperature and the medium in which they are carried out. The conditions for the conditioning and priming can be the same in some instances.
The conditioning and priming step in the process of the preparation of the drug delivery devices is performed to obtain a determined rate of release of a specific drug. The conditioning and priming step of the implant containing a hydrophilic drug is preferably carried out in an aqueous medium, more preferably in a saline solution. For hydrophobic drugs, the medium can be a plasma-like medium, including, but not limited to, cyclodextrin. The conditioning and priming steps are carried out by controlling three specific factors, namely the temperature, the medium and the period of time.
A person skilled in the art would understand that the conditioning and priming step of the drug delivery device will be affected by the medium in which the device is placed. Histrelin and naltrexone implants, for example, can be conditioned
-23-and primed in saline solution, more specifically, conditioned in saline solution of 0.9% sodium content and primed in saline solution of 1.8% sodium chloride content.
The temperature used to condition and prime the drug delivery device can vary across a wide range of temperatures, but, in some instances 37 C, is used.
The time period used for the conditioning and priming of the drug delivery devices can vary from about a single day to several weeks depending on the release rate desired for the specific implant or drug.
A person skilled in the art will understand the steps of conditioning and priming the implants, where appropriate or necessary, is to optimize the rate of release of the drug contained within the implant. As such, a shorter time period spent on the conditioning and the priming of a drug delivery device can result, for example, in a lower rate of release of the drug compared to a similar drug delivery device that has undergone a longer conditioning and priming step. Without priming, however, it was unexpectedly found that effective release occurred over a longer period of time (e.g., 28 weeks and beyond), and lower serum concentrations of the active ingredient were found to have ameliorative effects.
The temperature in the conditioning and priming step can also affect the rate of release in that a lower temperature results in a lower rate of release of the drug contained in the drug delivery device when compared to a similar drug delivery device that has undergone a treatment at a higher temperature.
Similarly, in the case of aqueous solutions, which are in some cases preferably saline solutions, the sodium chloride content of the solution will also determine what type of rate of release will be obtained for the drug delivery device.
More specifically, a lower content of sodium chloride can result in a higher rate of release of drug when compared to a drug delivery device that has undergone a conditioning and priming step where the sodium chloride content was higher.
To identify the location of the implant, radiopaque material can be incorporated into the delivery device by inserting it into the reservoir or by making it into end plug to be used to seal the cartridge.
The formulation of the present invention can contain a pharmaceutically acceptable carrier that can include, for example, suspending media, solvents, aqueous systems and solid substrates or matrices.
-24-Suspending media and solvents useful as the carrier include, for example, oils such as silicone oil (particularly medical grade), corn oil, castor oil, peanut oil and sesame oil; condensation products of castor oil and ethylene oxide; liquid glyceryl triesters of a lower molecular weight fatty acid; lower alkanols;
glycols; and polyalkylene glycols.
The aqueous systems include, for example, sterile water, saline, dextrose, dextrose in water or saline, and the like. The presence of electrolytes in the aqueous systems may tend to lower the solubility of the macromolecular drug in them.
The solid substrates or matrices include, for example, starch, gelatin, sugars (e.g., glucose), natural gums (e.g., acacia, sodium alginate, carboxymethyl cellulose), and the like. In a preferred embodiment, the pharmaceutical formulation further comprises about 2% to about 20%, more preferably about 10%
hydroxypropylcellulose. In addition, the pharmaceutical formulations may also contain hydroxyporopylcellulose, hydoxyethyl cellulose, methyl cellulose, sodium carboxymethyl cellulose, modified starch or crosslinked polyvinyl pyrrolidone.
The carrier can also contain adjuvants such as preserving, stabilizing, wetting and emulsifying agents and the like.
In one embodiment, a pharmaceutical formulation of the present invention comprises a formulation of octreotide acetate within a mixture of HEMA and HPMA copolymer, preferably about 20% HEMA and about 80% HPMA. The pharmaceutical formulation can comprise, for example, about 20 to about 150 milligrams of octreotide, preferably about 40 to about 90 milligrams. The formulation can further comprise between about 2 to about 20% excipients. The formulation can also contain about 10% hydroxypropylcellulose and/or about 2%
magnesium stearat.
A pharmaceutical formulation of the present invention can comprise a formulation of about 50 milligrams of octreotide within a mixture of HEMA and HPMA copolymer, preferably about 20% HEMA and about 80% HPMA. In a further embodiment, the formulation further comprises about 10%
hydroxypropylcellulose and 2% magnesium stearate with the octreotide acetate.
A pharmaceutical formulation of the present invention also can comprise a formulation of about 83 mg of octreotide within a mixture of HEMA and HPMA
-25-copolymer, preferably about 40% HEMA and about 60% HPMA. In a further embodiment, the formulation further comprises about 10% hydroxypropylcellulose and 2% magnesium stearate with the octreotide acetate. The pharmaceutical formulations may also contain stearic acid, vegetable stearin, talc and silica.
A pharmaceutical formulation of the present invention can also comprise a formulation of about 20 milligrams to about 150 milligrams, more preferably about 40 milligrams to about 90 milligrams, of octreotide in a xerogel, preferably a hydrogel or a polyurethane based polymer.
A method of treating a disease associated with a hormonal disorder is provided. The method can include administering octreotide and maintaining a plasma concentration at steady state of octreotide between about 0.1 ng/ml and about 9 ng/ml over an extended period of time, preferably at least about two months, and more preferably about six months and up to about two years. In preferred embodiment, the plasma concentration at steady state of octreotide is maintained between about 1 ng/ml and about 2 ng/ml, more preferably about 1.2 ng/ml to about 1.6 ng/ml, over an extended period of time. Hormonal disorders include, for example, acromegaly.
The invention is also directed to methods for decreasing GH levels by administering octreotide and maintaining a steady state plasma concentration of zo octreotide between about 0.1 ng/ml and about 9 ng/ml, about 0.5 ng/ml to about 1 ng/ml, about 1 ng/ml to about 2 ng/ml, or about 1.2 to about 1.6 ng/ml, over an extended period of time, preferably at least about two months, and more preferably about six months, and up to about two years.
The invention is also directed to methods for decreasing IGF-1 levels by administering octreotide and maintaining a plasma concentration of octreotide between about 0.1 ng/ml and about 9 ng/ml, about 0.5 ng/ml to about 1 ng/ml, about 1 ng/ml to about 2 ng/ml, or about 1.2 to about 1.6 ng/ml, over an extended period of time, preferably at least about two months, and more preferably about six months, and up to about two years.
The invention is further directed to methods of treating acromegaly comprising administering at least one implant of the present invention, preferably two implants, of the present invention. In the method, each implant administered
-26-can contain between about 20 to about 150 milligrams of octreotide, preferably about 40 to about 90 milligrams of octreotide, more preferably about 50 milligrams of octreotide, and release a therapeutically effective amount of octreotide over a period of at least two months, preferably about six months, and up to about two years.
The invention is further directed to methods of treating symptoms associated with carcinoid tumors and VIPomas. Methods of treating severe diarrhea and flushing episodes associated with carcinoid tumors by administering an implantable formulation of octreotide, which releases a therapeutically effective amount of octreotide over at least about two months, preferably about six months and up to about two years, are also encompassed by the present invention. Methods of treating watery diarrhea associated with VIPomas by administering an implantable formulation of octreotide, which release a therapeutically effective amount of octreotide over at least about two months, preferably about six months and up to about two years, are also encompassed by the present invention.
The formulations of the present invention exhibit a specific, desired release profile that maximizes the therapeutic effect while minimizing adverse side effects of the implant. The desired release profile can be described in terms of the maximum plasma concentration of the drug or active agent (Cmax) and the plasma concentration of the drug or active agent at steady state (Css).
The present invention is also directed to therapeutic compositions of a hydrogel and octreotide, wherein, upon implantation, the octreotide is released at a rate that provides and/or maintains a Css of about 0.1 ng/ml to about 9 ng/ml, about 0.5 ng/ml to about 1 ng/ml, about 1 ng/ml to about 2 ng/ml, or about 1.2 ng/ml to about 1.6 ng/ml. A further embodiment is a therapeutic composition of a hydrogel and octreotide, wherein, upon implantation, the octreotide is released at a rate of from about 10 jig to about 1000 itg per day over an extended period of time, preferably about 20 lig to about 800 jig, more preferably about 30 jig to about 300 jig per day. The octreotide can be release over at least about two months, about six months, or up to about two years. The hydrogel can comprise methacrylate based polymers or polyurethane based polymers.
-27-Another embodiment is a controlled-release formulation comprising octreotide and a hydrophilic polymer, which permits release of the octreotide at a rate of about 30 jig to about 250 jig per day over at least about two months, about six months or about two years in vitro. In some embodiment, delivery is about 100 ,g to about 130 jig per day. In a further embodiment, the hydrophilic polymer of the formulation permits release of octreotide at an average rate of about 100 jig per day in vitro. The hydrophilic polymer can be selected from polyurethane based polymers and methacrylate based polymers.
A further embodiment of the present invention is a controlled-release formulation comprising octreotide for implantation, wherein the formulation comprises octreotide in a hydrophilic polymer effective to permit in vitro release of no more than about 20% of said octreotide from the formulation after about 6 weeks;
and about 60% of said octreotide from said formulation after about six months.

The amount of a pharmaceutically-acceptable octreotide (e.g., various salts, salvation states, or prodrugs thereof) included in the pharmaceutical composition of the present invention will vary depending upon a variety of factors, including, for example, the specific octreotide used, the desired dosage level, the type and amount of hydrogel used, and the presence, types and amounts of additional materials included in the composition. The amount of octreotide, or a derivative thereof, in the formulation varies depending on the desired dose for efficient drug delivery, the molecular weight, and the activity of the compound. The actual amount of the used drug can depend on a patient's age, weight, sex, medical condition, disease or any other medical criteria. The actual drug amount is determined according to intended medical use by techniques known in the art. The pharmaceutical dosage formulated according to the invention can be administered, for example, about once every six months as determined by the attending physician.
The octreotide typically is formulated in the implant or other pharmaceutical composition in amounts of about 20 milligrams to about 150 milligrams, preferably about 40 to about 90 milligrams of octreotide, more preferably about 50 to about 85 milligrams. For adults, the daily dose for treatment of acromegaly is typically about 300 [1g to about 600 jig of immediate release octreotide per day (100 /..tg or 200 ,g Sandostatine). The amount of octreotide in the composition can be selected, for
-28-example, to release from about 10 jig to about 1000 jig per day over an extended period of time, about 20 jig to about 800 jig per day, or about 30 jig to about 300 jig per day. Such release rates maintain desired therapeutic levels in a patient's blood at about 0.1 to about 9 ng/ml over an extended period of time.
The hydrogel device in which octreotide is contained provides a controlled-release of octreotide into the plasma of the patient. Hydrogels suitable for controlling the release rate of octreotide for use in the pharmaceutical compositions of the present invention include polymers of hydrophilic monomers, including, but not limited to HPMA, HEMA and the like. Such hydrogels are also i o capable of preventing degradation and loss of octreotide from the composition.
IA pharmaceutical formulation of the present invention can comprise octreotide acetate within a hydrophilic copolymer of 2-hydroxyethyl methacrylate and hydroxypropyl methacrylate. The copolymer of the pharmaceutical formulation can comprise, for example, about 20% HEMA and about 80% HPMA. The copolymer of the pharmaceutical formulation can alternatively comprise, for example, about 40% HEMA and about 60% HPMA.
The size, shape and surface area of the implant can be modified to increase or decrease the release rate of octreotide from the implant.
The pharmaceutical composition of the present invention can include also auxiliary agents or excipients, for example, glidants, dissolution agents, surfactants, diluents, binders including low temperature melting binders, disintegrants and/or lubricants. Dissolution agents increase the dissolution rate of octreotide from the dosage formulation and can function by increasing the solubility of octreotide.
Suitable dissolution agents include, for example, organic acids such as citric acid, fumaric acid, tartaric acid, succinic acid, ascorbic acid, acetic acid, malic acid, glutaric acid and adipic acid, which can be used alone or in combination.
These agents man also be combined with salts of the acids, e.g., sodium citrate with citric acid, to produce a buffer system.
Other agents that can alter the pH of the microenviromnent on dissolution and establishment of a therapeutically-effective plasma concentration profile of octreotide include salts of inorganic acids and magnesium hydroxide. Other agents that can be used are surfactants and other solubilizing materials. Surfactants that are
-29-suitable for use in the pharmaceutical composition of the present invention include, for example, sodium lauryl sulfate, polyethylene stearates, polyethylene sorbitan fatty acid esters, polyoxyethylene castor oil derivatives, polyoxyethylene alkyl ethers, benzyl benzoate, cetrimide, cetyl alcohol, docusate sodium, glyceryl monooleate, glyceryl monostearate, glyceryl palmitostearate, lecithin, medium chain triglycerides, monoethanolamine, oleic acid, poloxamers, polyvinyl alcohol and sorbitan fatty acid esters.
Diluents that are suitable for use in the pharmaceutical composition of the present invention include, for example, pharmaceutically acceptable inert fillers such o as microcrystalline cellulose, lactose, sucrose, fructose, glucose dextrose, or other sugars, dibasic calcium phosphate, calcium sulfate, cellulose, ethylcellulose, cellulose derivatives, kaolin, mannitol, lactitol, maltitol, xylitol, sorbitol, or other sugar alcohols, dry starch, saccharides, dextrin, maltodextrin or other polysaccharides, inositol or mixtures thereof. The diluent can be a water-soluble diluent. Examples of diluents include, for example: microcrystalline cellulose such as Avicel PH112, Avicel PH101 and Avicel PH102 available from FMC
Corporation; lactose such as lactose monohydrate, lactose anhydrous, and Pharmatose DCL 21; dibasic calcium phosphate such as Emcompress available from Penwest Pharmaceuticals; mannitol; starch; sorbitol; sucrose; and glucose.
Diluents are carefully selected to match the specific composition with attention paid to the compression properties. The diluent is preferably used in an amount of about 2% to about 80% by weight, preferably about 20% to about 50% by weight, of the controlled-release composition.
Glidants are used to improve the flow and compressibility of ingredients during processing. Suitable glidants include, for example, colloidal silicon dioxide, a sub-micron fumed silica that can be prepared, for example, by vapor-phase hydrolysis of a silicon compound such as, for example, silicon tetrachloride.
Colloidal silicon dioxide is a sub-micron amorphous powder that is commercially available from a number of sources, including Cabot Corporation (under the trade name Cab-O-Sile); Degussa, Inc. (under the trade name Aerosile); and E.I.
DuPont & Co. Colloidal silicon dioxide is also known as colloidal silica, fumed silica, light
-30-anhydrous silicic acid, silicic anhydride and silicon dioxide fumed, among others. In one embodiment, the glidant comprises Aerosil 200.
Disintegrants that are suitable for use in the pharmaceutical composition of the present invention include, for example, starches, sodium starch glycolate, crospovidone, croscarmellose, microcrystalline cellulose, low substituted hydroxypropyl cellulose, pectins, potassium methacrylate-divinylbenzene copolymer, poly(vinyl alcohol), thylamide, sodium bicarbonate, sodium carbonate, starch derivatives, dextrin, beta cyclodextrin, dextrin derivatives, magnesium oxide, clays, bentonite and mixtures thereof The active ingredient of the present invention can be mixed with excipients that are pharmaceutically-acceptable and compatible with the active ingredient and in amounts suitable for use in the therapeutic methods described herein.
Various excipients can be homogeneously mixed with octreotide of the present invention as would be known to those skilled in the art. Octreotide, for example, can be mixed or combined with excipients such as but not limited to microcrystalline cellulose, colloidal silicon dioxide, lactose, starch, sorbitol, cyclodextrin and combinations of these.
Lubricants that are suitable for use in the pharmaceutical composition of the present invention include agents that act on the flowability of the powder to be compressed include but are not limited to silicon dioxide such as, for example, Aerosil 200, talc; stearic acid, magnesium stearate, calcium stearate, hydrogenated vegetable oils, sodium benzoate, sodium chloride, leucine carbowax, magnesium lauryl sulfate, and glyceryl monostearate.
The invention is further directed to a controlled-release implantable dosage formulation that includes an effective amount a octreotide in a hydrogel, and that, upon administration to a patient or as part of a therapy regimen, provides a release profile (of therapeutically-effective blood plasma level of octreotide) extending for a period of at least about two months, about six months or up to about two years.
The dosage formulation of the present invention can comprise one or more pharmaceutically-acceptable excipients. In preferred embodiments, the dosage formulation will comprise diluents and a lubricant in addition to octreotide unit dose and the rate-controlling polymer. For this purpose, magnesium stearate is a suitable
-31-excipient. When these materials are used, the magnesium stearate component can comprise from about 0.5 to about 5% w/w of the dosage formulation (e.g., about 2%), and the hydrogel and octreotide comprise the remainder of the formulation.
Another suitable excipient is hydroxypropylcellulose. When used, the hydroxypropylcellulose component can comprise from about 0.5 to about 20% w/w of the dosage formulation (e.g., about 10%), and the hydrogel and octreotide comprise the remainder of the formulation.
In one embodiment, the formulation comprises both magnesium stearate and hydroxypropylcellulose, preferably about 2% magnesium stearate and about 10%
hydroxypropylcellulose and the hydrogel and octreotide comprise the remainder of the formulation.
The compositions of the present invention can be used for the treatment of hormonal diseases, e.g., acromegaly, or symptoms thereof characterized by increased levels of GH and IGF-1 by administering to a patient an implantable formulation of the present invention. The implant can be administered, for example, every about six months, and release a therapeutically-effective amount of octreotide.
The implantable composition releases a concentration of octreotide in the patient at about the minimum therapeutically-effective level to ameliorate the hormonal disorder, yet relatively lower compared to the maximum concentration to enhance restful periods for the patient during the day. The compositions can be administered to a subject at a dose and for a period sufficient to allow the subject to tolerate the dose without showing adverse effects and thereafter increasing the dose of the active agent, if needed, at selected intervals of time until a therapeutic dose is achieved in the subject. The active agent can be administered, for example, at a dose of from about 10 pg to about 1000 g, about 20 g to about 800 jig, or about 30 g to about 300 pg, of octreotide daily for a period of at least about two months, about six months, or up to about two years.
Compositions of the present invention where the octreotide is octreotide acetate are suitable for use in the treatment of hormonal disorders that are characterized by increased levels of GH and IGF-1, e.g., acromegaly. The octreotide acetate agent in accordance with the invention is also suitable for the treatment of symptoms associated with carcinoid syndrome and VIPomas.
-32-Additional features and embodiments of the present invention are illustrated by the following non-limiting examples.
EXEMPLIFICATION
Example 1. In Vitro Octreotide Release Rates This example illustrates preparation of implantable octreotide formulations of the present invention and their in vitro release of octreotide. In the present study, a series of implants were tested to determine stability and in vitro release characteristics of octreotide from the hydrogel formulations over about 22 weeks (No. 146), 28 weeks (No. 136) and 33 weeks (all other formulations). Each implant io contained about 50 milligrams of octreotide acetate and about 2% stearic acid, but the polymer cartridges contained different amounts of HEMA and HPMA and therefore exhibited different %EWCs, as depicted in Table 1.
Table 1.
Formulation %
Number HEMA HPMA EWC Excipients/Other Ingredients 146 0 99.5 22.9 2% stearic acid 145 10 89.5 23.4 2% stearic acid 147 15 84.5 24.4 2% stearic acid 133 20 79.5 25.2 2% stearic acid 144 25 74.5 25.6 2% stearic acid 143 30 69.5 26.1 2% stearic acid 142 35 64.5 26.6 2% stearic acid 136 40 59.5 27.6 2% stearic acid FIGS. 2, 3 and 4 depict the release of octreotide from the implant per day for each of the formulations provided above. As noted in FIG. 2, the initial release was relatively high and dropped relatively quickly for Formulation No. 136. As shown in FIG. 3, the initial release rate for Formulation No. 146 was relatively low. FIG. 4 presents the release profiles for Formulation Nos: 145, 147, 133, 144, 143 and 142.
As shown in FIG. 4, the initial release rates show a good relationship with the %EWC, ranging from 20 to 450 pg per day for %EWCs of 22.9 to 27.6%. Problems
-33-were encountered, however, with respect to the osmotic pressure differential within the implant and the elution media. To stabilize the octreotide formulations, a number of experiments were designed using excipients that would provide better stability based on a "preferential hydration" principle.
Example 2. Formulation Study in Calf Serum To determine the effect of osmotic pressure on the swelling problem two implants of the present invention corresponding to Formulation No. 136 and Formulation No. 143 were eluted in calf serum. In particular, Formulation No.
136, composed of about 40% HEMA and 60% HPMA, containing octreotide acetate with 2% stearic acid and Formulation No. 143, composed of about 30% HEMA and 70%
HPMA, containing a mixture of 20% PEG3300 and 80% octreotide acetate, were tested. After three months, the implants exhibited normal appearance, being relatively straight and only slightly swollen.
Example 3. Formulation Study Due to osmotic pressure differential, the implants described in Example 1 were seen to swell significantly- ultimately resulting in bursting of the implants.
This example illustrates formulations designed to screen agents useful in stabilization of the octreotide implant. A series of implants was monitored to determine the effect of excipient on implant shape and durability. Each of the polymer cartridges was composed of about 28% HEMA, about 66.5% HPMA and 5% glycerin. The contents contained octreotide acetate with various excipients, as shown in Table 2.
-34-Table 2.
Sample No. Excipients/Other Ingredients 1 None 2 20% PEG 3300 3 40% PEG 3300 4 2% Stearic acid (control) 10% Glycolic acid 6 20% Poly(lactic acid) 7 10% Mannitol 8 10% MCC (microcrystalline cellulose) 9 20% MCC
10% Sesame oil Hydrophobic agents such as sesame oil and MCC separated in the formulation and did not provide "preferential hydration" and were less preferable in accordance with the present invention. Hydrophilic agents like PEG 3300 increased 5 the osmotic pressure differential and increased swelling. Low molecular weight additives like marmitol and glycolic acid did not provide a stabilizing effect and resulted in a decrease in integrity. None of these agents provided satisfactory stabilization of the octreotide formulations.
Example 4. Formulation Study and In Vitro Octreotide Release Rates 10 This study was conducted to evaluate stability of octreotide in hydrogel implants using various excipients as shown in Table 3. The excipients were chosen to have high molecular weight and some hydrophilic nature. Each implant was made from polymer cartridges composed of about 20% HEMA and about 80%
HPMA. The appearance of the implants in saline was monitored and rated over the course of nine weeks. The results are shown in Table 3.
-35-
36 PCT/US2009/050215 Table 3.
Formulation Implant Appearance at 9 No. Excipients/Other Ingredients Weeks (see key below) 133 20% Dextran 3 133 20% TPGS (vitamin E derivative) 2 133 20% HEC (hydroxyethyl cellulose) 3 133 20% HPC (hydroxypropyl cellulose) 2 133 20% Albumin 2 133 20 % Pectin 2 133 20 % AcDiSol 1.5 133 20 % Carbopol 1 133 2% SA (stearic acid) - control 4 As depicted in FIG. 5, the formulation containing dextran had the highest elution rate. The formulations containing pectin, AcDiSol and Carbopol exhibited less than satisfactory release after two weeks hydration and nine weeks elution.
Accordingly, a preferred embodiment having superior stabilizing effect, combination of good elution and appearance, was achieved with hydroxypropylcellulose.
Example 5. One-Month Implantation Study in a Healthy Dog This example illustrates preparation of formulations of the present invention o and their release of octreotide or pharmaceutically-acceptable salts thereof. A
healthy dog was implanted with one octreotide subdermal implant of the present invention. The octreotide subdermal implant formulation had a water content of 26.6%, containing 44 mg octreotide acetate. In vitro release rates were estimated at about 500 ktg/day in week 1 and decreasing to about 300 ktg/day in week 4 for a total release of about 10 mg of octreotide over the duration of the study. The implant was removed at 28 days after implantation. The implant used in this study was about 3.5 cm in length. Blood samples (1.5 ml) to obtain the serum concentration of octreotide acetate, IGF-1 and GH were obtained on days 0, 1-7, 11, 14, 18, 21, and 28 by jugular puncture without anesthesia and without fasting.

Clinical observations included that the octreotide implant formulation was well-tolerated, food intake was normal, and no abnormal behavior was noted.
Serum analysis showed a peak of octreotide acetate at day 4 and detectable amounts of octreotide acetate at all intervals measured. IGF-1 concentrations decreased after implantation until day 4, then returned to pre-dose levels by day 25.
IGF-1 levels declined from 40 to 90% of pre-implantation level, as can be seen in FIG. 6.
Example 6. Six-Month Implantation Study in Six Healthy Dogs This example illustrates preparation of formulations of the present invention and their release of octreotide or pharmaceutically-acceptable salts thereof.
Six healthy dogs were divided into two groups and implanted with one or two octreotide subdermal implants of the present invention, respectively. The octreotide subdermal implants had a water content of about 25.2% and contained about 60 mg octreotide acetate. The implants were removed six months after implantation. Blood samples (10 ml) to obtain the serum concentration of octreotide acetate, IGF-1 and GH
were obtained once daily for the first 7 days following implantation followed by twice a week sampling for three weeks, and then once a week until conclusion of the six month period. Four days prior to implantation, baseline serum samples were taken as a control.
Results indicate octreotide serum levels ranged from 200 to 700 tg/m1 in dogs receiving one implant and 400 to 1000 pg/m1 in dogs receiving two implants.
IGF-1 levels were reduced as much as 90% in both treatment groups as can be seen in FIGS. 7 and 8. Measurement of serum GH levels was abandoned after about the first month of the study because levels in healthy animals are too low to detect further reductions. Clinical observations included the octreotide implant formulation was well-tolerated, food intake was normal, and no abnormal behavior was noted.
Example 7. Six-Month Implantation Study in Humans This example illustrates preparation of formulations of the present invention and their release of octreotide or pharmaceutically acceptable salts thereof.
A six-month study was conducted in eleven patients with acromegaly. One or two
-37-implants of the present invention were implanted subcutaneously in 11 patients diagnosed with acromegaly, who were previously treated with a commercially-available octreotide LAR formulation. Levels of GH and IGF-1 were measured at baseline and every month thereafter for a period of six months.
Each implant contained approximately 60 mg of octreotide acetate in a copolymer of 20%
HEMA and 79.5% HPMA, with an EWC of about 25.2%. The implants used in this study were about 44 mm in length in a dry state and 50 mm in length in a hydrated state. The diameters of the implants were about 2.8 mm in a dry state and about 3.5 to about 3.6 mm in a hydrated state. The implants were hydrated for a period of about 1 week prior to implantation.
The reference ranges for GH is up to 2.5 mg/L, age-independent. Table 4 illustrates the basal levels of GH in mg/L over six months after implantation of octreotide implants of the present invention. Patient No. 11 did not participate in the study due to failure to meet screening criteria.
Table 4.
Visit 1 Visit 2 Visit 3 Visit 4 Visit 5 Visit 6 Visit 7 # (Insertion) (Month 1) (Month 2) (Month 3) (Month 4) (Month 5) (Month 6) Implants Screening GHBasal GH Basal GH Basal GH Basal GH Basal Gil Basal GH Basal GH
Patient Age Rec'd (mg/L) (mg/L) (mg/L) (mg/L) (mg/L) (mg/L) (mg/L) (mg/L) 001 39 1 26 16.3 0.9 1.5 1.1 1.1 1.1 2.1 002 38 2 17.8 20.7 1.4 0.2 0.3 0.2 0.3 0.48 003 49 1 67 55 2.8 3.1 3.3 5.0 5.3 5.8 004 47 2 7.9 7 2.6 3.8 2.8 3.7 4.0 2.4 005 43 1 10.8 11 2.2 1.8 2.2 1.6 2.2 1.3 006 43 1 1.7 1.7 1.8 2.3 1.9 1.7 1.8 1.9 007 30 2 23.3 21.8 2.4 2.2 2.9 2.0 1.1 0.51 008 58 2 1.9 3.2 0.1 0.1 2.0 0.1 0.6 0.11 009 47 2 14.9 14.1 1.4 0.9 1.5 1.1 1.4 1.4 010 78 1 4 5.2 0.4 0.2 0.5 0.2 0.3 1.0 012 40 2 21.1 27.8 13.5 13.7 14 11.9 8.9 13.1 mean 16.7 2.7 2.7 3.0 2.6 2.7 2.7 As shown above, by month six 89% of subjects exhibited normalized growth hormone levels. Reference ranges for IGF-1 are as follows: (i) 17-24 years old about 180-780 ng/mL; (ii) 25-39 years old about 114-400 ng/mL; (iii) 40-54 years old about 90-360 ng/mL; and (iv) >54 years old about 70-290 ng/mL.
-38-Table 5 illustrates the basal levels of IGF-1 in ng/ml over six months after implantation of octreotide implants of the present invention.
Table 5.
Visit I Visit 2 Visit 3 Visit 4 Visit 5 Visit 6 Visit 7 # Screening (Insertion) (Month 1) (Month 2) (Month 3) (Month 4) (Month 5) (Month 6) Implants IGF-1 IGF-1 IGF-I IGF-I IGF-1 IGF-I IGF-I
IGF-I
Patient Age Rec'd (nghnL) (ng/mL) (ng/mL) (ng/mL) (ng/mL) (nghnL) (nghnL) (nghnL) mean 1288 589 624 661 699 602 648 As shown above, by month six, 22% of subjects exhibited a normalized IGF-1 level.
FIGS. 9A and 9B demonstrate a comparison of the octreotide implant of the present invention with a commercially-available formulation of octreotide acetate.
The efficacy of the implant appeared to be at least as good as that of the commercially-available octreotide LAR formulation. The therapeutic effect of these io implants continued successfully for the entire 6 months of the study duration.
IGF-1 levels were decreased in all patients, with normalization in 2 patients.

The decrease was already observed at one month of therapy and the mean IGF-1 level was stable for the following 5 months. A comparison with decreases previously observed in the same patients while on the commercially available octreotide LAR formulation therapy was possible in 8 of the 9 patients. In 6 of the 8 patients, the percentage decrease in IGF-1 during the implant was greater than that while on the commercially-available octreotide LAR formulation, whereas in 2, it was less. After 6 months of therapy with the implant, GH levels in 3 patients were <1 ng/ml and in another 5, were <2.5 ng/ml. This compared favorably with the
-39-results on the commercially-available octreotide LAR formulation, where GH
levels in only 2 patients were <1 ng/ml and in another 2, were under 2.5 ng/ml.
Levels of octreotide in the serum of patients was also measured, as shown in Table 6.
Table 6.
Month 1 2 3 4 5 6 7 #Implants Patient ID Visit 2 Visit 3 Visit 4 Visit 5 Visit 6 Visit 7 Visit 8 Gender 1 Patient 1 1181 874.5 738.0 894.3 699.2 722.3 169.0 F
2 Patient 2 2686 2478 1625 1833 1388 1203 280 'M
1 Patient 3 2570 2351 1332 980.5 1131 775.2 2 Patient 4 4268 3308 2582 2650 2455 1 Patient 5 1218 1022 610.0 783.2 709.4 545.8 144 F
1 Patient 6 1899 1445 1427 1123 1148 747.7 206 F
2 Patient 7 5524 2621 3656 3141 2205 2 Patient 8 8684 3387 4899 3336 3454 2 Patient 9 3850 860.6 2638 1766 1729 1 Patient 10 2055 1628 1192 863.9 1641 1231 1130 F
2 Patient 12 2527 1366 2006 962.8 1484 A comparison of the octreotide levels achieved with one and two implants is depicted in the graph in FIG. 10. Overall, results indicated that the octreotide implant of the present invention is at least as effective as the commercially available LAR formulation of octreotide acetate in reducing GH levels and IGF-1 levels in patients with acromegaly.
Example 8. In Vitro Octreotide Delivery Using Dry Implants This example illustrates preparation of formulations of the present invention and their release of octreotide or pharmaceutically acceptable salts thereof.
Two healthy dogs were implanted with one octreotide subdermal implant of the present invention. The implants were not hydrated prior to implantation. The octreotide subdermal implants were composed of about 59.5% HPMA and about 40% HEMA
and had an equilibrium water content of about 27.6%. The implants contained about 84 mg of octreotide acetate, hydroxypropylcellulose and magnesium stearate.
The implants were removed six months after implantation. Blood samples (10 ml) were drawn to obtain the serum concentration of octreotide acetate and IGF-1 once daily every other day for the first four weeks following implantation followed by twice a
-40-week sampling for four weeks, and then once a week until conclusion of the six month period. Two days prior to implantation, baseline serum samples were taken as a control.
FIG. 11 shows the octreotide levels in the serum of the dogs and FIG. 12 shows the levels of IGF-1 in the dogs.
Example 9. Implant Compositions Possible compositions for the implants of the invention include, for example, those listed in Table 7, below. Implant cartridges greater than about 3.2-3.4 mm (dry) are aided by the use of release agents, e.g., vitamin E TPGS, during the formation process.
Table 7. Composition of Implant Small Implant Large Implant API 60 mg Octreotide Acetate 84 mg Octreotide Acetate Pellet Excipients 10 % Hydroxypropyl 10 % Hydroxypropyl cellulose (¨ 6.8 cellulose (¨ 9.5 mg/implant) mg/implant) 2 % Magnesium Stearate 2 % Magnesium Stearate (-1.3 mg/implant) (-2 mg/implant) Monomer Mixture 20 % HEMA 40 % HEMA
Composition 79.5 % HPMA 59.5 % HPMA
0.5 % TMPTMA 0.5 % TMPTMA
Added to mixture: Added to mixture:
1 % Triton X-100 1 % Vitamin E TPGS
0.3 % BME 0.3 % BME
0.1 %P-16 0.1 %P-16 Dry Implant Size 2.8 mm x 43 mm 3.4 mm x 43 mm Surface Area 378 mm2 459 mm2 Hydrated Implant 3.4 mm x 50 mm 4.3 mm x 50 mm Size Surface Area 534 mm2 675 mm2 EWC 26.0% 28.7%
Sterilization Gamma Irradiation Gamma Irradiation Packaging Solution Implants packaged dry in 2 Implants packaged dry in
-41-compartment package with 2 compartment package 0.9% saline solution in the with second compartment. 0.9% saline solution in Implant is combined with the second compartment.
saline 7-14 days prior to Implant is combined with implantation to allow for saline 3 -7 days prior to implant hydration. implantation to allow for implant hydration.
Packaging Divided Pouch with Divided Pouch with LF4835W Foil JT48FLLP Foil Barrier/FR5500 PET/PE B arrier/IT-CB 259B
Clear Sleeve as Aluminum Oxide CTD
components. LF4835W ¨ PET Clear Sleeve as DMF # 15796 components.
FR5500 ¨ Approved for For use in sterile medical food contact packaging Average Daily 130 vig/day for 6 months 250 jig/day for 6 months Release Rate Example 10. An Open-Label Study to the Evaluate the Pharmacokinetic and Pharmacodynamic Response of a Hydrated and Non-Hydrated 84 mg Octreotide Implant in Patients with Acromegaly Approximately 30 patients with acromegaly were enrolled after written informed consent was obtained. Patients were divided in 2 groups per the study randomization schedule: 15 patients received one hydrated 84 mg octreotide implant and 15 patients received one non-hydrated 84 mg octreotide implant. Eligible patients received the implant within 7 days of their screening visit. The octreotide implant was inserted subcutaneously in the inner aspect of their non-dominant arm under local anesthesia. Blood samples for the determination of IGF-1, GH and octreotide serum concentrations were collected at predetermined time points within the first 6 weeks after implantation. Patients then return for visits at Week 8, 12, 16, and 24 to have blood samples collected for the determination of IGF-1, GH and octreotide serum concentrations, as well as safety assessments. At the end of the 6-15 month (24-week) treatment phase, the implant will be removed. Following implant
-42-removal, the patient will be instructed to return in 4 weeks for the End of Study Visit (Week 28). Safety and efficacy will be carefully monitored throughout the study.
Investigational Products:
Hydrated octreotide implant (84 mg octreotide acetate) for subcutaneous implantation Non-hydrated octreotide implant (84 mg octreotide acetate) for subcutaneous implantation Duration of Treatment:
Eligible patients receive one implant, either hydrated or non-hydrated. At the end of the 6-month (24-week) treatment phase, the implant will be removed.
Following implant removal, the patient will be instructed to return in 4 weeks for the End of Study Visit.
Criteria for Inclusion:
1. Male and female patients with acromegaly 2. Must be 2 8 years of age 3. Confirmed diagnosis of a growth hormone-secreting tumor (elevation of IGF-1 level 20 % above upper limit of age-and sex-adjusted normal value and either a post-glucose GH of 2 .0 ng/ml or a pituitary tumor demonstrable on MRI). If patient has undergone pituitary surgery and has residual tumor present, it must be at least 3 mm in distance from the optic chiasm (unless patient is not a surgical candidate) and IGF-1 level must be elevated as described above. If no residual tumor is present or patient is inoperable then patient must meet both IGF-1 and GH criteria as described above.
4. Must be either a full or partial responder to octreotide demonstrated by historical laboratory values, as defined below:
a. Full responder: suppression of serum IGF-1 to normal age-and sex-adjusted levels and suppression of serum GH to <1.0 ng/ml after OGTT
b. Partial responder: a 20% decrease in IGF-1 and GH values when compared to pre-treatment values, but not meeting criteria for full responder
-43-c. OR
d. Must be a responder to octreotide demonstrated by laboratory values obtained via an acute aqueous test during the Screening Visit for octreotide naïve patients or patients in whom response to octreotide is unknown, as defined below:
e. Responder via acute aqueous test: a 0% decrease in GH
values at any time point of the 4 hour test period in response to a subcutaneous injection of 100 ftg of aqueous octreotide 5. Must be able to communicate, provide written informed consent, and willing to participate and comply with study requirements 6. Patient is eligible to participate in the opinion of the Investigator Criteria for Exclusion:
1. Women who are pregnant, lactating, or of child-bearing potential who are not practicing a medically acceptable method of birth control 2. Patients with pituitary surgery less than 12 weeks prior to screening 3. Patients with liver disease (e.g., cirrhosis, chronic active or persistent hepatitis or persistent abnormalities of ALT, AST (level >2x normal), alkaline phosphatase (level >2x normal), or direct bilirubin (level >1.5x normal) 4. Other laboratory values considered by the Investigator or Sponsor to be clinically significant 5. Patients with unstable angina, sustained ventricular arrhythmias, heart failure (NYHA III and IV), or a history of an acute myocardial infarction within 3 months of screening 6. Patients with symptomatic cholelithiasis 7. Patients with a history of drug or alcohol abuse within 6 months of screening 8. Patients who have received any investigational drug within 1 month of screening 9. Patients receiving radiotherapy for their pituitary tumor at any time before screening
-44-10.
Patients who have discontinued octreotide due to tolerability or efficacy issues.
Serum levels of octreotide were determined (see FIGS. 13 and 14 for graphical data).
The efficacy of cytokine concentration modulation after the octreotide implant was inserted, either in the dry form or after hydration, is shown in FIGS. 15, 16, and 17.
Example 11. Treatment of tumors with octreotide.
Sandostatin LAR Depot, as indicated on the FDA label (the entire contents of which are herein incorporated by reference), is a long-acting dosage form consisting of microspheres of the biodegradable glucose star polymer, D,L-lactic and glycolic acids copolymer, containing octreotide. It maintains all of the clinical and pharmacological characteristics of the immediate-release dosage form Sandostatin (octreotide acetate) Injection with the added feature of slow release of octreotide from the site of injection, reducing the need for frequent administration.
This slow release occurs as the polymer biodegrades, primarily through hydrolysis.
Sandostatin LAR Depot is designed to be injected intramuscularly (intragluteally) once every four weeks.
Octreotide exerts pharmacologic actions similar to the natural hormone, somatostatin. It is an even more potent inhibitor of growth hormone, glucagon, and insulin than somatostatin. Like somatostatin, it also suppresses LH response to GnRH, decreases splanchnic blood flow, and inhibits release of serotonin, gastrin, vasoactive intestinal peptide, secretin, motilin, and pancreatic polypeptide.
By virtue of these pharmacological actions, octreotide has been used to treat the symptoms associated with, for example, metastatic carcinoid tumors (flushing and diarrhea), and Vasoactive Intestinal Peptide (VIP) secreting adenomas (watery diarrhea). Octreotide substantially reduces and in many cases can normalize growth hormone and/or IGF-I (somatomedin C) levels in patients with acromegaly.
Single doses of Sandostatin Injection given subcutaneously have been shown to inhibit gallbladder contractility and to decrease bile secretion in normal volunteers.
-45-(.
In patients with acromegaly, the pharmacolcinetics differ somewhat from those in healthy volunteers. A mean peak concentration of 2.8 ng/mL (100 mcg dose) was reached in 0.7 hours after subcutaneous dosing. The volume of distribution (Vdss) was estimated to be 21.6 8.5 L and the total body clearance was increased to 18 L/h. The mean percent of the drug bound was 41.2%. The disposition and elimination half-lives were similar to normals.
Treating these tumors typically involves surgery as the first-line therapy.
Failing surgery, patients are usually given octreotide injections (such as S-Lar).
Chemotherapy may also prove beneficial- and does so in about 30% of patients.
Patients with carcinoid tumors were treated with six doses of S-Lar at 10, 20, or 30 mg given by i.m. injection every 4 weeks. Resulting serum concentrations were 1.2, 2.5, and 4.2 ng/mL. Steady state was achieved after two doses at 20 or 30 mg and after 3 doses at 10 mg.
Treatment with S-Lar reduced daily stool frequency to 2 ¨ 2.5 stools/day, is decreased mean daily flushing episodes to 0.5 to 1 episode/day, and reduced median 24-hr urinary 5-HIAA levels by 38-50%. It should be noted that over a 6 m trial, 50-70% of patients who completed the trial required supplemental Sandostatin injections to help control exacerbation of symptoms.
Although the present invention has been described in considerable detail zo with reference to certain preferred embodiments thereof, other versions are possible.
-46-

Claims (25)

What is claimed is:
1. Use of at least one subcutaneously implantable device for delivering octreotide to a subject with a substantially zero-order release profile over an extended period of time, but no less than six months, wherein the subject is a mammal that is not a dog, wherein the at least one implantable device comprises a composition comprising octreotide, wherein the composition is encased in a hydrophilic polymer, and wherein the implantable device is implanted in a dry state, such that the subject receives on a daily basis over a period of at least about six months dose amounts of octreotide, which are effective to treat the subject but which result in lower serum concentrations of octreotide compared with implantation of the implantable device in a hydrated state.
2. The use of Claim 1, wherein the hydrophilic polymer comprises one or more polyurethane-based polymers or one or more methacrylate-based polymers.
3. The use of Claim 1, wherein the octreotide is in free form, salt form or in the form of a complex thereof
4. The use of Claim 1, wherein the octreotide is octreotide acetate.
5. The use of Claim 1, wherein the subject is afflicted with a GH or IGF-1 hormone disorder or its symptoms.
6. The use of Claim 5, wherein the GH or IGF-1 disorder is acromegaly.
7. The use of Claim 1, wherein the at least one subcutaneously implantable device is for delivering to the subject octreotide at an average rate ranging from about 75 µg per day to about 300 µg per day over a period of at least six months.
8. The use of Claim 1, wherein the at least one subcutaneously implantable device is for delivering a dosage of octreotide to the subject resulting in average octreotide serum levels ranging from about 0.5 ng/mL to about 1.4 ng/mL.
9. The use of Claim 8, wherein the dosage of octreotide to the subject results in average octreotide serum levels ranging from about 0.8 ng/mL to about 1.2 ng/mL.
10. The use of Claim 1, wherein the at least one subcutaneously implantable device is for delivering a dosage of octreotide to the subject resulting in C max for octreotide serum levels below 1.2 ng/mL.
11. The use of Claim 1, wherein the at least one subcutaneously implantable device is for delivering a dosage of octreotide to the subject resulting in C max for octreotide serum levels below 1.0 ng/mL.
12. The use of Claim 1, wherein the at least one subcutaneously implantable device is for delivering an effective amount of octreotide for a period of at least about twelve months.
13. The use of Claim 1, wherein the at least one subcutaneously implantable device is for releasing octreotide for at least three to about ten days after implantation.
14. The use of Claim 1, wherein the subject is afflicted with a condition, wherein the condition is: carcinoid syndrome, VIPomas, neuroendocrine tumors, proliferative diabetic retinopathy, rosacea, pancreatitis, gastrointestinal bleeding, pancreatic and intestinal fistulas, Graves-Basedow ophthalmopathy, glaucoma, or symptoms associated with chemotherapy or AIDS.
15. An implantable device, comprising a controlled-release formulation comprising octreotide and hydroxpropylcellulose wherein the formulation is encased in a hydrophilic polymer comprising a co-polymer obtained from the co-polymerization of a mixture comprising at least two hydrophilic, ethylenically unsaturated monomers, wherein the implantable device delivers octreotide to a subject with a substantially zero-order release profile over an extended period of time, but no less than six months, wherein the subject is a mammal that is not a dog, when the implantable device is implanted in a dry state into the subject.
16. The implantable device of Claim 15, wherein the hydrophilic polymer comprises one or more polyurethane-based polymers.
17. The implantable device of Claim 15, wherein the octreotide is in free form, salt form or in the form of a complex thereof.
18. The implantable device of Claim 15, wherein the octreotide is octreotide acetate.
19. The implantable device of Claim 15, wherein when the implantable device is implanted into the subject, the subject receives octreotide at an average rate ranging from about 75 [tg per day to about 300 g per day over a period of at least about six months.
20. The implantable device of Claim 15, wherein when the implantable device is implanted into the subject, the dose amounts of octreotide received by the subject result in average octreotide serum levels ranging from about 0.5 ng/mL to about 1.4 ng/mL.
21. The implantable device of Claim 15, wherein when the implantable device is implanted into the subject, the dose amounts of octreotide received by the subject result in average octreotide serum levels ranging from about 0.8 ng/mL to about 1.2 ng/mL.
22. The implantable device of Claim 15, wherein when the implantable device is implanted into the subject, the dose amounts of octreotide received by the subject result in C max for octreotide serum levels below 1.2 ng/mL.
23. The implantable device of Claim 15, wherein when the implantable device is implanted into the subject, the dose amounts of octreotide received by the subject result in C max for octreotide serum levels below 1.0 ng/mL.
24. The implantable device of Claim 15, wherein when the implantable device is implanted into the subject, release of octreotide occurs at least three to about ten days after implantation.
25. Use of octreotide for the manufacture of a medicament for treating a mammalian subject that is not a dog, wherein the medicament is a controlled-release formulation comprising octreotide and hydroxpropylcellulose, wherein the formulation is encased in a hydrophilic polymer comprising a co-polymer obtained from the co-polymerization of a mixture comprising at least two hydrophilic, ethylenically unsaturated monomers, thereby forming an implantable device, wherein, upon implantation into a mammalian subject that is not a dog, the implantable device delivers octreotide to the subject with a substantially zero-order release profile over an extended period of time, but no less than six months, when the implantable device is implanted in a dry state into the subject.
CA2730391A 2008-07-11 2009-07-10 Delivery of dry formulations of octreotide Expired - Fee Related CA2730391C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US12/171,999 2008-07-11
US12/171,999 US7759312B2 (en) 2005-03-11 2008-07-11 Delivery of dry formulations of octreotide
PCT/US2009/050215 WO2010006236A1 (en) 2008-07-11 2009-07-10 Delivery of dry formulations of octreotide

Publications (2)

Publication Number Publication Date
CA2730391A1 CA2730391A1 (en) 2010-01-14
CA2730391C true CA2730391C (en) 2016-12-06

Family

ID=41268227

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2730391A Expired - Fee Related CA2730391C (en) 2008-07-11 2009-07-10 Delivery of dry formulations of octreotide

Country Status (14)

Country Link
US (4) US7759312B2 (en)
EP (1) EP2309979A1 (en)
JP (2) JP2011527705A (en)
KR (1) KR20110038056A (en)
CN (1) CN102231977A (en)
AU (1) AU2009268480C1 (en)
BR (1) BRPI0915523A2 (en)
CA (1) CA2730391C (en)
IL (1) IL210545A (en)
MX (1) MX336209B (en)
RU (1) RU2518745C2 (en)
UA (1) UA105496C2 (en)
WO (1) WO2010006236A1 (en)
ZA (1) ZA201100800B (en)

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070219131A1 (en) * 2004-04-15 2007-09-20 Ben-Sasson Shmuel A Compositions capable of facilitating penetration across a biological barrier
US7759312B2 (en) * 2005-03-11 2010-07-20 Endo Pharmaceuticals Solutions Inc. Delivery of dry formulations of octreotide
CA2601304C (en) * 2005-03-11 2010-08-17 Indevus Pharmaceuticals, Inc. Controlled release formulations of octreotide
NZ580803A (en) * 2007-04-27 2012-03-30 Endo Pharmaceuticals Solutions Implant device release agents and methods of using same
CN102231976B (en) * 2008-06-25 2014-05-07 Endo药物方法有限公司 Octreotide implant having a release agent
WO2009158412A2 (en) * 2008-06-25 2009-12-30 Endo Pharmaceuticals Solutions Inc. Sustained delivery of exenatide and other polypeptides
ES2629131T3 (en) 2008-09-17 2017-08-07 Chiasma Inc. Pharmaceutical compositions and related delivery methods.
CN103039688B (en) 2012-01-20 2016-01-06 奥驰亚客户服务公司 Oral product
CN103040090B (en) 2012-01-20 2016-03-30 奥驰亚客户服务公司 Remove the oral product of tobacco
US9854831B2 (en) 2012-01-20 2018-01-02 Altria Client Services Llc Oral product
CN102754908B (en) * 2012-01-20 2015-06-10 奥驰亚客户服务公司 Oral tobacco product
CN102754907B (en) 2012-01-20 2015-06-24 奥驰亚客户服务公司 Oral product
EP3007704B1 (en) * 2013-06-13 2021-01-06 Antisense Therapeutics Ltd Combination therapy for acromegaly
US10238709B2 (en) 2015-02-03 2019-03-26 Chiasma, Inc. Method of treating diseases
CN110732037B (en) 2018-07-20 2023-05-26 广州倍绣生物技术有限公司 Hemostatic paste and preparation method thereof
US11141457B1 (en) 2020-12-28 2021-10-12 Amryt Endo, Inc. Oral octreotide therapy and contraceptive methods
WO2023137018A1 (en) * 2022-01-11 2023-07-20 Crinetics Pharmaceuticals, Inc. Uses of a somatostatin modulator for the treatment of carcinoid syndrome

Family Cites Families (88)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2265226A (en) * 1936-10-17 1941-12-09 Du Pont Cast resins having integral sheen
US2513014A (en) * 1946-11-18 1950-06-27 Abbott Lab Apparatus for implanting medicinal pellets subcutaneously
US3975350A (en) * 1972-08-02 1976-08-17 Princeton Polymer Laboratories, Incorporated Hydrophilic or hydrogel carrier systems such as coatings, body implants and other articles
US3921632A (en) * 1974-08-16 1975-11-25 Frank M Bardani Implant device
US4131604A (en) * 1977-11-23 1978-12-26 Thermo Electron Corporation Polyurethane elastomer for heart assist devices
US4285987A (en) * 1978-10-23 1981-08-25 Alza Corporation Process for manufacturing device with dispersion zone
US4298002A (en) * 1979-09-10 1981-11-03 National Patent Development Corporation Porous hydrophilic materials, chambers therefrom, and devices comprising such chambers and biologically active tissue and methods of preparation
US4386039A (en) * 1980-02-11 1983-05-31 Thermo Electron Corporation Process for forming an optically clear polyurethane lens or cornea
US4523005A (en) * 1981-10-30 1985-06-11 Thermedics, Inc. Extrudable polyurethane for prosthetic devices prepared from a diisocyanate, a polytetramethylene ether polyol, and 1,4-butane diol
US4751133A (en) * 1984-11-13 1988-06-14 Thermedics, Inc. Medical patches and processes for producing same
US5342622A (en) * 1986-05-16 1994-08-30 The State Of Victoria Subdermal biocompatible implants
US4959217A (en) 1986-05-22 1990-09-25 Syntex (U.S.A.) Inc. Delayed/sustained release of macromolecules
US4743673A (en) * 1986-12-19 1988-05-10 Tyndale Plains-Hunter, Ltd. Hydrophilic carboxy polyurethanes
US4871094A (en) * 1986-12-31 1989-10-03 Alcon Laboratories, Inc. Means and method for dispensing substances
US4994028A (en) * 1987-03-18 1991-02-19 Endocon, Inc. Injector for inplanting multiple pellet medicaments
AU608891B2 (en) 1987-09-24 1991-04-18 Merck & Co., Inc. Solubility modulated drug delivery device
US5035891A (en) * 1987-10-05 1991-07-30 Syntex (U.S.A.) Inc. Controlled release subcutaneous implant
US4954587A (en) * 1988-07-05 1990-09-04 Ciba-Geigy Corporation Dimethylacrylamide-copolymer hydrogels with high oxygen permeability
GB8820353D0 (en) * 1988-08-26 1988-09-28 Staniforth J N Controlled release tablet
PH30995A (en) * 1989-07-07 1997-12-23 Novartis Inc Sustained release formulations of water soluble peptides.
US5538739A (en) * 1989-07-07 1996-07-23 Sandoz Ltd. Sustained release formulations of water soluble peptides
US5273752A (en) * 1989-07-18 1993-12-28 Alza Corporation Controlled release dispenser comprising beneficial agent
US5468811A (en) * 1989-11-02 1995-11-21 National Patent Development Corporation Hydrophilic composite polymer articles formed from a settable paste comprising a mixture of hydrophilic polymer and unsaturated monomer
US5254662A (en) * 1990-09-12 1993-10-19 Polymedia Industries, Inc. Biostable polyurethane products
US5266325A (en) * 1990-09-28 1993-11-30 Hydro Med Science Division Of National Patent Development Corp. Preparation of homogeneous hydrogel copolymers
AU651654B2 (en) * 1992-01-14 1994-07-28 Endo Pharmaceuticals Solutions Inc. Manufacture of water-swellable hydrophilic articles and drug delivery devices
EP0550641B1 (en) * 1990-09-28 1994-05-25 Pfizer Inc. Dispensing device containing a hydrophobic medium
HU222249B1 (en) * 1991-03-08 2003-05-28 Amylin Pharmaceuticals Inc. Process for producing amyline antagonistic peptide derivatives and pharmaceutical preparatives containing them
AU652984B2 (en) 1992-01-14 1994-09-15 Endo Pharmaceuticals Solutions Inc. Preparation of homogeneous hydrogel copolymers
JP2530079B2 (en) 1992-02-05 1996-09-04 ナショナル・パテント・ディベロプメント・コーポレーション Manufacture of water-swellable hydrophilic products and drug release devices
DK0637323T3 (en) * 1992-04-24 1999-05-25 Polymer Technology Group Inc Copolymers and non-porous, semi-permeable membrane thereof and its use for permeation of molecules with a predetermined size
US5614223A (en) * 1992-05-04 1997-03-25 Digestive Care Inc. Intraoral medicament-releasing device
US5464933A (en) * 1993-06-07 1995-11-07 Duke University Synthetic peptide inhibitors of HIV transmission
EP0628959B1 (en) * 1993-06-11 2000-09-06 Nippon Chemi-Con Corporation Apparatus for supporting and connecting a magnetic field modulation head to be used for a photomagnetic recording
US5354835A (en) * 1993-07-23 1994-10-11 Saudi Basic Industries Corporation Desalination process
KR950007873A (en) 1993-09-20 1995-04-15 후꾸하라 요시하루 Biologics Sustained-Release Pharmaceutical Formulations
JPH0797338A (en) 1993-09-20 1995-04-11 Shiseido Co Ltd Sustained release preparation
JPH07252166A (en) 1993-09-20 1995-10-03 Shiseido Co Ltd Sustained release preparation
US5443505A (en) * 1993-11-15 1995-08-22 Oculex Pharmaceuticals, Inc. Biocompatible ocular implants
US6143718A (en) * 1995-06-07 2000-11-07 Amylin Pharmaceuticals, Inc. Treatment of Type II diabetes mellutis with amylin agonists
US5904935A (en) 1995-06-07 1999-05-18 Alza Corporation Peptide/protein suspending formulations
US5817343A (en) * 1996-05-14 1998-10-06 Alkermes, Inc. Method for fabricating polymer-based controlled-release devices
WO1998008533A1 (en) * 1996-08-30 1998-03-05 Peptech Limited Sustained peptide-release formulation
US5945128A (en) * 1996-09-04 1999-08-31 Romano Deghenghi Process to manufacture implants containing bioactive peptides
AUPO251096A0 (en) * 1996-09-23 1996-10-17 Cardiac Crc Nominees Pty Limited Polysiloxane-containing polyurethane elastomeric compositions
US5756127A (en) * 1996-10-29 1998-05-26 Wright Medical Technology, Inc. Implantable bioresorbable string of calcium sulfate beads
DK0996459T3 (en) * 1997-01-07 2006-01-16 Amylin Pharmaceuticals Inc Use of exendins and agonists thereof to reduce food intake
US5854127A (en) * 1997-03-13 1998-12-29 Micron Technology, Inc. Method of forming a contact landing pad
FI971385A0 (en) 1997-04-04 1997-04-04 Bioxid Oy Biocompatible composition, methodological method
US7101853B2 (en) * 1997-05-06 2006-09-05 Amylin Pharmaceuticals, Inc. Method for treating or preventing gastritis using amylin or amylin agonists
US7157555B1 (en) * 1997-08-08 2007-01-02 Amylin Pharmaceuticals, Inc. Exendin agonist compounds
US7220721B1 (en) * 1997-11-14 2007-05-22 Amylin Pharmaceuticals, Inc. Exendin agonist peptides
US6770623B1 (en) * 1997-12-09 2004-08-03 Eli Lilly And Company Stabilized teriparatide solutions
US6822016B2 (en) * 2001-09-10 2004-11-23 Johnson & Johnson Vision Care, Inc. Biomedical devices containing internal wetting agents
WO2000004037A1 (en) * 1998-07-14 2000-01-27 Amylin Pharmaceuticals, Inc. Uncoupling protein 4 (ucp-4) and methods of use
US6087334A (en) * 1998-08-21 2000-07-11 Amylin Pharmaceuticals, Inc. Anti-diabetic peptides
US20040071736A1 (en) * 1998-08-25 2004-04-15 Health Protection Agency Methods and compounds for the treatment of mucus hypersecretion
US7259136B2 (en) * 1999-04-30 2007-08-21 Amylin Pharmaceuticals, Inc. Compositions and methods for treating peripheral vascular disease
DK1143989T3 (en) * 1999-01-14 2007-04-16 Amylin Pharmaceuticals Inc Exendins for glucagon suppression
EP1067911B1 (en) * 1999-01-28 2008-09-10 Indevus Pharmaceuticals, Inc. Hydrogel compositions useful for the sustained release of macromolecules and methods of making same
US6454933B2 (en) * 1999-08-26 2002-09-24 Exxonmobil Research And Engineering Company Fluid atomization process
ES2227115T5 (en) * 2000-01-10 2014-10-30 Amylin Pharmaceuticals, Inc. Use of exendins and their agonists for the treatment of hypertriglyceridemia
AU2001252201A1 (en) * 2000-03-14 2001-09-24 Amylin Pharmaceuticals, Inc. Effects of glucagon-like peptide-1 (7-36) on antro-pyloro-duodenal motility
US6969480B2 (en) * 2000-05-12 2005-11-29 Matregen Corp. Method of producing structures using centrifugal forces
MXPA02011339A (en) * 2000-05-19 2003-04-25 Amylin Pharmaceuticals Inc Treatment of acute coronary syndrome with glp 1.
JP3498041B2 (en) * 2000-05-29 2004-02-16 科研製薬株式会社 Nasal formulation containing pralmorelin
US7118737B2 (en) * 2000-09-08 2006-10-10 Amylin Pharmaceuticals, Inc. Polymer-modified synthetic proteins
CN1568195B (en) * 2000-12-14 2011-03-02 安米林药品公司 Peptide YY and peptide YY agonists for treatment of metabolic disorders
US20050004000A1 (en) 2001-04-01 2005-01-06 Yoram Shechter Oral absorbed drugs
US7105489B2 (en) * 2002-01-22 2006-09-12 Amylin Pharmaceuticals, Inc. Methods and compositions for treating polycystic ovary syndrome
DE10208335A1 (en) * 2002-02-27 2003-09-04 Roehm Gmbh Pharmaceutical form and process for its preparation
US20060122106A1 (en) * 2002-06-14 2006-06-08 Amylin Pharmaceuticals, Inc Prevention and/or treatment of inflammatory bowel disease using pyy or agonists thereof
US20040097419A1 (en) * 2002-11-19 2004-05-20 Holger Petersen Organic compounds
US6942264B1 (en) * 2003-02-10 2005-09-13 Richard Mendez Disposable pet waste receptacle
CA2437639C (en) * 2003-08-11 2016-07-05 Valera Pharmaceuticals, Inc. Long term drug delivery devices with polyurethane based polymers and their manufacture
WO2005021023A1 (en) * 2003-09-02 2005-03-10 Pharmacia & Upjohn Company Therapeutical conversion
US20050143303A1 (en) * 2003-12-26 2005-06-30 Nastech Pharmaceutical Company Inc. Intranasal administration of glucose-regulating peptides
US8076288B2 (en) * 2004-02-11 2011-12-13 Amylin Pharmaceuticals, Inc. Hybrid polypeptides having glucose lowering activity
FR2870398B1 (en) * 2004-05-12 2006-07-07 Canon Europa Nv Naamlooze Venn SECURE CONNECTION DEVICE AND CORRESPONDING CONNECTION PLUG
WO2006009801A2 (en) * 2004-06-17 2006-01-26 Valera Pharmaceuticals, Inc. Compositions and methods for treating central precocious puberty
SE0402345L (en) * 2004-09-24 2006-03-25 Mederio Ag Measured drug dose
CA2601304C (en) * 2005-03-11 2010-08-17 Indevus Pharmaceuticals, Inc. Controlled release formulations of octreotide
US7759312B2 (en) * 2005-03-11 2010-07-20 Endo Pharmaceuticals Solutions Inc. Delivery of dry formulations of octreotide
US20070037897A1 (en) * 2005-08-12 2007-02-15 Guigui Wang Method for making contact lenses
EP1937297A2 (en) 2005-09-08 2008-07-02 Gastrotech Pharma A/S Use of a glp-1 molecule for treatment of biliary dyskinesia and/or biliary pain/discomfort
NZ580803A (en) * 2007-04-27 2012-03-30 Endo Pharmaceuticals Solutions Implant device release agents and methods of using same
WO2009158412A2 (en) * 2008-06-25 2009-12-30 Endo Pharmaceuticals Solutions Inc. Sustained delivery of exenatide and other polypeptides
CN102231976B (en) * 2008-06-25 2014-05-07 Endo药物方法有限公司 Octreotide implant having a release agent

Also Published As

Publication number Publication date
UA105496C2 (en) 2014-05-26
US20190388500A1 (en) 2019-12-26
IL210545A (en) 2015-06-30
BRPI0915523A2 (en) 2016-01-26
CN102231977A (en) 2011-11-02
ZA201100800B (en) 2011-10-26
AU2009268480A1 (en) 2010-01-14
US20090035343A1 (en) 2009-02-05
JP5840758B2 (en) 2016-01-06
JP2011527705A (en) 2011-11-04
RU2518745C2 (en) 2014-06-10
AU2009268480B2 (en) 2014-11-20
MX2011000354A (en) 2011-04-05
KR20110038056A (en) 2011-04-13
US20170049850A1 (en) 2017-02-23
CA2730391A1 (en) 2010-01-14
AU2009268480C1 (en) 2015-04-09
IL210545A0 (en) 2011-03-31
US20100247594A1 (en) 2010-09-30
JP2015071632A (en) 2015-04-16
EP2309979A1 (en) 2011-04-20
MX336209B (en) 2016-01-11
RU2011105029A (en) 2012-08-20
WO2010006236A1 (en) 2010-01-14
US7759312B2 (en) 2010-07-20

Similar Documents

Publication Publication Date Title
US20200353055A1 (en) Controlled release formulations of octreotide
US20190388500A1 (en) Delivery of dry formulations of octreotide
CA2729139C (en) Octreotide implant having a release agent

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20140703

MKLA Lapsed

Effective date: 20180710