US20120208755A1 - Compositions, Devices and Methods of Use Thereof for the Treatment of Cancers - Google Patents

Compositions, Devices and Methods of Use Thereof for the Treatment of Cancers Download PDF

Info

Publication number
US20120208755A1
US20120208755A1 US13/372,326 US201213372326A US2012208755A1 US 20120208755 A1 US20120208755 A1 US 20120208755A1 US 201213372326 A US201213372326 A US 201213372326A US 2012208755 A1 US2012208755 A1 US 2012208755A1
Authority
US
United States
Prior art keywords
glp
formulation
receptor agonist
suspension
cancer
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/372,326
Inventor
Karling Alice Leung
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.)
Intarcia Therapeutics Inc
Original Assignee
Intarcia Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Intarcia Therapeutics Inc filed Critical Intarcia Therapeutics Inc
Priority to US13/372,326 priority Critical patent/US20120208755A1/en
Assigned to INTARCIA THERAPEUTICS, INC. reassignment INTARCIA THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LEUNG, KARLING ALICE
Publication of US20120208755A1 publication Critical patent/US20120208755A1/en
Assigned to U.S. BANK NATIONAL ASSOCIATION, AS COLLATERAL AGENT reassignment U.S. BANK NATIONAL ASSOCIATION, AS COLLATERAL AGENT SECURITY AGREEMENT Assignors: INTARCIA THERAPEUTICS, INC.
Priority to US14/525,201 priority patent/US20150057227A1/en
Assigned to INTARCIA THERAPEUTICS, INC. reassignment INTARCIA THERAPEUTICS, INC. RELEASE BY SECURED PARTY (SEE DOCUMENT FOR DETAILS). Assignors: U.S. BANK NATIONAL ASSOCIATION, AS COLLATERAL AGENT
Priority to US15/597,788 priority patent/US10159714B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • A61K38/26Glucagons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M31/00Devices for introducing or retaining media, e.g. remedies, in cavities of the body
    • A61M31/002Devices for releasing a drug at a continuous and controlled rate for a prolonged period of time
    • 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
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/605Glucagons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M37/00Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin

Definitions

  • the present invention relates to formulations and methods for treating cancer. Aspects of the present invention provide formulations of glucagon-like peptide-1 (GLP-1) receptor agonists for use in mammals for the treatment of cancers.
  • GLP-1 glucagon-like peptide-1
  • Glycolysis is the metabolic pathway that converts glucose into pyruvate. The free energy released in this process is used to form the high-energy compounds ATP and NADH.
  • Increased aerobic glycolysis is seen in a variety of cancer cells, a phenomenon known as the Warburg theory. Under aerobic conditions, some tumor cells produce as much as 60% of their ATP through glycolysis (Nakashima et al., Cancer Res . (1984) 44:5702-5706) as opposed to normal cells which normally generate ATP through mitochondrial oxidative phosphorylation. In addition to increased aerobic glycolysis, increased glycolysis is also seen in tumors that reach a size that exceeds the capacity of blood supply due to hypoxia. For a review of the Warburg theory and implications thereof, see, e.g., Chen et al., J. Bioenerg. Biomenzbr . (2007) 39:267-274.
  • Glucagon-like peptide-1 (GLP-1) is an important hormone and a fragment of the human proglucagon molecule. GLP-1 is rapidly metabolized by a peptidase (dipeptidylpeptidase IV or DPP-IV). A fragment of GLP-1, glucagon-like peptide-1 (7-36) amide (also known as GLP-1 (7-36) amide, glucagon-like insulinotropic peptide, or GLIP) is a gastrointestinal peptide that potentiates the release of insulin in physiologic concentrations (Gutniak et al., N Engl J Med (1992) 14:326(20):1316-22).
  • GLP-1(7-36)amide and GLP-1(7-37) normalize fasting hyperglycemia in type 2 diabetic patients (Nauck, M. A., et al., Diabet. Med. 15(11):937-45 (1998)).
  • Exendin-4 a GLP-1 receptor agonist
  • Heloderma suspectuni venom is a molecule purified from Heloderma suspectuni venom (Eng, et al., Biol. Chem . (1992) 267:7402-7405) and shows structural relationship to the hormone GLP-1(7-36)amide.
  • Exendin-4 and truncated exendin-(9-39)amide specifically interact with the GLP-1 receptor on insulinoma-derived cells and on lung membranes (Göke et al., J Biol. Chem. (1993) 268:19650-19655).
  • Exendin-4 has approximately 53% identity to human GLP-1 (Pohl, et al., J. Biol. Chem .
  • exendin-4 is resistant to degradation by DPP-IV.
  • a glycine substitution confers resistance to degradation by DPP-1V (Young, et al., Diabetes (1999) 48(5):1026-1034).
  • the increased dependency of cancer cells on the glycolytic pathway is an important metabolic difference between normal and malignant cells.
  • the present invention provides a unique solution to disrupting cancer cell energy reliance on the glycolytic pathway.
  • the present invention relates to compositions, devices and methods for treating cancer.
  • the invention utilizes GLP-1 receptor agonists to restrict glucose as an energy source for cancer cells and tumors.
  • the GLP-1 receptor agonists can be used alone or in combination with other beneficial agents, such as anticancer agents, antidiabetic agents and the like, as well as in combination with anticancer treatment modalities, such as radiation, surgery and chemotherapeutic regimens.
  • the invention relates to a method of treating cancer in a subject in need of such treatment, comprising administering a GLP-1 receptor agonist to said subject.
  • the GLP-1 receptor agonist is a glucagon-like peptide-1 (GLP-1), a derivative of GLP-1, or an analog of GLP-1.
  • GLP-1 glucagon-like peptide-1
  • the GLP-1 receptor agonist is GLP(7-36)amide comprising the sequence of SEQ ID NO:1.
  • the GLP-1 receptor agonist is exenatide, a derivative of exenatide, or an analog of exenatide, such as a synthetic exenatide peptide comprising the sequence of SEQ ID NO:2.
  • the GLP-1 receptor agonist is selected from the group consisting of lixisenatide, liraglutide (VICTOZA®), albiglutide (SYNCRIATM), semaglutide, taspoglutide, BYETTA®, BYDUREON® and LY2189265.
  • formulations comprising the GLP-1 receptor agonist are delivered by injection.
  • the GLP-1 receptor agonist is delivered using an implanted drug delivery device, such as an osmotic delivery device, that provides continuous delivery of a suspension formulation of GLP-1 receptor agonist for a period of at least one month.
  • an implanted drug delivery device such as an osmotic delivery device
  • the GLP-1 receptor agonist and/or other beneficial agent is provided in a suspension formulation comprising: (a) a particle formulation comprising said GLP-1 receptor agonist and/or beneficial agent; and (b) a vehicle formulation, wherein the particle formulation is dispersed in the vehicle.
  • the suspension formulation may further comprise a particle formulation comprising a GLP-1 receptor agonist and/or beneficial agent and one or more stabilizers selected from the group consisting of carbohydrates, antioxidants, amino acids, buffers, and inorganic compounds.
  • the suspension formulation further comprises a non-aqueous, single-phase suspension vehicle comprising one or more polymers and one or more solvents. The suspension vehicle typically exhibits viscous fluid characteristics and the particle formulation is dispersed in the vehicle.
  • the suspension formulation comprises a particle formulation comprising a GLP-1 receptor agonist and/or a beneficial agent, a disaccharide (e.g., sucrose), methionine, and a buffer (e.g., citrate), and a non-aqueous, single-phase suspension vehicle comprising one or more pyrrolidone polymer (e.g., polyvinylpyrollidone) and one or more solvent (e.g., lauryl lactate, lauryl alcohol, benzyl benzoate, or mixtures thereof.
  • a GLP-1 receptor agonist e.g., sucrose
  • methionine e.g., methionine
  • a buffer e.g., citrate
  • a non-aqueous, single-phase suspension vehicle comprising one or more pyrrolidone polymer (e.g., polyvinylpyrollidone) and one or more solvent (e.g., lauryl lactate, lauryl alcohol, benzyl benzo
  • the particle formulations of the present invention may further comprise a buffer, for example, selected from the group consisting of citrate, histidine, succinate, and mixtures thereof.
  • a buffer for example, selected from the group consisting of citrate, histidine, succinate, and mixtures thereof.
  • the particle formulations of the present invention may further comprise an inorganic compound, for example, selected from the group consisting of citrate, histidine, succinate, and mixtures thereof.
  • an inorganic compound for example, selected from the group consisting of citrate, histidine, succinate, and mixtures thereof.
  • the one or more stabilizers in the particle formulations may comprise, for example, a carbohydrate selected from the group consisting of lactose, sucrose, trehalose, mannitol, cellobiose, and mixtures thereof.
  • the one or more stabilizers in the particle formulations may comprise, for example, a antioxidant selected from the group consisting of methionine, ascorbic acid, sodium thiosulfate, ethylenediaminetetraacetic acid (EDTA), citric acid, cysteins, thioglycerol, thioglycolic acid, thiosorbitol, butylated hydroxanisol, butylated hydroxyltoluene, and propyl gallate, and mixtures thereof.
  • a antioxidant selected from the group consisting of methionine, ascorbic acid, sodium thiosulfate, ethylenediaminetetraacetic acid (EDTA), citric acid, cysteins, thioglycerol, thioglycolic acid, thiosorbitol, butylated hydroxanisol, butylated hydroxyltoluene, and propyl gallate, and mixtures thereof.
  • the one or more stabilizers in the particle formulations may comprise an amino acid.
  • the solvent of the suspension vehicle of the present invention is selected from the group consisting of lauryl lactate, lauryl alcohol, benzyl benzoate, and mixtures thereof.
  • a polymer that can be used to formulate the suspension vehicle is a pyrrolidone (e.g., polyvinylpyrrolidone).
  • the polymer is a pyrrolidone and the solvent is benzyl benzoate.
  • the suspension formulation typically has an overall moisture content less than about 10 wt % and in a preferred embodiment less than about 5 wt %.
  • a beneficial agent such as an anticancer agent, in addition to the GLP-1 receptor agonist is delivered to said subject.
  • the anticancer agent is a chemotherapeutic agent and/or an anticancer antibody.
  • the additional beneficial agent can be delivered prior to, subsequent to or concurrent with the GLP-1 receptor agonist.
  • an implantable drug delivery device may be used to deliver formulations comprising an anticancer agent.
  • the device is an osmotic delivery device.
  • implantable drug delivery devices deliver a GLP-1 receptor agonist formulations and/or other beneficial agent formulations at a substantially uniform rate for a period of about one month to about a year.
  • Such devices may, for example, be implanted subcutaneously in convenient locations.
  • the present invention also includes methods of manufacturing the suspension formulations, particle formulations, suspension vehicles, and devices of the present invention as described herein.
  • FIGS. 1A and 1B present the sequences of two representative GLP-1 receptor agonists: FIG. 1A , glucagon-like peptide 1 (7-36)amide (GLP-1(7-36)amide) (SEQ ID NO:1), and FIG. 1B , synthetic exenatide peptide (SEQ ID NO:2).
  • FIG. 2 presents a partial cross-sectional view of one embodiment of an osmotic delivery device useful in the practice of the present invention.
  • peptide typically refers to a molecule comprising a chain of two or more amino acids (e.g., most typically L-amino acids, but also including, e.g., D-amino acids, modified amino acids, amino acid analogs, and/or amino acid mimetic). Peptides may also comprise additional groups modifying the amino acid chain, for example, functional groups added via post-translational modification.
  • post-translation modifications include, but are not limited to, acetylation, alkylation (including, methylation), biotinylation, glutamylation, glycylation, glycosylation, isoprenylation, lipoylation, phosphopantetheinylation, phosphorylation, selenation, and C-terminal amidation.
  • the term peptide also includes peptides comprising modifications of the amino terminus and/or the carboxy terminus. Modifications of the terminal amino group include, but are not limited to, des-amino, N-lower alkyl, N-di-lower alkyl, and N-acyl modifications.
  • Modifications of the terminal carboxy group include, but are not limited to, amide, lower alkyl amide, dialkyl amide, and lower alkyl ester modifications (e.g., wherein lower alkyl is C 1 -C 4 alkyl).
  • the terminal amino acid at one end of the peptide chain typically has a free amino group (i.e., the amino terminus).
  • the terminal amino acid at the other end of the chain typically has a free carboxyl group (i.e., the carboxy terminus).
  • the amino acids making up a peptide are numbered in order, starting at the amino terminus and increasing in the direction of the carboxy terminus of the peptide.
  • amino acid residue refers to an amino acid that is incorporated into a peptide by an amide bond or an amide bond mimetic.
  • GLP-1 receptor agonist refers to an agent capable of binding and activating the GLP-1 receptor.
  • the term includes GLP-1 hormones, as well as GLP-1 peptides, peptide analogs thereof, or peptide derivatives thereof.
  • GLP-1 receptor agonist are other molecules that are capable of binding and activating the GLP-1 receptor, such as without limitation, an exenatide peptide, a peptide analog thereof, or a peptide derivative thereof.
  • GLP-1 receptor agonists include exenatide having the amino acid sequence of exendin-4, GLP-1(7-36)amide, lixisenatide, liraglutide (VICTOZA®), albiglutide (SYNCRIATM), semaglutide, taspoglutide, BYETTA®, BYDUREON® and LY2189265.
  • the term also includes small molecules capable of binding and activating the GLP-1 receptor. See, e.g., Sloop et al., Diabetes (2010) 59:3099-3107.
  • anticancer agent refers to any agent that exhibits anti-tumor activity as defined below.
  • agents include, without limitation, chemotherapeutic agents (i.e., a chemical compound or combination of compounds useful in the treatment of cancer), anticancer antibodies, agents that disrupt nucleic acid transcription and/or translation, such as antisense oligonucleotides, small interfering RNA (siRNA), and the like.
  • anti-tumor activity is intended a reduction in the rate of cell proliferation, and hence a decline in growth rate of an existing tumor or in a tumor that arises during therapy, and/or destruction of existing neoplastic (tumor) cells or newly formed neoplastic cells, and hence a stabilization or decrease in the overall size of a tumor during therapy.
  • agents for treating types 1 and 2 diabetes such as but not limited to, GLP-1 receptor agonists; small molecules such as metformin, tolbutamide, glibenclamide, glipizide, gliquidone, glibornuride, tolazamide, sulfonylureas, meglitinides (e.g., repaglinide, and nateglinide); thiazolidinediones (TZDs), such as pioglitazone; SGLT 1 and SGLT 2 inhibitors; alpha glucosidase inhibitors; amylin (as well as synthetic analogs such as pramlintide); dipeptidyl peptidase IV (DPP-1V) inhibitors (e.g., saxagliptin, sitagliptin, alogliptin and
  • DPP-1V dipeptidyl peptidase IV
  • DPP-IV oral dipeptidyl peptidase-IV
  • DPP-IV or DPP-4 a GLP-1 that is cleavable by dipeptidyl peptidase-1V
  • an “antibody” intends a molecule that binds to an epitope of interest present in an antigen.
  • the term “antibody” as used herein includes antibodies obtained from both polyclonal and monoclonal preparations, as well as, the following: hybrid (chimeric) antibody molecules (see, for example, Winter et al., Nature (1991) 349:293-299; and U.S. Pat. No.
  • F(ab′)2 and F(ab) fragments Fv molecules (non-covalent heterodimers, see, for example, Inbar et al., Proc Natl Acad Sci USA (1972) 69:2659-2662; and Ehrlich et al., Biochem (1980) 19:4091-4096); single-chain Fv molecules (sFv) (see, for example, Huston et al., Proc Natl Acad Sci USA (1988) 85:5879-5883); dimeric and trimeric antibody fragment constructs; diabodies; avamers; aptamers; affitins; affitins; anticalins; affibody molecules; designed ankyrin repeat proteins; domain antibodies; minibodies (see, e.g., Pack et al., Biochem (1992) 31:1579-1584; Cumber et al., J Immunology (1992) 149B:120-126); humanized antibody molecules (see, for example
  • the term “monoclonal antibody” refers to an antibody composition having a homogeneous antibody population.
  • the term is not limited regarding the species or source of the antibody, nor is it intended to be limited by the manner in which it is made.
  • the term encompasses whole immunoglobulins as well as fragments such as Fab, F(ab′) 2 , Fv, and other fragments, as well as chimeric and humanized homogeneous antibody populations, that exhibit immunological binding properties of the parent monoclonal antibody molecule.
  • anti-cancer antibody encompasses antibodies that have been designed to target cancer cells, particularly cell-surface antigens residing on cells of a particular cancer of interest.
  • Vehicle refers to a medium used to carry a compound, e.g., a drug.
  • Vehicles of the present invention typically comprise components such as polymers and solvents.
  • the suspension vehicles of the present invention typically comprise solvents and polymers that are used to prepare suspension formulations further comprising drug particle formulations.
  • phase separation refers to the formation of multiple phases (e.g., liquid or gel phases) in the suspension vehicle, such as when the suspension vehicle contacts the aqueous environment.
  • the suspension vehicle is formulated to exhibit phase separation upon contact with an aqueous environment having less than approximately 10% water.
  • single-phase refers to a solid, semisolid, or liquid homogeneous system that is physically and chemically uniform throughout.
  • dissolving refers to dissolving, dispersing, suspending, or otherwise distributing a compound, for example, a peptide, in a suspension vehicle.
  • a “homogeneous suspension” typically refers to a particle that is insoluble in a suspension vehicle and is distributed uniformly in a suspension vehicle.
  • chemically stable refers to formation in a formulation of an acceptable percentage of degradation products produced over a defined period of time by chemical pathways, such as deamidation, (usually by hydrolysis), aggregation, or oxidation.
  • a physically stable formulation refers to formation in a formulation of an acceptable percentage of aggregates (e.g., dimers and other higher molecular weight products). Further, a physically stable formulation does not change its physical state as, for example, from liquid to solid, or from amorphous to crystal form.
  • viscosity typically refers to a value determined from the ratio of shear stress to shear rate (see, e.g., Considine, D. M. & Considine, G. D., Encyclopedia of Chemistry, 4th Edition, Van Nostrand, Reinhold, N.Y., 1984) essentially as follows:
  • V/L the velocity per layer thickness (shear rate).
  • the ratio of shear stress to shear rate defines viscosity.
  • Measurements of shear stress and shear rate are typically determined using parallel plate rheometery performed under selected conditions (for example, a temperature of about 37° C.).
  • Other methods for the determination of viscosity include, measurement of a kinematic viscosity using a viscometer, for example, a Cannon-Fenske viscometer, a Ubbelohde viscometer for the Cannon-Fenske opaque solution, or a Ostwald viscometer.
  • suspension vehicles of the present invention have a viscosity sufficient to prevent particles suspended therein from settling during storage and use in a method of delivery, for example, in an implantable, drug delivery device.
  • non-aqueous refers to an overall moisture content, for example, of a suspension formulation, typically of less than or equal to about 10 wt %, preferably less than or equal to about 5 wt %, and more preferably less than about 4 wt %.
  • subject refers to any member of the subphylum chordata, including, without limitation, humans and other primates, including non-human primates such as rhesus macaque, chimpanzees and other apes and monkey species; farm animals such as cattle, sheep, pigs, goats and horses; domestic mammals such as dogs and cats; laboratory animals including rodents such as mice, rats and guinea pigs; birds, including domestic, wild and game birds such as chickens, turkeys and other gallinaceous birds, ducks, geese, and the like.
  • the term does not denote a particular age. Thus, both adult and newborn individuals are intended to be covered.
  • the terms “drug,” “therapeutic agent”, and “beneficial agent” are used interchangeably to refer to any therapeutically active substance that is delivered to a subject to produce a desired beneficial effect.
  • the drug is a GLP-1 receptor agonist, e.g., GLP-1 (7-36)amide, exenatide, and derivatives or analogs thereof.
  • GLP-1 receptor agonist e.g., GLP-1 (7-36)amide, exenatide, and derivatives or analogs thereof.
  • the devices and methods of the present invention are well suited for the delivery of polypeptides as well as small molecules and combinations thereof.
  • osmotic delivery device typically refers to a device used for delivery of one or more GLP-1 receptor agonists, or other beneficial agents to a subject, wherein the device comprises, for example, a reservoir (made, for example, from a titanium alloy) having a lumen that contains, in one chamber, a beneficial agent formulation (e.g., comprising one or more beneficial agent) and, in another chamber, an osmotic agent formulation.
  • a beneficial agent formulation e.g., comprising one or more beneficial agent
  • a piston assembly positioned in the lumen isolates the beneficial agent formulation from the osmotic agent formulation.
  • a semi-permeable membrane also termed a semi-permeable plug
  • a diffusion moderator (which defines a delivery orifice through which the beneficial agent formulation exits the device) is positioned at a second distal end of the reservoir adjacent the suspension formulation.
  • the piston assembly and the diffusion moderator define a chamber that contains the beneficial agent formulation and the piston assembly and the semipermeable membrane define a chamber that contains the osmotic agent formulation.
  • the terms “flow modulator,” “diffusion modulator,” “flow moderator,” and “diffusion moderator” are used interchangeably herein.
  • the osmotic delivery device is implanted within the subject, for example, subcutaneously (e.g., in the inside, outside, or back of the upper arm; or in the abdominal area).
  • An exemplary osmotic delivery device is the DUROe delivery device.
  • the present invention relates to methods of treating cancer in a subject in need of treatment, including, but not limited to, treating hematological tumors and solid tumors.
  • the method comprises providing delivery of a GLP-1 receptor agonist formulation to a subject in need thereof.
  • the GLP-1 receptor agonist formulation is delivered using an osmotic delivery device at a substantially uniform rate. The length of delivery of the formulation is determined based on the cancer being treated.
  • the administration period is for at least about one month, at least about one month to about one year, at least about three months to about one year, at least about four months to about one year, at least about five months to about one year, at least about six months to about one year, at least about eight months to about one year, at least about nine months to about one year, or at least about 10 months to about one year.
  • the period of administration can also exceed one year if necessary, such as from one year to two years.
  • the method may further include subcutaneously inserting an osmotic delivery device, loaded with the GLP-1 receptor agonist formulation, into the subject.
  • the GLP-1 receptor agonist is delivered parenterally (including by subcutaneous, intravenous, intramedullary, intraarticular, intramuscular, or intraperitoneal injection) rectally, topically, transdermally, intranasally, by inhalation, or orally (for example, in capsules, suspensions, or tablets).
  • injectable formulations of GLP-1 agonists are known and include, without limitation, lixisenatide, liraglutide (VICTOZA®), albiglutide (SYNCRIATM), semaglutide, taspoglutide, BYETTA®, BYDLIREON® and LY2189265.
  • the formulation comprises a glucagon-like peptide-1 (GLP-1), a derivative of GLP-1, or an analog of GLP-1.
  • GLP-1 glucagon-like peptide-1
  • the GLP-1 receptor agonist is GLP-1(7-36)amide shown in FIG. 1A (SEQ ID NO:1).
  • the formulation comprises exenatide, a derivative of exenatide, or an analog of exenatide.
  • the exenatide is the exenatide peptide shown in FIG. 1B (SEQ ID NO:2).
  • additional beneficial agents are provided with the GLP-1 receptor agonist formulations, such as anticancer agents, including without limitation, chemotherapeutic agents, anticancer antibodies, antisense nucleotides, siRNA, anticancer vaccines, and the like. Such additional beneficial agents are described in detail below.
  • Administration of these agents is not limited to any particular delivery system and may include, without limitation, delivery using osmotic delivery devices as described herein if the agent is suitable for such delivery, or may be parenteral (including subcutaneous, intravenous, intramedullary, intraarticular, intramuscular, or intraperitoneal injection), rectal, topical, transdermal, intranasal, by inhalation, or oral (for example, in capsules, suspensions, or tablets).
  • Administration of the additional agents to an individual may occur in a single dose or in repeat administrations, and in any of a variety of physiologically acceptable salt forms, and/or with an acceptable pharmaceutical carrier and/or additive as part of a pharmaceutical composition.
  • physiologically acceptable salt forms and standard pharmaceutical formulation techniques and excipients are well known to persons skilled in the art (see, e.g., Physicians' Desk Reference (PDR) 2009, 63th ed. (PDR.net), Medical Economics Company; and Remington: The Science and Practice of Pharmacy, eds. Gennado et al., 21th ed, Lippincott, Williams & Wilkins, 2005).
  • the GLP-1 receptor agonist and/or suitable additional beneficial agents are provided in a suspension formulation, comprising a particle formulation and a suspension vehicle.
  • the particle formulation includes, but is not limited to, the GLP-1 receptor agonist or other agent of interest and one or more stabilizers.
  • the one or more stabilizers are typically selected from the group consisting of carbohydrates, antioxidants, amino acids, and buffers.
  • the suspension vehicle is typically a non-aqueous, single-phase suspension vehicle comprising one or more polymers and one or more solvents. The suspension vehicle exhibits viscous fluid characteristics.
  • the particle formulation is uniformly dispersed in the vehicle.
  • the particle formulation of the present invention typically includes one or more of the following stabilizers: one or more carbohydrates (e.g., a disaccharide, such as, lactose, sucrose, trehalose, cellobiose, and mixtures thereof); one or more antioxidants (e.g., methionine, ascorbic acid, sodium thiosulfate, ethylenediaminetetraacetic acid (EDTA), citric acid, butylated hydroxyltoluene, and mixtures thereof); and one or more buffers (e.g., citrate, histidine, succinate, and mixtures thereof).
  • the particle formulation comprises a GLP-1 receptor agonist, sucrose, methionine, and citrate buffer.
  • the ratio of the GLP-1 receptor agonist to sucrose+methionine is typically about 1/20, about 1/10, about 1/5, about 1/2, about 2/1, about 5/1, about 10/1, or about 20/1, preferably between about 1/5 to 5/1, more preferably between about 1/3 to 3/1.
  • the particle formulation is preferably a particle formulation prepared by spray drying and has a low moisture content, preferably less than or equal to about 10 wt %, more preferably less or equal to about 5 wt %.
  • the particle formulation can be lyophilized.
  • the suspension vehicle for use in the present formulations comprises one or more solvents and one or more polymers.
  • the solvent is selected from the group consisting of lauryl lactate, lauryl alcohol, benzyl benzoate, and mixtures thereof. More preferably the solvent is lauryl lactate or benzyl benzoate.
  • the polymer is a pyrrolidone polymer.
  • the polymer is polyvinylpyrrolidone (e.g., polyvinylpyrrolidone K-17, which typically has an approximate average molecular weight range of 7,900-10,800).
  • the solvent consists essentially of benzyl benzoate and polyvinylpyrrolidone.
  • the suspension formulation typically has a low overall moisture content, for example, less than or equal to about 10 wt % and in a preferred embodiment less than or equal to about 5 wt %.
  • GLP-1 including three forms of the peptide, GLP-1(1-37), GLP-1(7-37) and GLP-1(7-36)amide, as well as peptide analogs of GLP-1 have been shown to stimulate insulin secretion (i.e., they are insulinotropic), which results in decreases in serum glucose concentrations (see, e.g., Mojsov, S., Int. J. Peptide Protein Research (1992) 40:333-343).
  • the sequence of GLP-1(7-36)amide is shown in FIG. 1A and SEQ ID NO:1.
  • GLP-1 peptide derivatives and peptide analogs demonstrating insulinotropic action are known in the art (see, e.g., U.S. Pat. Nos. 5,118,666; 5,120,712; 5,512,549; 5,545,618; 5,574,008; 5,574,008; 5,614,492; 5,958,909; 6,191,102; 6,268,343; 6,329,336; 6,451,974; 6,458,924; 6,514,500; 6,593,295; 6,703,359; 6,706,689; 6,720,407; 6,821,949; 6,849,708; 6,849,714; 6,887,470; 6,887,849; 6,903,186; 7,022,674; 7,041,646; 7,084,243; 7,101,843; 7,138,486; 7,141,547; 7,144,863; and 7,199,217, all of which are incorporated herein by reference in their entireties), as well
  • GLP-1 peptide derivative useful in the practice of the present invention is VICTOZA® (liraglutide; U.S. Pat. Nos. 6,268,343, 6,458,924, 7,235,627, incorporated herein by reference in their entireties).
  • VICTOZA® liraglutide
  • Once-daily injectable VICTOZA® (liraglutide) is commercially available in the United States, Europe, and Japan.
  • Other injectable GLP-1 peptides for use with the present invention are described above and include, without limitation taspoglutide, albiglutide (SYNCRIATM), LY2189265 and semaglutide.
  • GLP-1 family of GLP-1 peptides, GLP-1 peptide derivatives and GLP-1 peptide analogs having insulinotropic activity is referred to collectively as “GLP-1.”
  • the molecule exenatide has the amino acid sequence of exendin-4 (Kolterman O. G., et al., J. Clin. Endocrinol. Metab . (2003) 88(7):3082-3089) and is produced by chemical synthesis or recombinant expression. Twice-daily injectable exenatide is commercially available in the United States and Europe, and sold under the tradename of BYETTA®. Another injectable exenatide under development is BYDUREON®. Exendin-3 and exendin-4 are known in the art and were originally isolated from Heloderma spp. (Eng, et al., J. Biol. Chem .
  • exenatide peptide derivatives and peptide analogs are known in the art (see, e.g., U.S. Pat. Nos.
  • exenatide derivative useful in the practice of the present invention is lixisenatide (see, e.g., U.S. Pat. No. 6,528,486, incorporated herein by reference in its entirety).
  • lixisenatide see, e.g., U.S. Pat. No. 6,528,486, incorporated herein by reference in its entirety.
  • exenatide peptides e.g., including exendin-3, exendin-4, and exendin-4-amide
  • exenatide peptide derivatives e.g., including exendin-3, exendin-4, and exendin-4-amide
  • exenatide peptide derivatives e.g., including exenatide peptide derivatives
  • exenatide peptide analogs is referred to collectively as “exenatide.”
  • the present invention utilizes particle formulations of GLP-1 receptor agonists described above that can be used to prepare suspension formulations.
  • the GLP-1 receptor agonists for use with the present invention shall not be limited by method of synthesis or manufacture and shall include those obtained from natural sources, or synthesized or manufactured by recombinant (whether produced from cDNA or genomic DNA), synthetic, transgenic, and gene-activated methods.
  • the GLP-1 receptor agonist is a GLP-1 peptide or an exendin peptide (as described above), for example, GLP-1(7-36)amide or exenatide, such as the exenatide peptide shown in FIG. 1B and SEQ ID NO:2.
  • the present invention also includes combinations of two or more such agents, for example, GLP-1(7-36)amide and GIP.
  • Particle formulations are preferably chemically and physically stable for at least one month, preferably at least three months, more preferably at least six months, more preferably at least 12 months at delivery temperature.
  • the delivery temperature is typically normal human body temperature, for example, about 37° C., or slightly higher, for example, about 40° C.
  • particle formulations are preferably chemically and physically stable for at least three months, preferably at least six months, more preferably at least 12 months, at storage temperature.
  • Examples of storage temperatures include refrigeration temperature, for example, about 5° C., or room temperature, for example, about 25° C.
  • a particle formulation may be considered chemically stable if less than about 25%, preferably less than about 20%, more preferably less than about 15%, more preferably less than about 10%, and more preferably less than about 5% breakdown products of the peptide particles are formed after about three months, preferably after about six months, preferably after about 12 months at delivery temperature and after about six months, after about 12 months, and preferably after about 24 months at storage temperature.
  • a particle formulation may be considered physically stable if less than about 10%, preferably less than about 5%, more preferably less than about 3%, more preferably less than 1% aggregates of the peptide particles are formed after about three months, preferably after about six months, at delivery temperature and about 6 months, preferably about 12 months, at storage temperature.
  • Tg glass transition temperature
  • Tg glass transition temperature
  • particle formulations are formable into particles using processes such as spray drying, lyophilization, desiccation, milling, granulation, ultrasonic drop creation, crystallization, precipitation, or other techniques available in the art for forming particles from a mixture of components.
  • the particles are preferably substantially uniform in shape and size.
  • a typical spray dry process may include, for example, loading a spray solution containing a peptide, for example, GLP-1(7-36)amide or exenatide, and stabilizing excipients into a sample chamber.
  • the sample chamber is typically maintained at a desired temperature, for example, refrigeration to room temperature. Refrigeration generally promotes stability of the protein.
  • a solution, emulsion, or suspension is introduced to the spray dryer where the fluid is atomized into droplets. Droplets can be formed by use of a rotary atomizer, pressure nozzle, pneumatic nozzle, or sonic nozzle. The mist of droplets is immediately brought into contact with a drying gas in a drying chamber. The drying gas removes solvent from the droplets and carries the particles into a collection chamber.
  • yield of the spray dry process depends in part on the particle formulation.
  • the particles are sized such that they can be delivered via an implantable drug delivery device. Uniform shape and size of the particles typically helps to provide a consistent and uniform rate of release from such a delivery device; however, a particle preparation having a non-normal particle size distribution profile may also be used.
  • the size of the particles is less than about 30%, preferably is less than about 20%, more preferably is less than about than 10%, of the diameter of the delivery orifice.
  • particle sizes may be preferably less than about 50 microns, more preferably less than about 10 microns, more preferably in a range from about 3 to about 7 microns.
  • the orifice is about 0.25 mm (250 microns) and the particle size is approximately 3-5 microns.
  • particle sizes may be preferably less than about 50 microns, more preferably less than about 10 microns, more preferably in a range from about 3 to about 7 microns.
  • a particle formulation for use with the present invention comprises one or more GLP-1 receptor agonists, as described above and one or more stabilizers.
  • the stabilizers may be, for example, carbohydrate, antioxidant, amino acid, buffer, or inorganic compound.
  • the amounts of stabilizers in the particle formulation can be determined experimentally based on the activities of the stabilizers and buffers and the desired characteristics of the formulation.
  • the amount of carbohydrate in the formulation is determined by aggregation concerns. In general, the carbohydrate level should not be too high so as to avoid promoting crystal growth in the presence of water due to excess carbohydrate unbound to insulinotropic peptide.
  • the amount of antioxidant in the formulation is determined by oxidation concerns, while the amount of amino acid in the formulation is determined by oxidation concerns and/or formability of particles during spray drying.
  • the amount of buffer components in the formulation is determined by pre-processing concerns, stability concerns, and formability of particles during spray drying. Buffer may be required to stabilize the GLP-1 receptor agonist during processing, e.g., solution preparation and spray drying, when all excipients are solubilized.
  • carbohydrates examples include, but are not limited to, monosaccharides (e.g., fructose, maltose, galactose, glucose, D-mannose, and sorbose), disaccharides (e.g., lactose, sucrose, trehalose, and cellobiose), polysaccharides (e.g., raffinose, melezitose, maltodextrins, dextrans, and starches), and alditols (acyclic polyols; e.g., mannitol, xylitol, maltitol, lactitol, xylitol sorbitol, pyranosyl sorbitol, and myoinsitol).
  • Preferred carbohydrates include non-reducing sugars, such as sucrose, trehalose, and raffinose.
  • antioxidants examples include, but are not limited to, methionine, ascorbic acid, sodium thiosulfate, catalase, platinum, ethylenediaminetetraacetic acid (EDTA), citric acid, cysteins, thioglycerol, thioglycolic acid, thiosorbitol, butylated hydroxanisol, butylated hydroxyltoluene, and propyl gallate.
  • amino acids examples include, but are not limited to, arginine, methionine, glycine, histidine, alanine, L-leucine, glutamic acid, iso-leucine, L-threonine, 2-phenylamine, valine, norvaline, praline, phenylalanine, trytophan, serine, asparagines, cysteine, tyrosine, lysine, and norleucine.
  • Preferred amino acids include those that readily oxidize, e.g., cysteine, methionine, and trytophan.
  • buffers examples include, but are not limited to, citrate, histidine, succinate, phosphate, maleate, tris, acetate, carbohydrate, and gly-gly.
  • Preferred buffers include citrate, histidine, succinate, and tris. It is to be understood that buffers can be added to the solution before formation of the particles, for example, by spray drying. However, after the dry particle formation is prepared, the buffer component no longer serves as a buffer in the dried particles. For ease of reference herein, when referring to buffer components, the term buffer is used.
  • inorganic compounds that may be included in the particle formulation include, but are not limited to, NaCl, Na 2 SO 4 , NaHCO 3 , KCl, KH 2 PO 4 , CaCl 2 , and MgCl 2 .
  • the particle formulation may include other excipients, such as but not limited to surfactants and salts.
  • surfactants include, but are not limited to, Polysorbate 20, Polysorbate 80, PLURONIC®, F68, and sodium docecyl sulfate (SDS).
  • excipients include, but are not limited to, mannitol and glycine.
  • salts include, but are not limited to, sodium chloride, calcium chloride, and magnesium chloride.
  • the particle formulation comprises, for example, exenatide peptide, sucrose (carbohydrate), methionine (antioxidant), and sodium citrate/citric acid.
  • All components included in the particle formulation are typically acceptable for pharmaceutical use in mammals, in particular, in humans.
  • Particle size distribution of the dry particle powder can be well controlled (0.1 micron ⁇ 20 micron), for example, by using the methods of spray drying or lyophilization to prepare the particle formulations.
  • the process parameters for formation of the dry powder are optimal to produce particles with desired particle size distribution, density, and surface area.
  • the selected excipients and stabilizers in the particle formulation may provide, for example, the following functions: density modification of the dry powder; preservation of the peptide chemical stability; maintenance of the peptide's physical stability (e.g., high glass transition temperature, and avoiding phase to phase transition); producing homogenous dispersions in suspension; and modification of hydrophobicity and/or hydrophilicity to manipulate dry powder solubility in selected solvents.
  • GLP-1 receptor agonists can be formulated into dried powders in solid state, which preserves maximum chemical and biological stability of proteins or peptides.
  • the particle formulation offers long term storage stability at high temperature, and therefore, allows delivery to a subject of stable and biologically effective peptide for extended periods of time.
  • particle formulations described above are with reference to GLP-1 receptor agonists, such particle formulations can also be formed with any other suitable agents, such as other suitable beneficial polypeptides, including suitable anticancer polypeptides, antibodies and the like, described in detail below.
  • the suspension formulation includes a suspension vehicle to provide a stable environment in which the GLP-1 receptor agonist particle formulation (or other suitable particle formulation) is dispersed.
  • the particle formulations are chemically and physically stable (as described above) in the suspension vehicle.
  • the suspension vehicle typically comprises one or more polymers and one or more solvents that form a solution of sufficient viscosity to uniformly suspend the particles comprising the GLP-1 receptor agonist or other suitable agent.
  • the suspension formulations can be used with any suitable agents, such as other suitable beneficial polypeptides, including suitable anticancer polypeptides, antibodies and the like, described in detail below.
  • the viscosity of the suspension vehicle is typically sufficient to prevent the particle formulation from settling during storage and use in a method of delivery, for example, in an implantable, drug delivery device.
  • the suspension vehicle is biodegradable in that the suspension vehicle disintegrates or breaks down over a period of time in response to a biological environment.
  • the disintegration of the suspension vehicle may occur by one or more physical or chemical degradative processes, such as by enzymatic action, oxidation, reduction, hydrolysis (e.g., proteolysis), displacement (e.g., ion exchange), or dissolution by solubilization, emulsion or micelle formation.
  • hydrolysis e.g., proteolysis
  • displacement e.g., ion exchange
  • dissolution by solubilization, emulsion or micelle formation emulsion or micelle formation.
  • the solvent in which the polymer is dissolved may affect characteristics of the suspension formulation, such as the behavior of the particle formulation during storage.
  • a solvent may be selected in combination with a polymer so that the resulting suspension vehicle exhibits phase separation upon contact with the aqueous environment.
  • the solvent may be selected in combination with the polymer so that the resulting suspension vehicle exhibits phase separation upon contact with the aqueous environment having less than approximately about 10% water.
  • the solvent may be an acceptable solvent that is not miscible with water.
  • the solvent may also be selected so that the polymer is soluble in the solvent at high concentrations, such as at a polymer concentration of greater than about 30%. However, typically particles comprising the GLP-1 receptor agonists are substantially insoluble in the solvent.
  • solvents useful in the practice of the present invention include, but are not limited to, lauryl alcohol, benzyl benzoate, benzyl alcohol, lauryl lactate, decanol (also called decyl alcohol), ethyl hexyl lactate, and long chain (C 8 to C 24 ) aliphatic alcohols, esters, or mixtures thereof.
  • the solvent used in the suspension vehicle may be “dry,” in that it has a low moisture content.
  • Preferred solvents for use in formulation of the suspension vehicle include lauryl lactate, lauryl alcohol, benzyl benzoate, and combinations thereof.
  • polymers for formulation of the suspension vehicles include, but are not limited to, a polyester (e.g., polylactic acid or polylacticpolyglycolic acid), pyrrolidone polymer (e.g., polyvinylpyrrolidone (PVP) having a molecular weight ranging from approximately 2,000 to approximately 1,000,000), ester or ether of an unsaturated alcohol (e.g., vinyl acetate), polyoxyethylenepolyoxypropylene block copolymer, or mixtures thereof.
  • the polymer is PVP having a molecular weight of 2,000 to 1,000,000.
  • the polymer is polyvinylpyrrolidone K-17 (typically having an approximate average molecular weight range of 7,900-10,800).
  • Polyvinylpyrrolidone can be characterized by its K-value (e.g., K-17), which is a viscosity index.
  • K-17 a viscosity index.
  • the polymer used in the suspension vehicle may include one or more different polymers or may include different grades of a single polymer.
  • the polymer used in the suspension vehicle may also be dry or have a low moisture content.
  • a suspension vehicle according to the present invention may vary in composition based on the desired performance characteristics.
  • the suspension vehicle may comprise about 40% to about 80% (w/w) polymer(s) and about 20% to about 60% (w/w) solvent(s).
  • Preferred embodiments of a suspension vehicle include vehicles formed of polymer(s) and solvent(s) combined at the following ratios: about 25% solvent and about 75% polymer; about 50% solvent and about 50% polymer; about 75% solvent and about 25% polymer.
  • the suspension vehicle may exhibit Newtonian behavior.
  • the suspension vehicle is typically formulated to provide a viscosity that maintains a uniform dispersion of the particle formulation for a predetermined period of time. This helps facilitate making a suspension formulation tailored to provide controlled delivery of the insulinotropic peptide at a desired rate.
  • the viscosity of the suspension vehicle may vary depending on the desired application, the size and type of the particle formulation, and the loading of the particle formulation in the suspension vehicle.
  • the viscosity of the suspension vehicle may be varied by altering the type or relative amount of the solvent or polymer used.
  • the suspension vehicle may have a viscosity ranging from about 100 poise to about 1,000,000 poise, preferably from about 1,000 poise to about 100,000 poise.
  • the viscosity may be measured at a selected temperature, for example, 33° C., at a shear rate of 10 ⁇ 4 /sec, using a parallel plate rheometer.
  • the viscosity of the suspension vehicle ranges from approximately 5,000 poise to approximately 50,000 poise, such as about 7,000 poise to about 40,000 poise, about 8,000 poise to about 20,000 poise, about 9,000 poise to about 25,000 poise, about 10,000 poise to about 20,000 poise, and the like.
  • the viscosity range is between about 12,000 to about 18,000 poise at 33° C.
  • the suspension vehicle may exhibit phase separation when contacted with the aqueous environment; however, typically the suspension vehicle exhibits substantially no phase separation as a function of temperature. For example, at a temperature ranging from approximately 0° C. to approximately 70° C. and upon temperature cycling, such as cycling from 4° C. to 37° C. to 4° C., the suspension vehicle typically exhibits no phase separation.
  • the suspension vehicle may be prepared by combining the polymer and the solvent under dry conditions, such as in a dry box.
  • the polymer and solvent may be combined at an elevated temperature, such as from approximately 40° C. to approximately 70° C., and allowed to liquefy and form the single phase.
  • the ingredients may be blended under vacuum to remove air bubbles produced from the dry ingredients.
  • the ingredients may be combined using a conventional mixer, such as a dual helix blade or similar mixer, set at a speed of approximately 40 rpm. However, higher speeds may also be used to mix the ingredients.
  • the suspension vehicle may be cooled to room temperature.
  • Differential scanning calorimetry (DSC) may be used to verify that the suspension vehicle is a single phase.
  • the components of the vehicle e.g., the solvent and/or the polymer
  • the particle formulation comprising a GLP-1 receptor agonist, or other suitable agent, is added to the suspension vehicle to form a suspension formulation.
  • the suspension formulation may be prepared by dispersing the particle formulation in the suspension vehicle.
  • the suspension vehicle may be heated and the particle formulation added to the suspension vehicle under dry conditions.
  • the ingredients may be mixed under vacuum at an elevated temperature, such as from about 40° C. to about 70° C.
  • the ingredients may be mixed at a sufficient speed, such as from about 40 rpm to about 120 rpm, and for a sufficient amount of time, such as about 15 minutes, to achieve a uniform dispersion of the particle formulation in the suspension vehicle.
  • the mixer may be a dual helix blade or other suitable mixer.
  • the resulting mixture may be removed from the mixer, sealed in a dry container to prevent water from contaminating the suspension formulation, and allowed to cool to room temperature before further use, for example, loading into an implantable, drug delivery device, unit dose container, or multiple-dose container.
  • the suspension formulation typically has an overall moisture content of less than about 10 wt %, preferably less than about 5 wt %, and more preferably less than about 4 wt %.
  • suspension formulations of the present invention are exemplified herein below with reference to exenatide and GLP-1(7-36)amide as representative GLP-1 receptor agonists (see, Example 3 and Example 4). These examples are not intended to be limiting.
  • the components of the suspension vehicle provide biocompatibility.
  • Components of the suspension vehicle offer suitable chemico-physical properties to form stable suspensions of, for example, dry powder particle formulations. These properties include, but are not limited to, the following: viscosity of the suspension; purity of the vehicle; residual moisture of the vehicle; density of the vehicle; compatibility with the dry powders; compatibility with implantable devices; molecular weight of the polymer; stability of the vehicle; and hydrophobicity and hydrophilicity of the vehicle. These properties can be manipulated and controlled, for example, by variation of the vehicle composition and manipulation of the ratio of components used in the suspension vehicle.
  • the suspension formulations described herein may be used in an implantable, drug delivery device to provide sustained delivery of a compound over an extended period of time, such as over weeks, months, or up to about one year.
  • Such an implantable drug delivery device is typically capable of delivering the compound at a desired flow rate over a desired period of time.
  • the suspension formulation may be loaded into the implantable, drug delivery device by conventional techniques.
  • the suspension formulation may be delivered, for example, using an osmotically, mechanically, electromechanically, or chemically driven drug delivery device.
  • the active agent in the suspension formulation is delivered at a flow rate that is therapeutically effective to the subject in need of treatment.
  • the active agent such as GLP-1(7-36)amide, exenatide, or other suitable beneficial agent, may be delivered over a period ranging from more than about one week to about one year or more, preferably for about one month to about a year or more, more preferably for about three months to about a year or more.
  • the implantable, drug delivery device may include a reservoir having at least one orifice through which the agent is delivered.
  • the suspension formulation may be stored within the reservoir.
  • the implantable, drug delivery device is an osmotic delivery device, wherein delivery of the drug is osmotically driven.
  • the DUROS® delivery device typically consists of a cylindrical reservoir which contains the osmotic engine, piston, and drug formulation.
  • the reservoir is capped at one end by a controlled-rate water-permeable membrane and capped at the other end by a diffusion moderator through which drug formulation is released from the drug reservoir.
  • the piston separates the drug formulation from the osmotic engine and utilizes a seal to prevent the water in the osmotic engine compartment from entering the drug reservoir.
  • the diffusion moderator is designed, in conjunction with the drug formulation, to prevent body fluid from entering the drug reservoir through the orifice.
  • the DUROS® device releases a therapeutic agent at a predetermined rate based on the principle of osmosis.
  • Extracellular fluid enters the DUROS® device through a semi-permeable membrane directly into a salt engine that expands to drive the piston at a slow and even delivery rate. Movement of the piston forces the drug formulation to be released through the orifice or exit port at a predetermined sheer rate.
  • the reservoir of the DUROS® device is loaded with a suspension formulation comprising, for example, GLP-1(7-36)amide or exenatide, wherein the device is capable of delivering the suspension formulation to a subject over an extended period of time (e.g., about one, about two, about three, about six, or about 12 months) at a predetermined, therapeutically effective delivery rate.
  • a suspension formulation comprising, for example, GLP-1(7-36)amide or exenatide
  • Implantable, drug delivery devices may be used in the practice of the present invention and may include regulator-type implantable pumps that provide constant flow, adjustable flow, or programmable flow of the compound, such as those available from Codman & Shurtleff, Inc. (Raynham, Mass.), Medtronic, Inc. (Minneapolis, Minn.), and Tricumed Medinzintechnik GmbH (Germany).
  • Implantable devices for example, the DUROS® device, provide the following advantages for administration of the formulations of the present invention: true zero-order release of the insulinotropic peptide pharmacokinetically; long-term release period time (e.g., up to about 12 months); and reliable delivery and dosing of the GLP-1 receptor agonist or other suitable beneficial agent.
  • FIG. 2 depicts a representative osmotic delivery device useful in the practice of the present invention.
  • an osmotic delivery device 10 is shown comprising a reservoir 12 .
  • a piston assembly 14 is positioned in the lumen of the reservoir and divides the lumen into two chambers.
  • the chamber 16 contains a beneficial agent formulation, such as a GLP-1 receptor agonist (e.g., GLP-1 (7-36)amide or exenatide) formulation, an anticancer agent, or the like and the chamber 20 contains an osmotic agent formulation.
  • a GLP-1 receptor agonist e.g., GLP-1 (7-36)amide or exenatide
  • the chamber 20 contains an osmotic agent formulation.
  • a semi-permeable membrane 18 is positioned at a distal end of the reservoir, adjacent the chamber 20 containing the osmotic agent formulation.
  • a diffusion moderator 22 is positioned in mating relationship at a distal end of the reservoir 12 , adjacent the chamber 16 containing the beneficial agent formulation.
  • the diffusion moderator 22 includes a delivery orifice 24 .
  • the diffusion moderator 22 may be any suitable flow device having a delivery orifice.
  • the flow path 26 is formed between a threaded diffusion moderator 22 and threads 28 formed on the interior surface of the reservoir 12 .
  • the diffusion moderator can, for example, (i) be press-fit (or friction fit) through an opening and contacting a smooth interior surface of the reservoir, or (ii) comprise two pieces with an outer shell constructed and arranged for positioning in an opening, an inner core inserted in the outer shell, and a fluid channel having a spiral shape defined between the outer shell and the inner core (e.g., U.S. Patent Publication No. 2007-0281024, incorporated herein by reference in its entirety).
  • Fluid is imbibed into the chamber 20 through the semi-permeable membrane 18 .
  • the beneficial agent formulation is dispensed from the chamber 16 through the delivery orifice 24 in the diffusion moderator 22 .
  • the piston assembly 14 engages and seals against the interior wall of the reservoir 12 , thereby isolating the osmotic agent formulation in chamber 20 and fluid imbibed through the semi-permeable membrane 18 from the beneficial agent formulation in chamber 16 .
  • the beneficial agent formulation is expelled through the delivery orifice 24 in the diffusion moderator 22 at a rate corresponding to the rate at which external fluid is imbibed into the chamber 20 through the semi-permeable membrane 18 .
  • the semi-permeable membrane 18 may be in the form of a plug that is resiliently engaged in sealing relationship with the interior surface of the reservoir 12 .
  • FIG. 2 it is shown to have ridges that serve to frictionally engage the semi-permeable membrane 18 with the interior surface of the reservoir 12 .
  • the amount of beneficial agent employed in the delivery device of the invention is that amount necessary to deliver a therapeutically effective amount of the agent to achieve the desired therapeutic result. In practice, this will vary depending upon such variables, for example, as the particular agent, the site of delivery, the severity of the condition, and the desired therapeutic effect.
  • the volume of a beneficial agent chamber comprising the beneficial agent formulation is between about 100 ⁇ l to about 1000 ⁇ l, more preferably between about 120 ⁇ l and about 500 ⁇ l, more preferably between about 150 ⁇ l and about 200 ⁇ l.
  • the osmotic delivery device is implanted within the subject, for example, subcutaneously.
  • the device(s) can be inserted in either or both arms (e.g., in the inside, outside, or back of the upper arm) or into the abdomen. Preferred locations in the abdomen are under the abdominal skin in the area extending below the ribs and above the belt line.
  • the abdominal wall can be divided into 4 quadrants as follows: the upper right quadrant extending 5-8 centimeters below the right ribs and about 5-8 centimeters to the right of the midline, the lower right quadrant extending 5-8 centimeters above the belt line and 5-8 centimeters to the right of the midline, the upper left quadrant extending 5-8 centimeters below the left ribs and about 5-8 centimeters to the left of the midline, and the lower left quadrant extending 5-8 centimeters above the belt line and 5-8 centimeters to the left of the midline.
  • This provides multiple available locations for implantation of one or more devices on one or more occasions.
  • the suspension formulation may also be delivered from a drug delivery device that is not implantable or implanted, for example, an external pump such as a peristaltic pump used for subcutaneous delivery in a hospital setting.
  • an external pump such as a peristaltic pump used for subcutaneous delivery in a hospital setting.
  • suspension formulations of the present invention may also be used in infusion pumps, for example, the ALZET® osmotic pumps which are miniature, infusion pumps for the continuous dosing of laboratory animals (e.g., mice and rats).
  • ALZET® osmotic pumps which are miniature, infusion pumps for the continuous dosing of laboratory animals (e.g., mice and rats).
  • suspension formulations of the present invention may also be used in the form of injections to provide highly concentrated bolus doses of biologically active agents, such as the GLP-1 receptor agonists, anti-cancer agents, etc.
  • the GLP-1 receptor agonists such as GLP-1(7-36)amide and exenatide, can be delivered to a patient as a single modality treatment or in combination with other beneficial agents, including anticancer agents as described below, chemotherapeutic drugs, anticancer antibodies, antisense molecules, siRNA, and the like.
  • one useful combination is with a tyrosine kinase inhibitor, such as SUTENT®, NEXAVAR®, BIBF 1120, ZD1839 (gefitinib), erlotinib, TYKERBTM, and the like.
  • a tyrosine kinase inhibitor such as SUTENT®, NEXAVAR®, BIBF 1120, ZD1839 (gefitinib), erlotinib, TYKERBTM, and the like.
  • mTOR inhibitors such as rapamycin (sirolimus), AZD8055, NVP-BEZ235, deforolimus, everolimus, temsirolimus, GSK1059615, WYE354, KU0063794, XL765 (all available from Selleck Chemicals) will also find use in a combination treatment.
  • GLP-1 receptor agonists e.g., exenatide and GLP-1(7-36)amide
  • drugs that cause hypoxia in tumor tissues such as metformin
  • drugs that inhibit the hypoxia inducible factor 1 such as CCAA/enhancer binding protein a, PX-478, resveratrol, and the various small molecule inhibitors described in Jones et al., Mol. Cancer. Ther . (2006) 5:2193-2202.
  • drugs that inhibit IGF-1 such as octreonide acetate and tyrosine kinase inhibitors, that serve to block IGF-1 receptor signaling.
  • VEGF-inhibitors such as anti-VEGF antibodies including bevacizumab) (AVASTIN®, as well as prolactin, sunitinib and sorafenib, may also be used in combination with the GLP-1 receptor agonists.
  • anti-VEGF antibodies including bevacizumab) (AVASTIN®, as well as prolactin, sunitinib and sorafenib, may also be used in combination with the GLP-1 receptor agonists.
  • Another useful combination therapy is the use of a sugar analog, such as 2DG, subsequent to reducing glucose availability to the cancer cells using GLP-1 receptor agonists, such as exenatide and GLP-1(7-36)amide.
  • a sugar analog such as 2DG
  • GLP-1 receptor agonists such as exenatide and GLP-1(7-36)amide.
  • Cell cycle blockers will also find use herein, such as a cyclin-dependent kinase (cdk)-inhibitor, e.g., olomoucin, butyrolactone-I, n-butyrate, upregulators of cdk activity, e.g., flavopiridol, Chalcones (1,3-diphenylpropen-1-ones) and derivatives thereof.
  • cdk cyclin-dependent kinase
  • HDAC histone deacetylase
  • peptides that induce cell apoptosis such as TRAIL, antagonists or antibodies against integrin ⁇ v ⁇ 3 , anti-survivin antibodies and antagonists of survivin, and numerous pro-apoptotic peptides, well known in the art, such as described in Ellerby et al., Nat. Med. (1999) 5:1032-1038.
  • chemokines which can be administered include BCA-1/BLC, BRAK, Chemokine CC-2, CTACK, CXCL-16, ELC, ENA, ENA-70, ENA-74, ENA-78, Eotaxin, Exodus-2, Fractalkine, GCP-2, GRO, GRO alpha (MGSA), GRO-beta, GRO-gamma, HCC-1, HCC-4, 1-309, IP-10, 1-TAC, LAG-1, LD78-beta, LEC/NCC-4, LL-37, Lymphotactin, MCP, MCAF (MCP-1), MCP-2, MCP-3, MCP-4, MDC, MDC, MDC-2, MDC-4, MEC/CCL28, MIG, MIP, MIP-1 alpha, MIP-1 beta, MIP-1 delta, MIP-3/MPIF-1, MIP-3 alpha, MIP-3 bet, MIP-4 (PARC), MIP-5, NAP-2, PARC
  • growth factors which can be delivered include Human Amphiregulin, Human Angiogenesis Proteins, Human ACE, Human Angiogenin, Human Angiopoietin, Human Angiostatin, Human Endostatin, Human Betacellulin, Human BMP, Human BMP-13/CDMP-2, Human BMP-14/CDMP-1, Human BMP-2, Human BMP-3, Human BMP-4, Human BMP-5, Human BMP-6, Human BMP-7, Human BMP-8, Human BMP-9, Human Colony Stimulating Factors, Human flt3-Ligand, Human G-CSF, Human GM-CSF, Human M-CSF, Human Connective Tissue Growth Factor, Human Cripto-1, Human Cryptic, Human ECGF, Human EGF, Human EG-VEGF, Human Erythropoietin, Human Fetuin, Human FGF, Human FGF-1, Human FGF-10, Human FGF-16, Human FGF-17, Human FGF-18, Human FGF-19, Human F
  • chemotherapeutic agents used in the methods of the invention are selected from antimetabolites; enzyme inhibitors including topoisomerase I and II inhibitors, tyrosine and serine/threonine kinase inhibitors and COX2 inhibitors, tubulin binders, proteasome inhibitors, anticancer alkylating agents including bifunctional and monofunctional alkylating agents and methylating agents, anticancer antibiotics, anticancer antibodies and active fragments and fusions thereof and antibody-drug conjugates, bisphosphonates, antiestrogens and antiandrogens, anticancer cytokines, anticancer enzymes, immunomodulatory agents, anticancer peptides, anticancer retinoids, anticancer steroids and related agents, anticancer phototherapeutics, normal tissue protectors and antihormonal agents including aromatase inhibitors.
  • Antimetabolites may include folate analogs, which inhibit dihydrofolate reductase resulting in DNA breaks by blocking purine and thymidylate synthesis.
  • folate analogs include methotrexate (FOLEXTM), trimetrexate (NEUTREXIN®) and pemetrexed (ALIMTA®).
  • Other anitmetabolites are nucleoside analogs that disrupt DNA or RNA synthesis, such as purine or pyrimidine analogs.
  • purine analogs include allopurinol (ZYLOPRIM®), mercaptopurine (PURINETHOL®), fludarabine (FLUDARATM), thioguanine (6-TG), cladribine (LEUSTATIN®, 2-CdA), and pentostatin (NIPENT®).
  • pyrimidine analogs include capecitabine (XELODA®), cytarabine (CYTOSARTM), liposomal cytarabine (DEPOCYT®), floxuridine (FUDRTM), fluororouracil (ADRUCIL®), gemcitabine (GEMZAR®), and clofarabine (CLOLAR®), decitabine (DACOGEN®) and azacitadine (VIDAZA®).
  • Topoisomerase II inhibitors bind to topoisomerase II and DNA, preventing the resealing of DNA strands during replication, and leading to DNA strand breaks, such as epipodophyllotoxins.
  • epipodophyllotoxins include etoposide (VEPESID®, ETOPOPHOS®) and teniposide (VUMON®, VM26TM).
  • topoisomerase II inhibitors such as anthracycline antibiotics, intercalate between DNA base pairs leading to free radicals and also topoisomerase II inhibition.
  • anthracyclines examples include daunorubicin (DANOIJXOME®, CERUBIDINETM), liposomal daunorubicin (DAUNOXOME®), doxorubicin (ADRIAMYCINTM, RUBEXTM), liposomal doxorubicin (DOXILTM), epirubicin (ELLENCETM), valrubicin (VALSTAR®), and idarubicin (IDAMYCINTM). Mitoxantrone (NOVANTRONE®) also inhibits topoisomerase II and is an anticancer therapeutic.
  • Topoisomerase I inhibitors bind to topoisomerase I and DNA, preventing DNA strand breaks, such as, e.g., camptothecins, including irinotecan (CAMPTOSAR®) and topotecan (HYCAMTIN®).
  • camptothecins including irinotecan (CAMPTOSAR®) and topotecan (HYCAMTIN®).
  • Anticancer kinase inhibitors inhibit phosphorylation of a protein or small molecule messenger in a an intracellular signaling pathway in malignant cells or vascular or stromal cells, such as, e.g., imatinib mseylate (GLEEVEC®), gefitinib (IRESSA®) or erlotinib (TARCEVA®), sorafenib (NEXAVAR®), sunitinib (SUTENT®), nilotinib (TASIGN®), everolimus (AFINITOR®), lapatinib (TYKERB®), dasatinib (SPRYCEL®), BRAF inhibitors such as GSK218436 (GlaxoSmithKline, London UK) and vemurafenib (Plexxikon Inc., CA) and MEK inhibitors.
  • GLEEVEC® imatinib mseylate
  • IRESSA® gefitinib
  • TARCEVA® so
  • Tubulin binders include agents that bind to microtubules, shift the microtubules toward polymerization, and are active in the M phase, such as taxanes including docetaxel (TAXOTERE®) and paclitaxel (TAXOL®) and epothilones including ixabepilone (IXEMPRA®) and eribulin mesylate.
  • Other tubulin binders act by inhibiting polymerization and mitotic spindle formation, and are active in the S phase, such as, e.g., vinca alkaloids, including vinblastine (VELBAN®), vincristine (ONCOVINTM), and vinorelbine (NAVELBINE®).
  • Other tubulin binders include ILX-651 (TASIDOTINTM) and estramustine (EMCYT®), which inhibit microtubule assembly and disassembly.
  • proteasome inhibitors block the trypsin-like, chymotrypsin-like and/or peptidylglutamyl peptide hydrolyzing-like protease activities in nuclear and cytoplasmic proteasomes.
  • proteasome inhibitors include bortezomib (VELCADE®).
  • Anticancer alkylating agents are reactive molecules that bind to DNA and interfere with DNA replication. These agents include, but are not limited to, alkyl sulfonates such as busulfan (MYLERAN®), platinum analogs such as carboplatin (PARAPLATIN®), cisplatin (PLATINOL®-AQ, and oxaliplatin (ELOXATIN®), nitrosoureas such as carmustine (BICNU®), lomustine (CCNUTM, CEENU®), and streptozocin (ZANOSAle), nitrogen mustards including chlorambucil (LEUKERAN®), uracil mustard, cyclophosphamide (CYTOXAN®), ifosfamide (IFEX®), meclorethamine (MUSTARGEN®), and melphalan (ALKERAN®, L-PAM), bendamustine (TREANDA®), triazenes such as dacarbazine (DTIC-DOME®), procarbazine
  • Anticancer antibiotics act by a variety of mechanisms including inhibition of protein synthesis generation of oxygen free radicals in the vicinity of DNA and other mechanisms.
  • Examples of anticancer antibiotics include actinomycin D (COSMEGEN®), bleomycin sulfate (BLENOXANE®) and plicamycin (MITHRACINTM).
  • Anticancer antibodies bind to specific molecular targets on cells or in the extracellular space. Anticancer antibodies act by neutralizing the activity of the target, attracting immune cells to the target cell or by being directly or indirectly cytotoxic toward the target cell. Anticancer antibodies include, but are not limited to, anti-CD52 antibodies such as alemtuzumab (CAMPATH®); anti-VEGF antibodies including bevacizumab (AVASTIN®); anti-CD33 antibodies, including gemtuzumab ozogamicin (MYLOTARG®); anti-CD20 antibodies including ibritumomab (ZEVALINTM), rituximab (RITUXANTM), tositumomab (BEXXAR®) and ofatumumab (ARZERRA®); anti-EGFR antibodies such as cetuximab (ERBITUX®) and panitumumab (VECTIBEX®); anti-Her2 antibodies, including trastuzumab (HERCEPTIN®); anti-CTLA
  • Anticancer cytokines include, but are not limited to, aldesleukin (PROLEUKIN®), denileukin diftitox (ONTAK®), GM-CSF (sargramostim, PROKINETM, LEUKINETM), interferon alfa-2b (INTRON®-A), PEGinterferon alpha (PEGASYS® or PEGINTRON®) and consensus interferon (INFERGEN®).
  • Immunomodulatory agents are effective by increasing the response of the immune system of the host to the malignancy.
  • Immunomodulatory agents include, but are not limited to, Bacillus Calmette-Gurerin (BCG Vaccine), levamisole (ERGAMISOLTM), thalidomide (THALIDOMID®), sipuleucel-T (PROVENGE®), and lenalidomide (REVLIMID®).
  • Anticancer retinoids include, but are not limited to, aliretinoin (PANRETIN®), bexarotene (TARGRETIN®) and tretinoin (VESANOID®, ATRATM); other agents include octreotide acetate (SANDOSTATIN®).
  • Anticancer enzymes include asparaginase (ELSPAR®), pegademase (ADAGEN®), and pegaspargase (ONCASPAR®).
  • Anticancer steroids and related agents include dexamethasone (DECADRONTM), predisone (DELTASONE®), prednisolone (DELTA-CORTEFTM) and mitotane (LYSODREN®).
  • Normal tissue protectors include, but are not limited to, amifostine (ETHYOL®), darbepoetin alfa (ARANESP®), dexrazoxane (ZINECARD®), epoetin alfa (EPOGEN®, PROCRIT®), filgrastim (NEUPOGEN®), folinic acid (leucovorin), allopurinol (ALOPRIM®) mesna (MESNEX®), oprelvekin (NEUMEGA®), pegfilgrastim (NEULASTA®), GM-CSF (sargramostim, PROKINETM, LEUKINE®), raloxifene (EVISTA®) and eltrombopag (PROMACTA®).
  • ETHYOL® amifostine
  • ARANESP® darbepoetin alfa
  • ZINECARD® dexrazoxane
  • EPOGEN® epoetin alfa
  • Phototherapeutics are agents that sensitize cells so that exposure to a specific frequency of laser light induces abundant free radical formation and DNA alkylation. These agents include, but are not limited to, porfimer sodium (PHOTOFRIN®).
  • Antihormones include LHRH agonists, which compete with gonadotropin by binding to the hypothalamus causing an initial surge of LH and FSH followed by down regulation by negative feedback, including goserelin (ZOLADEX®), leuprolide (LUPRON® or ELIGARD®), and triptorelin (TRELSTAR®); and antiandrogens, which competitively bind and inhibit the binding of androgens to androgen receptors, such as hicalutamide (CASODEX®), flutamide (EULEXINTM), nilutamide (NILANDRON®), aminoglutethimide (CYTADREN®), and abarelix (PLENAXIS®); and antiestrogens, which competitively bind and inhibit the binding of estrogens to estrogen receptors such as tamoxifen (NOLVADEX®), fluoxymesterone (HALOTESTIN®) and megestrol (MEGACE®), bisphosphonates including pamidronate (AREDIA®) and
  • ATP-competitive inhibitors of c-Met/HGF receptor and/or the nucleophosmin-anaplastic lymphoma kinase include crizotinib, CH5424802 (Chugai Pharmaceutical Co., Ltd., Japan), and AP26113 (ARIAD Pharmaceuticals, Inc., MA).
  • agents including beneficial agents and anticancer agents that can be delivered with the GLP-1 receptor agonist compositions described herein include those described above and/or shown in Table 1.
  • Tests can be performed prior to treatment to specifically tailor a treatment for a patient. Such tests may include genetic or protein marker testing of tumor markers to determine susceptibility or resistance to a particular drug or class of drugs. For example, recently a mutation in von Hippel-Landau (VHL) gene have been found to be associated with a more favorable drug response for drugs such as SUTENT®, NEXAVAR®, and AVASTIN®. Other genetic and protein tests can be performed to link a treatment to an appropriate patient population.
  • VHL von Hippel-Landau
  • the agents described above can be provided in formulations obtained from the manufacturer.
  • Such formulations typically include the active components mixed with a pharmaceutically acceptable vehicle or excipient.
  • the vehicle may contain minor amounts of auxiliary substances such as wetting or emulsifying agents.
  • the formulations may also include ancillary substances, such as pharmacological agents, cytokines, or other biological response modifiers.
  • the pharmaceutical composition comprising the agent is a sustained-release formulation, and/or a formulation that is administered using a sustained-release device.
  • sustained-release devices include, for example, transdermal patches, and miniature implantable pumps (such as described herein) that can provide for drug delivery over time in a continuous, steady-state fashion at a variety of doses to achieve a sustained-release effect with either a non-sustained-release or a sustained release pharmaceutical composition.
  • polypeptide agents and antibodies described herein are suitable agents for delivery using an osmotic delivery device such as the DUROS® implantable device described above.
  • two or more such implantable delivery devices can be used, one including the GLP-1 receptor agonist and one or more including one or more additional beneficial agents, such as anticancer polypeptide formulations, antibodies, and the like. See, e.g., U.S. Patent Publication 2009/0202608, incorporated herein by reference in its entirety, for a description of the use of two or more implantable delivery devices.
  • the additional beneficial agents may also be formulated as particle and suspension formulations as described herein, if appropriate.
  • particle and suspension formulations are useful with polypeptide agents and antibodies and can be delivered using implantable devices as described above.
  • some polypeptide agents e.g., leuprolide acetate
  • some polypeptide agents can be directly dissolved or dispersed in a vehicle for delivery from implantable devices.
  • some polypeptides e.g., leuprolide acetate
  • non-aqueous polar aprotic solvents e.g., dimethylsulfoxide
  • peptide formulations include, but are not limited to, non-aqueous protic peptide formulations (see, e.g., U.S. Pat. No. 6,066,619, incorporated herein by reference in its entirety) and aqueous formulations of peptides (see, e.g., U.S. Pat. No. 6,068,850, incorporated herein by reference in its entirety).
  • compositions for example s.c., intraperitoneal (i.p.), intramuscular (i.m.), intravenous (i.v.), or infusion, oral (p.o.) and pulmonary, nasal, topical, transdermal, and suppositories.
  • parenteral administration such as subcutaneous (s.c.), intraperitoneal (i.p.), intramuscular (i.m.), intravenous (i.v.), or infusion, oral (p.o.) and pulmonary, nasal, topical, transdermal, and suppositories.
  • the therapeutically effective dose is adjusted such that the soluble level of the agent in the bloodstream, is equivalent to that obtained with a therapeutically effective dose that is administered parenterally, for example s.c., i.p., i.m., or i.v.
  • the pharmaceutical composition comprising the beneficial agent is administered by i.m. or s.c. injection, particularly by i.m. or s.c. injection
  • One or more therapeutically effective dose of the additional beneficial agent such as an anticancer agent will be administered.
  • therapeutically effective dose or amount of each of these agents is intended an amount that when administered in combination with the other agents, brings about a positive therapeutic response with respect to treatment of an individual with cancer. Of particular interest is an amount of these agents that provides an anti-tumor effect, as defined herein. In certain embodiments, multiple therapeutically effective doses of the additional beneficial agent will be provided.
  • the additional beneficial agents can be administered prior to, concurrent with, or subsequent to administration of the GLP-1 receptor agonist.
  • initial treatment with a chemotherapeutic agent can be performed, followed by implantation of a delivery device including the GLP-1 receptor agonist formulation or vice versa.
  • the additional beneficial agent may be administered over the time that the GLP-1 receptor agonist formulation is also being delivered.
  • concurrent therapy is intended administration to a subject such that the therapeutic effect of the combination of the substances is caused in the subject undergoing therapy.
  • the GLP-1 receptor agonists e.g., exenatide and GLP-1(7-36)amide, optionally in combination with other beneficial agents, can be used to treat various cancers.
  • cancer cells are known to exhibit increased glycolysis as compared to normal cells.
  • An advantage of the present invention is that inhibiting glucose availability to cancer cells by using a GLP-1 receptor agonist, such as exenatide and GLP-1(7-36)amide, effectively reduces the amount of energy metabolites such as ATP and NADH produced, thereby starving the cancer cell of energy.
  • tumors or cancers such as hemangiomas, neufibromatosis, breast, colorectal, lung, brain and CNS, renal, gynecological (e.g.
  • Table 2 A list of cancers that may benefit from delivery of the GLP-1 receptor agonists is shown in Table 2.
  • Acute Lymphoblastic Leukemia Adult Acute Lymphoblastic Leukemia, Childhood Acute Myeloid Leukemia, Adult Acute Myeloid Leukemia, Childhood Adrenocortical Carcinoma Adrenocortical Carcinoma, Childhood AIDS-Related Cancers AIDS-Related Lymphoma
  • Anal Cancer Appendix Cancer Atypical Teratoid/Rhabdoid Tumor, Childhood, Central Nervous System Basal Cell Carcinoma, see Skin Cancer (Non-melanoma) Bladder Cancer Bladder Cancer, Childhood Bone Cancer, Osteosarcoma and Malignant Fibrous Histiocytoma Brain Stem Glioma, Childhood Brain Tumor, Adult Brain Tumor, Brain Stem Glioma, Childhood Brain Tumor, Central Nervous System Atypical Teratoid/Rhabdoid Tumor, Childhood Brain Tumor, Central Nervous System Embryonal Tumors, Childhood Brain Tumor, Cerebellar Astrocytoma, Childhood
  • the GLP-1 receptor agonists are used in the treatment of hematological tumors and/or solid tumors.
  • the GLP-1 receptor agonists for example, exenatide and GLP-1(7-36)amide, are used in the treatment of solid tumors.
  • the GLP-1 receptor agonists are delivered in order to provide a positive therapeutic response.
  • positive therapeutic response it is intended the individual undergoing the combination treatment of a GLP-1 receptor agonist, such as exenatide and GLP-1(7-36)amide, and an additional beneficial agent exhibits an improvement in one or more symptoms of the cancer for which the individual is undergoing therapy.
  • a positive therapeutic response refers to one or more of the following improvements in the disease: (1) reduction in tumor size; (2) reduction in the number of cancer cells; (3) inhibition (i.e., slowing to some extent, preferably halting) of tumor growth; (4) inhibition (i.e., slowing to some extent, preferably halting) of cancer cell infiltration into peripheral organs; (5) inhibition (i.e., slowing to some extent, preferably halting) of tumor metastasis; and (6) some extent of relief from one or more symptoms associated with the cancer.
  • Such therapeutic responses may be further characterized as to degree of improvement.
  • an improvement may be characterized as a complete response.
  • partial response is intended a reduction of greater than 50% in the sum of the products of the perpendicular diameters of one or more measurable lesions when compared with pretreatment measurements (for patients with evaluable response only, partial response does not apply).
  • the GLP-1 receptor agonist is delivered in a suspension formulation, administered using an osmotic delivery device as described above.
  • target rates of delivery for suspension formulations of the present invention include, but are not limited to: suspension formulations comprising particle formulations comprising GLP-1 (e.g., GLP-1(7-36)amide), between about 20 ⁇ g/day and about 900 ⁇ g/day, preferably between about 100 ⁇ g/day and about 600 ⁇ g/day, for example, at about 480 ⁇ g/day; and suspension formulations comprising particle formulations comprising exenatide, between about 5 ⁇ g/day and about 320 ⁇ g/day, preferably between about 5 ⁇ g/day and about 160 ⁇ g/day, for example, at about 10 ⁇ g/day to about 20 ⁇ g/day, such as 10, 20, 40, 60, 80, 100, 120 ⁇ g/day, or any integers between the above ranges.
  • GLP-1 e.g., GLP-1(7-36)amide
  • An exit sheer rate of the suspension formulation from the osmotic delivery device is determined such that the target daily target delivery rate of the GLP-1 receptor agonist is reasonably achieved by substantially continuous, uniform delivery of the suspension formulation from the osmotic delivery device.
  • exit sheer rates include, but are not limited to, about 1 to about 1 ⁇ 10 4 reciprocal second, preferably about 4 ⁇ 10 ⁇ 2 to about 6 ⁇ 10 4 reciprocal second, more preferably 5 ⁇ 10 ⁇ 3 to 1 ⁇ 10 ⁇ 3 reciprocal second.
  • a subject being treated with the GLP-1 receptor agonist formulations of the present invention may also benefit from co-treatment with other beneficial agents, including anticancer agents described above, as well as antidiabetic agents.
  • Additional beneficial agents that can be delivered include, but are not limited to, pharmacologically beneficial peptides proteins, polypeptides, genes, gene products, other gene therapy agents, or other small molecules.
  • the additional beneficial agents are useful for the treatment of a variety of conditions including but not limited to hemophilia and other blood disorders, growth disorders, diabetes, leukemia and lymphoma, hepatitis, renal failure, bacterial infection, viral infection (e.g., infection by HIV, HCV, etc.), hereditary diseases such as cerbrosidase deficiency and adenosine deaminase deficiency, hypertension, septic shock, autoimmune diseases (e.g., Graves disease, systemic lupus erythematosus and rheumatoid arthritis), shock and wasting disorders, cystic fibrosis, lactose intolerance, Crohn's disease, inflammatory bowel disease, Alzheimer's disease, metabolic disorders (such as obesity), and cancers.
  • the polypeptides may include but are not limited to the following: glucagon-like peptide 2 (GLP-2), cholecystokinin (CCK), CCK octapeptide, growth hormone, somatostatin; somatropin, somatotropin, somatotropin analogs, somatomedin-C, somatotropin plus an amino acid, somatotropin plus a protein; follicle stimulating hormone; luteinizing hormone, luteinizing hormone-releasing hormone (LHRH), LHRH analogs/agonists such as leuprolide, nafarelin and goserelin, LHRH antagonists; growth hormone releasing factor; calcitonin; colchicine; gonadotropins such as chorionic gonadotropin; antiandrogens such as flutamide, nilutamide and cytoprerone; aromatase inhibitors such as exemastane, letrozole and anastrazole; selective
  • beneficial agents include but are not limited to the following: alpha antitrypsin; factor VII; factor IX, thrombin and other coagulation factors; insulin; peptide hormones; adrenal cortical stimulating hormone, thyroid stimulating hormone and other pituitary hormones; erythropoietin; growth factors such as granulocyte-colony stimulating factor, granulocyte-macrophage colony stimulating factor, thrombopoietin, insulin-like growth factor 1; tissue plasminogen activator; CD4; 1-deamino-8-D-arginine vasopressin; interleukin-1 receptor antagonist; tumor necrosis factor, tumor necrosis factor receptor; tumor suppresser proteins; pancreatic enzymes; lactase; cytokines, including lymphokines, chemokines or interleukins such as interleukin-1, interleukin-2 and other members of the interleukin family (e.g., IL-1, 6, 12, 15, 17, 18, 32); cytotoxic
  • beneficial agents include, but are not limited to, organic compounds including those compounds that transport across a vessel.
  • beneficial agents include, but are not limited to, the following: hypnotics and sedatives such as pentobarbital sodium, phenobarbital, secobarbital, thiopental, amides and ureas exemplified by diethylisovaleramide and alpha-bromo-isovaleryl urea, urethanes, or disulfanes; heterocyclic hypnotics such as dioxopiperidines, and glutarimides; antidepressants such as isocarboxazid, nialamide, phenelzine, imipramine, tranylcypromine, pargyline; tranquilizers such as chloropromazine, promazine, fluphenazine reserpine, deserpidine, meprobamate, benzodiazepines such
  • peptides, proteins, or polypeptides that are useful in the practice of the present invention are described herein.
  • modifications of these peptides, proteins, or polypeptides are also known to one of skill in the art and can be used in the practice of the present invention following the guidance presented herein. Such modifications include, but are not limited to, amino acid analogs, amino acid mimetics, analog polypeptides, or derivative polypeptides.
  • the beneficial agents disclosed herein may be formulated singly or in combination (e.g., mixtures).
  • Peptide YY inhibits gut motility and blood flow (Laburthe, M., Trends Endocrinol Metab. 1(3):168-74 (1990), mediates intestinal secretion (Cox, H. M., et al., Br J Pharmacol 101(2):247-52 (1990); Playford, R. J., et al., Lancet 335(8705):1555-7 (1990)), stimulate net absorption (MacFayden, R. J., et al., Neuropeptides 7(3):219-27 (1986)), and two major in vivo variants (PYY and PYY 3-36 ) have been identified (e.g., Eberlein, G.
  • PYY polypeptides, PYY derivatives, variants and analogs are referred to collectively as PYY.
  • GIP is an insulinotropic peptide hormone (Efendic, S., Horm Metab Res . (2004) 36:742-746) and is secreted by the mucosa of the duodenum and jejunum in response to absorbed fat and carbohydrate that stimulate the pancreas to secrete insulin. GIP stimulates insulin secretion from pancreatic beta cells in the presence of glucose (Tseng et al., PATAS (1993) 90:1992-1996). GIP circulates as a biologically active 42-amino acid peptide. GIP is also known as glucose-dependent insulinotropic protein. The sequence of GIP, as well as peptide analogs and peptide derivatives thereof, are known in the art (see, e.g., Meier J.
  • GIP GIP polypeptides, GIP derivatives, variants and analogs are referred to collectively as GIP.
  • Oxyntomodulin is a naturally occurring 37 amino acid peptide hormone found in the colon that has been found to suppress appetite and facilitate weight loss (Wynne K, et al., Int J Obes (Lond) 30(12):1729-36 (2006)).
  • the sequence of oxyntomodulin, as well as analogs and derivatives thereof, are known in the art (e.g., U.S. Patent Publication Nos. 2005-0070469 and 2006-0094652).
  • the family of oxyntomodulin polypeptides, oxyntomodulin derivatives, variants and analogs are referred to collectively as oxyntomodulin.
  • Amylin as well as analogs and derivatives thereof: are known in the art (e.g., U.S. Pat. Nos. 5,686,411, 5,814,600, 5,998,367, 6,114,304, 6,410,511, 6,608,029, and 6,610,824).
  • the family of amylin polypeptides, amylin derivatives, variants and analogs are referred to collectively as amylin.
  • the cDNA sequence encoding the human leptin protein hormone is known (e.g., Masuzaki, H., et al. (Diabetes 44: 855-858, 1995)).
  • Leptin, as well as analogs and derivatives thereof, are known in the art (e.g., U.S. Pat. Nos.
  • RNA molecules may include, but are not limited to, small nuclear RNAs (snRNAs), and small interfering RNA strands (siRNA) for use in RNA interference (RNAi) inhibition of gene expression.
  • snRNAs small nuclear RNAs
  • siRNA small interfering RNA strands
  • RNAi inhibition typically occurs at the stage of translation or by hindering the transcription of specific genes.
  • RNAi targets include, but are not limited to, RNA from viruses and genes with roles in regulating development and genome maintenance.
  • the beneficial agents can also be in various forms including, but not limited to, the following: uncharged molecules; components of molecular complexes; and pharmacologically acceptable salts such as hydrochloride, hydrobromide, sulfate, laurates, palmatates, phosphate, nitrate, borate, acetate, maleate, tartrate, oleates, or salicylates.
  • pharmacologically acceptable salts such as hydrochloride, hydrobromide, sulfate, laurates, palmatates, phosphate, nitrate, borate, acetate, maleate, tartrate, oleates, or salicylates.
  • salts of metals, amines or organic cations for example, quaternary ammonium
  • simple derivatives of the drug such as esters, ethers, amides and the like that have solubility characteristics suitable for the purpose of the invention can also be used herein.
  • the formulation used can have been in various art known forms such as solution, dispersion, paste, cream, particle, granule, tablet, emulsions, suspensions, powders and the like.
  • the beneficial agent formulation may optionally include pharmaceutically acceptable carriers and/or additional ingredients such as antioxidants, stabilizing agents, buffers, and permeation enhancers.
  • the amount of beneficial agent used is that amount necessary to deliver a therapeutically effective amount of the agent to achieve the desired therapeutic result. In practice, this will vary depending upon such variables, for example, as the particular agent, the site of delivery, the severity of the condition, and the desired therapeutic effect.
  • Beneficial agents and their dosage unit amounts are known to the prior art in Goodman & Gilman's The Pharmacological Basis of Therapeutics, 11th Ed., (2005), McGraw Hill; Remington's Pharmaceutical Sciences, 18th Ed., (1995), Mack Publishing Co.; and Martin's Physical Pharmacy and Pharmaceutical Sciences, 1.00 edition (2005), Lippincott Williams & Wilkins.
  • the additional beneficial agent can be delivered using any of the various delivery techniques outlined above, including without limitation parenterally (including by subcutaneous, intravenous, intramedullary, intraarticular, intramuscular, or intraperitoneal injection) rectally, topically, transdermally, intranasally, by inhalation, or orally (for example, in capsules, suspensions, or tablets).
  • parenterally including by subcutaneous, intravenous, intramedullary, intraarticular, intramuscular, or intraperitoneal injection
  • rectally including by subcutaneous, intravenous, intramedullary, intraarticular, intramuscular, or intraperitoneal injection
  • rectally including by subcutaneous, intravenous, intramedullary, intraarticular, intramuscular, or intraperitoneal injection
  • rectally rectally, topically, transdermally, intranasally, by inhalation, or orally (for example, in capsules, suspensions, or tablets).
  • the agent is in a sustained-release formulation, or
  • Such devices are well known in the art, and include, for example, transdermal patches, and miniature implantable pumps (such as the DUROS® delivery device described herein) that can provide for drug delivery over time in a continuous, steady-state fashion at a variety of doses to achieve a sustained-release effect with a non-sustained-release pharmaceutical composition.
  • the volume of a beneficial agent chamber comprising the beneficial agent formulation is between about 50 ⁇ l to about 1000 ⁇ l, more preferably between about 100 ⁇ l and about 500 ⁇ l, more preferably between about 150 ⁇ l and about 200 ⁇ l.
  • two or more such devices can be used, one including the GLP-1 receptor agonist and one or more including one or more additional beneficial agents, such as an antidiabetic compound. See, e.g., U.S. Patent Publication 2009/0202608, incorporated herein by reference in its entirety, for a description of the use of two or more implantable delivery devices.
  • Example 5 An example of a cancer treatment using delivery of an anticancer agent from a first osmotic delivery device and delivery of a GLP-1 receptor agonist from a second osmotic delivery device is presented below in Example 5.
  • the cancer is prostate cancer
  • the anticancer agent is leuprolide acetate
  • the GLP-1 receptor agonist is exenatide.
  • compositions produced according to the present invention meet the specifications for content and purity required of pharmaceutical products.
  • This example describes making exenatide particle formulations.
  • Exenatide (0.25 g) was dissolved in 50 mM sodium citrate buffer at pH 6.04. The solution was dialyzed with a formulation solution containing sodium citrate buffer, sucrose, and methionine. The formulated solution was then spray dried using Buchi 290 with 0.7 mm nozzle, outlet temperature of 75° C., atomization pressure of 100 Psi, solid content of 2%, and flow rate of 2.8 mL/min. The dry powder contained 21.5% of exenatide with 4.7% residual moisture and 0.228 g/ml density.
  • GLP-1(7-36)amide This example describes making a GLP-1(7-36)amide particle formulation.
  • GLP-1(7-36)amide (1.5 g) was dissolved in 5 mM sodium citrate buffer at pH 4. The solution was dialyzed with a formulation solution containing sodium citrate buffer and methionine. The formulated solution was then spray dried using Buchi 290 with 0.7 mm nozzle, outlet temperature of 70° C., atomization pressure of 100 Psi, solid content of 1.5%, and flow rate of 5 mL/min. The dry powder contained 90% of GLP-1(7-36)amide.
  • This example describes making suspension formulations comprising a suspension vehicle and an exenatide particle formulation.
  • exenatide particle formulation was generated by spray-drying, and contained 20 wt % exenatide, 32 wt % sucrose, 16 wt % methionine and 32 wt % citrate buffer.
  • a suspension vehicle was formed by dissolving the polymer polyvinylpyrrolidone in the solvent benzyl benzoate at approximately a 50/50 ratio by weight.
  • the vehicle viscosity was approximately 12,000 to 18,000 poise when measured at 33° C.
  • Particles containing the peptide exenatide were dispersed throughout the vehicle at a concentration of 10% particles by weight.
  • a suspension vehicle was formed by dissolving the polymer polyvinylpyrrolidone K-17 (typically having an approximate average molecular weight range of 7,900-10,800) in the solvent benzyl benzoate heated to approximately 65° C. under a dry atmosphere and reduced pressure at approximately a 50/50 ratio by weight.
  • the vehicle viscosity was approximately 12,000 to 18,000 poise when measured at 33° C.
  • Particle formulations 1-3, described in Example 1 were dispersed throughout the vehicle at the concentrations (by weight percent) shown in Table 4.
  • This example describes making a suspension formulation comprising a suspension vehicle and an GLP-1(7-36)amide particle formulation.
  • a GLP-1(7-36)amide particle formulation was generated by spray-drying, and contained 90 wt % GLP-1, 5 wt % methionine and 5 wt % citrate buffer.
  • a suspension vehicle containing the polymer polyvinylpyrrolidone was dissolved in the solvent benzyl benzoate at approximately a 50/50 ratio by weight.
  • the vehicle viscosity was approximately 12,000 to 18,000 poise when measured at 33° C.
  • Particles containing the peptide GLP-1(7-36)amide were dispersed throughout the vehicle at a concentration of 33% particles by weight.
  • Leuprolide acetate acts as a potent inhibitor of gonadotropin secretion when given continuously and in therapeutic doses.
  • Animal and human studies indicate that following an initial stimulation, chronic administration of leuprolide acetate results in suppression of testicular steroidogenesis. This effect is reversible upon discontinuation of drug therapy.
  • Administration of leuprolide acetate has resulted in inhibition of the growth of certain hormone-dependent tumors (prostatic tumors in Noble and Dunning male rats and DMBA-induced mammary tumors in female rats) as well as atrophy of the reproductive organs.
  • leuprolide acetate In humans, administration of leuprolide acetate results in an initial increase in circulating levels of luteinizing hormone (LH) and follicle stimulating hormone (FSH), leading to a transient increase in levels of the gonadal steroids (testosterone and dihydrotestosterone in males).
  • LH luteinizing hormone
  • FSH follicle stimulating hormone
  • continuous administration of leuprolide acetate results in decreased level of LH and FSH.
  • testosterone is reduced to castrate levels. These decreases occur within two to six weeks after initiation of treatment, and castrate levels of testosterone in prostatic cancer patients have been demonstrated for multiyear periods.
  • Leuprolide acetate is not active when given orally.
  • An implantable device containing leuprolide acetate for the treatment of prostate cancer is assembled as described in U.S. Pat. No. 5,728,396, incorporated herein by reference in its entirety.
  • the device includes the following components:
  • Lubricant silicone medical fluid
  • Compressed osmotic engine 76.4% NaCl, 15.5% sodium carboxymethyl cellulose, 6% povidone, 0.5% Mg Stearate, 1.6% water
  • PEG 400 8 mg added to osmotic engine to fill air spaces
  • Membrane plug polyurethane polymer, injection molded to desired shape
  • Back diffusion Regulating Outlet polyethylene
  • Drug formulation (1) 0.150 g of 60% water and 40% leuprolide acetate; or (2) leuprolide acetate dissolved in DMSO to a measured content of 65 mg leuprolide.
  • the piston and inner diameter of the reservoir are lightly lubricated.
  • the piston is inserted about 0.5 cm into the reservoir at the membrane end.
  • PEG 400 is added into the reservoir.
  • Two osmotic engine tablets (40 mg each) are then inserted into the reservoir from the membrane end. After insertion, the osmotic engine is flush with the end of the reservoir.
  • the membrane plug is inserted by lining up the plug with the reservoir and pushing gently until the retaining features of the plug are fully engaged in the reservoir.
  • Formulation is loaded into a syringe which is then used to fill the reservoir from the outlet end by injecting formulation into the open tube until the formulation is about 3 mm from the end.
  • the filled reservoir is centrifuged (outlet end “up”) to remove any air bubbles that have been trapped in the formulation during filling.
  • the outlet is screwed into the open end of the reservoir until completely engaged. As the outlet is screwed in, excess formulation exits out of the orifice ensuring a uniform fill.
  • These devices deliver about 0.35 ⁇ L/day leuprolide formulation containing on average 150 ⁇ g leuprolide in the amount delivered per day. They provide delivery of leuprolide at this rate for at least one year. The devices can achieve approximately 70% steady-state delivery by day 14.
  • Exenatide suspension formulations are produced as described in Example 1 and loaded into an implantable delivery device as above.
  • Two implantable devices one including an exenatide formulation and one including a leuprolide formulation are implanted under local anesthetic and by means of an incision in a patient suffering from advanced prostatic cancer. Implantation can be accomplished using, for example, an implanter device. See e.g., U.S. Pat. No. 6,190,350, incorporated herein by reference in its entirety.
  • the implantable delivery devices are removed under local anesthetic. New devices may be inserted at that time.
  • Embodiments of the present invention include, but are not limited to, the following:
  • a method of treating cancer in a subject in need of such treatment comprising: administering a GLP-1 receptor agonist to said subject.
  • GLP-1 receptor agonist is a glucagon-like peptide-1 (GLP-1), a derivative of GLP-1, or an analog of GLP-1.
  • GLP-1 glucagon-like peptide-1
  • GLP-1 receptor agonist is selected from the group consisting of liraglutide, albiglutide, semaglutide and taspoglutide.
  • the GLP-1 receptor agonist is provided in a suspension formulation comprising: (a) a particle formulation comprising said GLP-1 receptor agonist; and (b) a vehicle formulation, wherein the particle formulation is dispersed in the vehicle.
  • the particle formulation additionally comprises a disaccharide, methionine and a buffer and (b) the vehicle formulation is a non-aqueous, single-phase suspension vehicle comprising one or more pyrrolidone polymers and one or more solvents selected from the group consisting of lauryl lactate, lauryl alcohol, benzyl benzoate, and mixtures thereof; wherein the suspension vehicle exhibits viscous fluid characteristics, and the particle formulation is dispersed in the vehicle.
  • the buffer is selected from the group consisting of citrate, histidine, succinate, and mixtures thereof.
  • disaccharide is selected from the group consisting of lactose, sucrose, trehalose, cellobiose, and mixtures thereof.

Abstract

The use of GLP-1 receptor agonists, such as glucagon-like peptide-1 (GLP-1) or exenatide, for the treatment of cancer is described. The GLP-1 receptor agonists are typically delivered using an implanted osmotic delivery device that provides for continuous delivery of the GLP-1 receptor agonist for at least one month. Additional beneficial agents, such as anticancer agents, can also be administered.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application claims the benefit of U.S. Provisional Application Ser. No. 61/443,628, filed 16 Feb. 2011, now pending, which application is herein incorporated by reference in its entirety.
  • TECHNICAL FIELD
  • The present invention relates to formulations and methods for treating cancer. Aspects of the present invention provide formulations of glucagon-like peptide-1 (GLP-1) receptor agonists for use in mammals for the treatment of cancers.
  • BACKGROUND OF THE INVENTION
  • Glycolysis is the metabolic pathway that converts glucose into pyruvate. The free energy released in this process is used to form the high-energy compounds ATP and NADH. Increased aerobic glycolysis is seen in a variety of cancer cells, a phenomenon known as the Warburg theory. Under aerobic conditions, some tumor cells produce as much as 60% of their ATP through glycolysis (Nakashima et al., Cancer Res. (1984) 44:5702-5706) as opposed to normal cells which normally generate ATP through mitochondrial oxidative phosphorylation. In addition to increased aerobic glycolysis, increased glycolysis is also seen in tumors that reach a size that exceeds the capacity of blood supply due to hypoxia. For a review of the Warburg theory and implications thereof, see, e.g., Chen et al., J. Bioenerg. Biomenzbr. (2007) 39:267-274.
  • Glucagon-like peptide-1 (GLP-1) is an important hormone and a fragment of the human proglucagon molecule. GLP-1 is rapidly metabolized by a peptidase (dipeptidylpeptidase IV or DPP-IV). A fragment of GLP-1, glucagon-like peptide-1 (7-36) amide (also known as GLP-1 (7-36) amide, glucagon-like insulinotropic peptide, or GLIP) is a gastrointestinal peptide that potentiates the release of insulin in physiologic concentrations (Gutniak et al., N Engl J Med (1992) 14:326(20):1316-22). Food intake, as well as stimulation of the sympathetic nervous system, stimulates secretion of GLP-1 in the small intestine of mammals. Further, GLP-1 stimulates the production and secretion of insulin, the release of somatostatin, glucose utilization by increasing insulin sensitivity, and, in animal studies, also stimulates beta-cell function and proliferation. GLP-1(7-36)amide and GLP-1(7-37) normalize fasting hyperglycemia in type 2 diabetic patients (Nauck, M. A., et al., Diabet. Med. 15(11):937-45 (1998)).
  • Exendin-4, a GLP-1 receptor agonist, is a molecule purified from Heloderma suspectuni venom (Eng, et al., Biol. Chem. (1992) 267:7402-7405) and shows structural relationship to the hormone GLP-1(7-36)amide. Exendin-4 and truncated exendin-(9-39)amide specifically interact with the GLP-1 receptor on insulinoma-derived cells and on lung membranes (Göke et al., J Biol. Chem. (1993) 268:19650-19655). Exendin-4 has approximately 53% identity to human GLP-1 (Pohl, et al., J. Biol. Chem. (1998) 273:9778-9784). Unlike GLP-1, however, exendin-4 is resistant to degradation by DPP-IV. A glycine substitution confers resistance to degradation by DPP-1V (Young, et al., Diabetes (1999) 48(5):1026-1034).
  • The increased dependency of cancer cells on the glycolytic pathway is an important metabolic difference between normal and malignant cells. The present invention provides a unique solution to disrupting cancer cell energy reliance on the glycolytic pathway.
  • SUMMARY OF THE INVENTION
  • The present invention relates to compositions, devices and methods for treating cancer. The invention utilizes GLP-1 receptor agonists to restrict glucose as an energy source for cancer cells and tumors. The GLP-1 receptor agonists can be used alone or in combination with other beneficial agents, such as anticancer agents, antidiabetic agents and the like, as well as in combination with anticancer treatment modalities, such as radiation, surgery and chemotherapeutic regimens.
  • Thus, in one aspect the invention relates to a method of treating cancer in a subject in need of such treatment, comprising administering a GLP-1 receptor agonist to said subject.
  • In certain aspects of the method, the GLP-1 receptor agonist is a glucagon-like peptide-1 (GLP-1), a derivative of GLP-1, or an analog of GLP-1. In some embodiments, the GLP-1 receptor agonist is GLP(7-36)amide comprising the sequence of SEQ ID NO:1.
  • In other aspects of the invention, the GLP-1 receptor agonist is exenatide, a derivative of exenatide, or an analog of exenatide, such as a synthetic exenatide peptide comprising the sequence of SEQ ID NO:2.
  • In additional aspects of the invention, the GLP-1 receptor agonist is selected from the group consisting of lixisenatide, liraglutide (VICTOZA®), albiglutide (SYNCRIA™), semaglutide, taspoglutide, BYETTA®, BYDUREON® and LY2189265. In some embodiments, formulations comprising the GLP-1 receptor agonist are delivered by injection.
  • In further aspects, the GLP-1 receptor agonist is delivered using an implanted drug delivery device, such as an osmotic delivery device, that provides continuous delivery of a suspension formulation of GLP-1 receptor agonist for a period of at least one month.
  • In other aspects, the GLP-1 receptor agonist and/or other beneficial agent is provided in a suspension formulation comprising: (a) a particle formulation comprising said GLP-1 receptor agonist and/or beneficial agent; and (b) a vehicle formulation, wherein the particle formulation is dispersed in the vehicle.
  • In additional aspects, the suspension formulation may further comprise a particle formulation comprising a GLP-1 receptor agonist and/or beneficial agent and one or more stabilizers selected from the group consisting of carbohydrates, antioxidants, amino acids, buffers, and inorganic compounds. The suspension formulation further comprises a non-aqueous, single-phase suspension vehicle comprising one or more polymers and one or more solvents. The suspension vehicle typically exhibits viscous fluid characteristics and the particle formulation is dispersed in the vehicle.
  • In another embodiment, the suspension formulation comprises a particle formulation comprising a GLP-1 receptor agonist and/or a beneficial agent, a disaccharide (e.g., sucrose), methionine, and a buffer (e.g., citrate), and a non-aqueous, single-phase suspension vehicle comprising one or more pyrrolidone polymer (e.g., polyvinylpyrollidone) and one or more solvent (e.g., lauryl lactate, lauryl alcohol, benzyl benzoate, or mixtures thereof.
  • The particle formulations of the present invention may further comprise a buffer, for example, selected from the group consisting of citrate, histidine, succinate, and mixtures thereof.
  • The particle formulations of the present invention may further comprise an inorganic compound, for example, selected from the group consisting of citrate, histidine, succinate, and mixtures thereof. NaCl, Na2SO4, NaHCO3, KCl, KH2PO4, CaCl2, and MgCl2.
  • The one or more stabilizers in the particle formulations may comprise, for example, a carbohydrate selected from the group consisting of lactose, sucrose, trehalose, mannitol, cellobiose, and mixtures thereof.
  • The one or more stabilizers in the particle formulations may comprise, for example, a antioxidant selected from the group consisting of methionine, ascorbic acid, sodium thiosulfate, ethylenediaminetetraacetic acid (EDTA), citric acid, cysteins, thioglycerol, thioglycolic acid, thiosorbitol, butylated hydroxanisol, butylated hydroxyltoluene, and propyl gallate, and mixtures thereof.
  • The one or more stabilizers in the particle formulations may comprise an amino acid.
  • In one embodiment, the solvent of the suspension vehicle of the present invention is selected from the group consisting of lauryl lactate, lauryl alcohol, benzyl benzoate, and mixtures thereof. An example of a polymer that can be used to formulate the suspension vehicle is a pyrrolidone (e.g., polyvinylpyrrolidone). In a preferred embodiment, the polymer is a pyrrolidone and the solvent is benzyl benzoate.
  • The suspension formulation typically has an overall moisture content less than about 10 wt % and in a preferred embodiment less than about 5 wt %.
  • In additional embodiments, a beneficial agent, such as an anticancer agent, in addition to the GLP-1 receptor agonist is delivered to said subject. In certain embodiments, the anticancer agent is a chemotherapeutic agent and/or an anticancer antibody. The additional beneficial agent can be delivered prior to, subsequent to or concurrent with the GLP-1 receptor agonist. In some embodiments, an implantable drug delivery device may be used to deliver formulations comprising an anticancer agent. In one embodiment, the device is an osmotic delivery device.
  • In some embodiments, implantable drug delivery devices deliver a GLP-1 receptor agonist formulations and/or other beneficial agent formulations at a substantially uniform rate for a period of about one month to about a year. Such devices may, for example, be implanted subcutaneously in convenient locations.
  • The present invention also includes methods of manufacturing the suspension formulations, particle formulations, suspension vehicles, and devices of the present invention as described herein.
  • These and other embodiments of the present invention will readily occur to those of ordinary skill in the art in view of the disclosure herein.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIGS. 1A and 1B present the sequences of two representative GLP-1 receptor agonists: FIG. 1A, glucagon-like peptide 1 (7-36)amide (GLP-1(7-36)amide) (SEQ ID NO:1), and FIG. 1B, synthetic exenatide peptide (SEQ ID NO:2).
  • FIG. 2 presents a partial cross-sectional view of one embodiment of an osmotic delivery device useful in the practice of the present invention.
  • DETAILED DESCRIPTION OF THE INVENTION
  • All patents, publications, and patent applications cited in this specification are herein incorporated by reference as if each individual patent, publication, or patent application was specifically and individually indicated to be incorporated by reference in its entirety for all purposes.
  • 1.0.0 DEFINITIONS
  • It is to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting. As used in this specification and the appended claims, the singular forms “a,” “an” and “the” include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to “a GLP-1 receptor agonist” includes a combination of two or more such molecules, reference to “a peptide” includes one or more peptides, mixtures of peptides, and the like.
  • Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the invention pertains. Although other methods and materials similar, or equivalent, to those described herein can be used in the practice of the present invention, the preferred materials and methods are described herein.
  • In describing and claiming the present invention, the following terminology will be used in accordance with the definitions set out below.
  • The terms “peptide,” “polypeptide,” and “protein” are used interchangeably herein and typically refer to a molecule comprising a chain of two or more amino acids (e.g., most typically L-amino acids, but also including, e.g., D-amino acids, modified amino acids, amino acid analogs, and/or amino acid mimetic). Peptides may also comprise additional groups modifying the amino acid chain, for example, functional groups added via post-translational modification. Examples of post-translation modifications include, but are not limited to, acetylation, alkylation (including, methylation), biotinylation, glutamylation, glycylation, glycosylation, isoprenylation, lipoylation, phosphopantetheinylation, phosphorylation, selenation, and C-terminal amidation. The term peptide also includes peptides comprising modifications of the amino terminus and/or the carboxy terminus. Modifications of the terminal amino group include, but are not limited to, des-amino, N-lower alkyl, N-di-lower alkyl, and N-acyl modifications. Modifications of the terminal carboxy group include, but are not limited to, amide, lower alkyl amide, dialkyl amide, and lower alkyl ester modifications (e.g., wherein lower alkyl is C1-C4 alkyl).
  • The terminal amino acid at one end of the peptide chain typically has a free amino group (i.e., the amino terminus). The terminal amino acid at the other end of the chain typically has a free carboxyl group (i.e., the carboxy terminus). Typically, the amino acids making up a peptide are numbered in order, starting at the amino terminus and increasing in the direction of the carboxy terminus of the peptide.
  • The phrase “amino acid residue” as used herein refers to an amino acid that is incorporated into a peptide by an amide bond or an amide bond mimetic.
  • The term “GLP-1 receptor agonist” as used herein refers to an agent capable of binding and activating the GLP-1 receptor. The term includes GLP-1 hormones, as well as GLP-1 peptides, peptide analogs thereof, or peptide derivatives thereof. Also encompassed by the term GLP-1 receptor agonist are other molecules that are capable of binding and activating the GLP-1 receptor, such as without limitation, an exenatide peptide, a peptide analog thereof, or a peptide derivative thereof. Specific examples of preferred GLP-1 receptor agonists include exenatide having the amino acid sequence of exendin-4, GLP-1(7-36)amide, lixisenatide, liraglutide (VICTOZA®), albiglutide (SYNCRIA™), semaglutide, taspoglutide, BYETTA®, BYDUREON® and LY2189265. The term also includes small molecules capable of binding and activating the GLP-1 receptor. See, e.g., Sloop et al., Diabetes (2010) 59:3099-3107.
  • The term “anticancer agent” refers to any agent that exhibits anti-tumor activity as defined below. Such agents include, without limitation, chemotherapeutic agents (i.e., a chemical compound or combination of compounds useful in the treatment of cancer), anticancer antibodies, agents that disrupt nucleic acid transcription and/or translation, such as antisense oligonucleotides, small interfering RNA (siRNA), and the like.
  • By “anti-tumor activity” is intended a reduction in the rate of cell proliferation, and hence a decline in growth rate of an existing tumor or in a tumor that arises during therapy, and/or destruction of existing neoplastic (tumor) cells or newly formed neoplastic cells, and hence a stabilization or decrease in the overall size of a tumor during therapy.
  • By “antidiabetic agent” is meant any agent that when administered to a subject either directly or indirectly causes a reduction in glucose levels. Such agents include, without limitation, agents for treating types 1 and 2 diabetes, such as but not limited to, GLP-1 receptor agonists; small molecules such as metformin, tolbutamide, glibenclamide, glipizide, gliquidone, glibornuride, tolazamide, sulfonylureas, meglitinides (e.g., repaglinide, and nateglinide); thiazolidinediones (TZDs), such as pioglitazone; SGLT1 and SGLT2 inhibitors; alpha glucosidase inhibitors; amylin (as well as synthetic analogs such as pramlintide); dipeptidyl peptidase IV (DPP-1V) inhibitors (e.g., saxagliptin, sitagliptin, alogliptin and vildagliptin); long/short acting insulins; glucagon receptor antagonists; GRP agonists (e.g., GRP-119 and GRP-40), and the like. Use of oral dipeptidyl peptidase-IV (DPP-IV or DPP-4) inhibitors orally to prevent cleavage of GLP-1 may be particularly useful when the formulation comprises a GLP-1 that is cleavable by dipeptidyl peptidase-1V (see, e.g., U.S. Pat. No. 7,205,409, incorporated herein by reference in its entirety).
  • An “antibody” intends a molecule that binds to an epitope of interest present in an antigen. The term “antibody” as used herein includes antibodies obtained from both polyclonal and monoclonal preparations, as well as, the following: hybrid (chimeric) antibody molecules (see, for example, Winter et al., Nature (1991) 349:293-299; and U.S. Pat. No. 4,816,567); F(ab′)2 and F(ab) fragments; Fv molecules (non-covalent heterodimers, see, for example, Inbar et al., Proc Natl Acad Sci USA (1972) 69:2659-2662; and Ehrlich et al., Biochem (1980) 19:4091-4096); single-chain Fv molecules (sFv) (see, for example, Huston et al., Proc Natl Acad Sci USA (1988) 85:5879-5883); dimeric and trimeric antibody fragment constructs; diabodies; avamers; aptamers; affitins; affitins; anticalins; affibody molecules; designed ankyrin repeat proteins; domain antibodies; minibodies (see, e.g., Pack et al., Biochem (1992) 31:1579-1584; Cumber et al., J Immunology (1992) 149B:120-126); humanized antibody molecules (see, for example, Riechmann et al., Nature (1988) 332:323-327; Verhoeyan et al., Science (1988) 239:1534-1536; and U.K. Patent Publication No. GB 2,276,169, published 21 Sep. 1994); and, any functional fragments obtained from such molecules, or fusions thereof, wherein such fragments and fusions retain immunological binding properties of the parent antibody molecule. Chimeric antibodies composed of human and non-human amino acid sequences may be formed from monoclonal antibody molecules to reduce their immunogenicity in humans (Winter et al. (1991) Nature 349:293; Lobuglio et al. (1989) Proc. Nat. Acad. Sci. USA 86:4220; Shaw et al. (1987) J Immunol. 138:4534; and Brown et al. (1987) Cancer Res. 47:3577; Rieclunann et al. (1988) Nature 332:323; Verhoeyen et al. (1988) Science 239:1534; and Jones et al. (1986) Nature 321:522; EP Publication No. 519,596, published 23 Dec. 1992; and U.K. Patent Publication No. GB 2,276,169, published 21 Sep. 1994).
  • As used herein, the term “monoclonal antibody” refers to an antibody composition having a homogeneous antibody population. The term is not limited regarding the species or source of the antibody, nor is it intended to be limited by the manner in which it is made. The term encompasses whole immunoglobulins as well as fragments such as Fab, F(ab′)2, Fv, and other fragments, as well as chimeric and humanized homogeneous antibody populations, that exhibit immunological binding properties of the parent monoclonal antibody molecule.
  • As used herein, the term “anti-cancer antibody” encompasses antibodies that have been designed to target cancer cells, particularly cell-surface antigens residing on cells of a particular cancer of interest.
  • The term “vehicle” as used herein refers to a medium used to carry a compound, e.g., a drug. Vehicles of the present invention typically comprise components such as polymers and solvents. The suspension vehicles of the present invention typically comprise solvents and polymers that are used to prepare suspension formulations further comprising drug particle formulations.
  • The phrase “phase separation” as used herein refers to the formation of multiple phases (e.g., liquid or gel phases) in the suspension vehicle, such as when the suspension vehicle contacts the aqueous environment. In some embodiments of the present invention, the suspension vehicle is formulated to exhibit phase separation upon contact with an aqueous environment having less than approximately 10% water.
  • The phrase “single-phase” as used herein refers to a solid, semisolid, or liquid homogeneous system that is physically and chemically uniform throughout.
  • The term “dispersed” as used herein refers to dissolving, dispersing, suspending, or otherwise distributing a compound, for example, a peptide, in a suspension vehicle.
  • A “homogeneous suspension” typically refers to a particle that is insoluble in a suspension vehicle and is distributed uniformly in a suspension vehicle.
  • The phrase “chemically stable” as used herein refers to formation in a formulation of an acceptable percentage of degradation products produced over a defined period of time by chemical pathways, such as deamidation, (usually by hydrolysis), aggregation, or oxidation.
  • The phrase “physically stable” as used herein refers to formation in a formulation of an acceptable percentage of aggregates (e.g., dimers and other higher molecular weight products). Further, a physically stable formulation does not change its physical state as, for example, from liquid to solid, or from amorphous to crystal form.
  • The term “viscosity” as used herein typically refers to a value determined from the ratio of shear stress to shear rate (see, e.g., Considine, D. M. & Considine, G. D., Encyclopedia of Chemistry, 4th Edition, Van Nostrand, Reinhold, N.Y., 1984) essentially as follows:

  • F/A=μ*V/L  (Equation 1)
  • where F/A=shear stress (force per unit area).
  • μ=a proportionality constant (viscosity), and
  • V/L=the velocity per layer thickness (shear rate).
  • From this relationship, the ratio of shear stress to shear rate defines viscosity. Measurements of shear stress and shear rate are typically determined using parallel plate rheometery performed under selected conditions (for example, a temperature of about 37° C.). Other methods for the determination of viscosity include, measurement of a kinematic viscosity using a viscometer, for example, a Cannon-Fenske viscometer, a Ubbelohde viscometer for the Cannon-Fenske opaque solution, or a Ostwald viscometer. Generally, suspension vehicles of the present invention have a viscosity sufficient to prevent particles suspended therein from settling during storage and use in a method of delivery, for example, in an implantable, drug delivery device.
  • The term “non-aqueous” as used herein refers to an overall moisture content, for example, of a suspension formulation, typically of less than or equal to about 10 wt %, preferably less than or equal to about 5 wt %, and more preferably less than about 4 wt %.
  • The term “subject” as used herein refers to any member of the subphylum chordata, including, without limitation, humans and other primates, including non-human primates such as rhesus macaque, chimpanzees and other apes and monkey species; farm animals such as cattle, sheep, pigs, goats and horses; domestic mammals such as dogs and cats; laboratory animals including rodents such as mice, rats and guinea pigs; birds, including domestic, wild and game birds such as chickens, turkeys and other gallinaceous birds, ducks, geese, and the like. The term does not denote a particular age. Thus, both adult and newborn individuals are intended to be covered.
  • The terms “drug,” “therapeutic agent”, and “beneficial agent” are used interchangeably to refer to any therapeutically active substance that is delivered to a subject to produce a desired beneficial effect. In one embodiment of the present invention, the drug is a GLP-1 receptor agonist, e.g., GLP-1 (7-36)amide, exenatide, and derivatives or analogs thereof. The devices and methods of the present invention are well suited for the delivery of polypeptides as well as small molecules and combinations thereof.
  • The term “osmotic delivery device” as used herein typically refers to a device used for delivery of one or more GLP-1 receptor agonists, or other beneficial agents to a subject, wherein the device comprises, for example, a reservoir (made, for example, from a titanium alloy) having a lumen that contains, in one chamber, a beneficial agent formulation (e.g., comprising one or more beneficial agent) and, in another chamber, an osmotic agent formulation. A piston assembly positioned in the lumen isolates the beneficial agent formulation from the osmotic agent formulation. A semi-permeable membrane (also termed a semi-permeable plug) is positioned at a first distal end of the reservoir adjacent the osmotic agent formulation. A diffusion moderator (which defines a delivery orifice through which the beneficial agent formulation exits the device) is positioned at a second distal end of the reservoir adjacent the suspension formulation. The piston assembly and the diffusion moderator define a chamber that contains the beneficial agent formulation and the piston assembly and the semipermeable membrane define a chamber that contains the osmotic agent formulation. The terms “flow modulator,” “diffusion modulator,” “flow moderator,” and “diffusion moderator” are used interchangeably herein. Typically, the osmotic delivery device is implanted within the subject, for example, subcutaneously (e.g., in the inside, outside, or back of the upper arm; or in the abdominal area). An exemplary osmotic delivery device is the DUROe delivery device.
  • 2.0.0 GENERAL OVERVIEW OF THE INVENTION
  • Before describing the present invention in detail, it is to be understood that this invention is not limited to particular types of drug delivery, particular types of drug delivery devices, particular sources of peptides, particular solvents, particular polymers, and the like, as use of such particulars may be selected in view of the teachings of the present specification. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments of the invention only, and is not intended to be limiting.
  • In one aspect, the present invention relates to methods of treating cancer in a subject in need of treatment, including, but not limited to, treating hematological tumors and solid tumors. The method comprises providing delivery of a GLP-1 receptor agonist formulation to a subject in need thereof. In certain embodiments, the GLP-1 receptor agonist formulation is delivered using an osmotic delivery device at a substantially uniform rate. The length of delivery of the formulation is determined based on the cancer being treated. In some embodiments, for example, the administration period is for at least about one month, at least about one month to about one year, at least about three months to about one year, at least about four months to about one year, at least about five months to about one year, at least about six months to about one year, at least about eight months to about one year, at least about nine months to about one year, or at least about 10 months to about one year. The period of administration can also exceed one year if necessary, such as from one year to two years. The method may further include subcutaneously inserting an osmotic delivery device, loaded with the GLP-1 receptor agonist formulation, into the subject.
  • In other embodiments of the invention, the GLP-1 receptor agonist is delivered parenterally (including by subcutaneous, intravenous, intramedullary, intraarticular, intramuscular, or intraperitoneal injection) rectally, topically, transdermally, intranasally, by inhalation, or orally (for example, in capsules, suspensions, or tablets). Injectable formulations of GLP-1 agonists are known and include, without limitation, lixisenatide, liraglutide (VICTOZA®), albiglutide (SYNCRIA™), semaglutide, taspoglutide, BYETTA®, BYDLIREON® and LY2189265.
  • In one embodiment of the present invention the formulation comprises a glucagon-like peptide-1 (GLP-1), a derivative of GLP-1, or an analog of GLP-1.
  • In certain embodiments, the GLP-1 receptor agonist is GLP-1(7-36)amide shown in FIG. 1A (SEQ ID NO:1).
  • In another embodiment of the present invention the formulation comprises exenatide, a derivative of exenatide, or an analog of exenatide. In certain embodiments, the exenatide is the exenatide peptide shown in FIG. 1B (SEQ ID NO:2).
  • In certain embodiments, additional beneficial agents are provided with the GLP-1 receptor agonist formulations, such as anticancer agents, including without limitation, chemotherapeutic agents, anticancer antibodies, antisense nucleotides, siRNA, anticancer vaccines, and the like. Such additional beneficial agents are described in detail below. Administration of these agents is not limited to any particular delivery system and may include, without limitation, delivery using osmotic delivery devices as described herein if the agent is suitable for such delivery, or may be parenteral (including subcutaneous, intravenous, intramedullary, intraarticular, intramuscular, or intraperitoneal injection), rectal, topical, transdermal, intranasal, by inhalation, or oral (for example, in capsules, suspensions, or tablets). Administration of the additional agents to an individual may occur in a single dose or in repeat administrations, and in any of a variety of physiologically acceptable salt forms, and/or with an acceptable pharmaceutical carrier and/or additive as part of a pharmaceutical composition. Physiologically acceptable salt forms and standard pharmaceutical formulation techniques and excipients are well known to persons skilled in the art (see, e.g., Physicians' Desk Reference (PDR) 2009, 63th ed. (PDR.net), Medical Economics Company; and Remington: The Science and Practice of Pharmacy, eds. Gennado et al., 21th ed, Lippincott, Williams & Wilkins, 2005).
  • In certain embodiments, the GLP-1 receptor agonist and/or suitable additional beneficial agents, if present, are provided in a suspension formulation, comprising a particle formulation and a suspension vehicle. The particle formulation includes, but is not limited to, the GLP-1 receptor agonist or other agent of interest and one or more stabilizers. The one or more stabilizers are typically selected from the group consisting of carbohydrates, antioxidants, amino acids, and buffers. The suspension vehicle is typically a non-aqueous, single-phase suspension vehicle comprising one or more polymers and one or more solvents. The suspension vehicle exhibits viscous fluid characteristics. The particle formulation is uniformly dispersed in the vehicle.
  • The particle formulation of the present invention typically includes one or more of the following stabilizers: one or more carbohydrates (e.g., a disaccharide, such as, lactose, sucrose, trehalose, cellobiose, and mixtures thereof); one or more antioxidants (e.g., methionine, ascorbic acid, sodium thiosulfate, ethylenediaminetetraacetic acid (EDTA), citric acid, butylated hydroxyltoluene, and mixtures thereof); and one or more buffers (e.g., citrate, histidine, succinate, and mixtures thereof). In a preferred embodiment, the particle formulation comprises a GLP-1 receptor agonist, sucrose, methionine, and citrate buffer. The ratio of the GLP-1 receptor agonist to sucrose+methionine is typically about 1/20, about 1/10, about 1/5, about 1/2, about 2/1, about 5/1, about 10/1, or about 20/1, preferably between about 1/5 to 5/1, more preferably between about 1/3 to 3/1. The particle formulation is preferably a particle formulation prepared by spray drying and has a low moisture content, preferably less than or equal to about 10 wt %, more preferably less or equal to about 5 wt %. Alternatively, the particle formulation can be lyophilized.
  • The suspension vehicle for use in the present formulations comprises one or more solvents and one or more polymers. Preferably the solvent is selected from the group consisting of lauryl lactate, lauryl alcohol, benzyl benzoate, and mixtures thereof. More preferably the solvent is lauryl lactate or benzyl benzoate. Preferably the polymer is a pyrrolidone polymer. In some embodiments the polymer is polyvinylpyrrolidone (e.g., polyvinylpyrrolidone K-17, which typically has an approximate average molecular weight range of 7,900-10,800). In one embodiment, the solvent consists essentially of benzyl benzoate and polyvinylpyrrolidone.
  • The suspension formulation typically has a low overall moisture content, for example, less than or equal to about 10 wt % and in a preferred embodiment less than or equal to about 5 wt %.
  • 2.1.0 Compositions and Formulations
  • 2.1.1 GLP-1 Receptor Agonists
  • GLP-1, including three forms of the peptide, GLP-1(1-37), GLP-1(7-37) and GLP-1(7-36)amide, as well as peptide analogs of GLP-1 have been shown to stimulate insulin secretion (i.e., they are insulinotropic), which results in decreases in serum glucose concentrations (see, e.g., Mojsov, S., Int. J. Peptide Protein Research (1992) 40:333-343). The sequence of GLP-1(7-36)amide is shown in FIG. 1A and SEQ ID NO:1.
  • Numerous GLP-1 peptide derivatives and peptide analogs demonstrating insulinotropic action are known in the art (see, e.g., U.S. Pat. Nos. 5,118,666; 5,120,712; 5,512,549; 5,545,618; 5,574,008; 5,574,008; 5,614,492; 5,958,909; 6,191,102; 6,268,343; 6,329,336; 6,451,974; 6,458,924; 6,514,500; 6,593,295; 6,703,359; 6,706,689; 6,720,407; 6,821,949; 6,849,708; 6,849,714; 6,887,470; 6,887,849; 6,903,186; 7,022,674; 7,041,646; 7,084,243; 7,101,843; 7,138,486; 7,141,547; 7,144,863; and 7,199,217, all of which are incorporated herein by reference in their entireties), as well as in clinical trials (e.g., taspoglutide and albiglutide). One example of a GLP-1 peptide derivative useful in the practice of the present invention is VICTOZA® (liraglutide; U.S. Pat. Nos. 6,268,343, 6,458,924, 7,235,627, incorporated herein by reference in their entireties). Once-daily injectable VICTOZA® (liraglutide) is commercially available in the United States, Europe, and Japan. Other injectable GLP-1 peptides for use with the present invention are described above and include, without limitation taspoglutide, albiglutide (SYNCRIA™), LY2189265 and semaglutide. For ease of reference the family of GLP-1 peptides, GLP-1 peptide derivatives and GLP-1 peptide analogs having insulinotropic activity is referred to collectively as “GLP-1.”
  • The molecule exenatide has the amino acid sequence of exendin-4 (Kolterman O. G., et al., J. Clin. Endocrinol. Metab. (2003) 88(7):3082-3089) and is produced by chemical synthesis or recombinant expression. Twice-daily injectable exenatide is commercially available in the United States and Europe, and sold under the tradename of BYETTA®. Another injectable exenatide under development is BYDUREON®. Exendin-3 and exendin-4 are known in the art and were originally isolated from Heloderma spp. (Eng, et al., J. Biol. Chem. (1990) 265:20259-62; Eng., et al., J. Biol. Chem. (1992) 267:7402-05). Numerous exenatide peptide derivatives and peptide analogs (including, e.g., exendin-4 agonists) are known in the art (see, e.g., U.S. Pat. Nos. 5,424,286; 6,268,343; 6,329,336; 6,506,724; 6,514,500; 6,528,486; 6,593,295; 6,703,359; 6,706,689; 6,767,887; 6,821,949; 6,849,714; 6,858,576; 6,872,700; 6,887,470; 6,887,849; 6,924,264; 6,956,026; 6,989,366; 7,022,674; 7,041,646; 7,115,569; 7,138,375; 7,141,547; 7,153,825; and 7,157,555, all of which are incorporated herein by reference in their entireties). One example of an exenatide derivative useful in the practice of the present invention is lixisenatide (see, e.g., U.S. Pat. No. 6,528,486, incorporated herein by reference in its entirety). For ease of reference herein, the family of exenatide peptides (e.g., including exendin-3, exendin-4, and exendin-4-amide), exenatide peptide derivatives, and exenatide peptide analogs is referred to collectively as “exenatide.”
  • 2.1.2 Suspension Formulations
  • In one aspect, the present invention utilizes particle formulations of GLP-1 receptor agonists described above that can be used to prepare suspension formulations. The GLP-1 receptor agonists for use with the present invention shall not be limited by method of synthesis or manufacture and shall include those obtained from natural sources, or synthesized or manufactured by recombinant (whether produced from cDNA or genomic DNA), synthetic, transgenic, and gene-activated methods. In preferred embodiments of the present invention, the GLP-1 receptor agonist is a GLP-1 peptide or an exendin peptide (as described above), for example, GLP-1(7-36)amide or exenatide, such as the exenatide peptide shown in FIG. 1B and SEQ ID NO:2. The present invention also includes combinations of two or more such agents, for example, GLP-1(7-36)amide and GIP.
  • Particle formulations are preferably chemically and physically stable for at least one month, preferably at least three months, more preferably at least six months, more preferably at least 12 months at delivery temperature. The delivery temperature is typically normal human body temperature, for example, about 37° C., or slightly higher, for example, about 40° C. Further, particle formulations are preferably chemically and physically stable for at least three months, preferably at least six months, more preferably at least 12 months, at storage temperature. Examples of storage temperatures include refrigeration temperature, for example, about 5° C., or room temperature, for example, about 25° C.
  • A particle formulation may be considered chemically stable if less than about 25%, preferably less than about 20%, more preferably less than about 15%, more preferably less than about 10%, and more preferably less than about 5% breakdown products of the peptide particles are formed after about three months, preferably after about six months, preferably after about 12 months at delivery temperature and after about six months, after about 12 months, and preferably after about 24 months at storage temperature.
  • A particle formulation may be considered physically stable if less than about 10%, preferably less than about 5%, more preferably less than about 3%, more preferably less than 1% aggregates of the peptide particles are formed after about three months, preferably after about six months, at delivery temperature and about 6 months, preferably about 12 months, at storage temperature.
  • To preserve protein stability, a GLP-1 receptor agonist solution is generally kept in a frozen condition and lyophilized or spray dried to a solid state. Tg (glass transition temperature) may be one factor to consider in achieving stable compositions of peptide. While not intending to be bound by any particular theory, the theory of formation of a high Tg amorphous solid to stabilize peptides, polypeptides, or proteins has been utilized in pharmaceutical industry. Generally, if an amorphous solid has a higher Tg, such as 100° C., peptide products will not have mobility when stored at room temp or even at 40° C. because the storage temperature is below the Tg. Calculations using molecular information have shown that if a glass transition temperature is above a storage temperature of 50° C. that there is zero mobility for molecules. No mobility of molecules correlates with no instability issues. Tg is also dependent on the moisture level in the product formulation. Generally, the more moisture, the lower the Tg of the composition.
  • Accordingly, in some aspects of the present invention, excipients with higher Tg may be included in the protein formulation to improve stability, for example, sucrose (Tg=75° C.) and trehalose (Tg=110° C.). Preferably, particle formulations are formable into particles using processes such as spray drying, lyophilization, desiccation, milling, granulation, ultrasonic drop creation, crystallization, precipitation, or other techniques available in the art for forming particles from a mixture of components. The particles are preferably substantially uniform in shape and size.
  • A typical spray dry process may include, for example, loading a spray solution containing a peptide, for example, GLP-1(7-36)amide or exenatide, and stabilizing excipients into a sample chamber. The sample chamber is typically maintained at a desired temperature, for example, refrigeration to room temperature. Refrigeration generally promotes stability of the protein. A solution, emulsion, or suspension is introduced to the spray dryer where the fluid is atomized into droplets. Droplets can be formed by use of a rotary atomizer, pressure nozzle, pneumatic nozzle, or sonic nozzle. The mist of droplets is immediately brought into contact with a drying gas in a drying chamber. The drying gas removes solvent from the droplets and carries the particles into a collection chamber. In spray drying, factors that can affect yield include, but are not limited to, localized charges on particles (which may promote adhesion of the particles to the spray dryer) and aerodynamics of the particles (which may make it difficult to collect the particles). In general, yield of the spray dry process depends in part on the particle formulation.
  • In one embodiment, the particles are sized such that they can be delivered via an implantable drug delivery device. Uniform shape and size of the particles typically helps to provide a consistent and uniform rate of release from such a delivery device; however, a particle preparation having a non-normal particle size distribution profile may also be used. For example, in a typical implantable osmotic delivery device having a delivery orifice, the size of the particles is less than about 30%, preferably is less than about 20%, more preferably is less than about than 10%, of the diameter of the delivery orifice. In an embodiment of the particle formulation for use with an osmotic delivery device, wherein the delivery orifice diameter of the implant is in a range of, for example, about 0.1 to about 0.5 mm, particle sizes may be preferably less than about 50 microns, more preferably less than about 10 microns, more preferably in a range from about 3 to about 7 microns. In one embodiment, the orifice is about 0.25 mm (250 microns) and the particle size is approximately 3-5 microns.
  • In a preferred embodiment, when the particles are incorporated in a suspension vehicle they do not settle in less than about three months at delivery temperature. Generally speaking, smaller particles tend to have a lower settling rate in viscous suspension vehicles than larger particles. Accordingly, micron- to nano-sized particles are typically desirable. In an embodiment of the particle formulation for use in an implantable osmotic delivery device, wherein the delivery orifice diameter of the implant is in a range of, for example, about 0.1 to about 0.5 mm, particle sizes may be preferably less than about 50 microns, more preferably less than about 10 microns, more preferably in a range from about 3 to about 7 microns.
  • In one embodiment, a particle formulation for use with the present invention comprises one or more GLP-1 receptor agonists, as described above and one or more stabilizers. The stabilizers may be, for example, carbohydrate, antioxidant, amino acid, buffer, or inorganic compound. The amounts of stabilizers in the particle formulation can be determined experimentally based on the activities of the stabilizers and buffers and the desired characteristics of the formulation. Typically, the amount of carbohydrate in the formulation is determined by aggregation concerns. In general, the carbohydrate level should not be too high so as to avoid promoting crystal growth in the presence of water due to excess carbohydrate unbound to insulinotropic peptide. Typically, the amount of antioxidant in the formulation is determined by oxidation concerns, while the amount of amino acid in the formulation is determined by oxidation concerns and/or formability of particles during spray drying. Typically, the amount of buffer components in the formulation is determined by pre-processing concerns, stability concerns, and formability of particles during spray drying. Buffer may be required to stabilize the GLP-1 receptor agonist during processing, e.g., solution preparation and spray drying, when all excipients are solubilized.
  • Examples of carbohydrates that may be included in the particle formulation include, but are not limited to, monosaccharides (e.g., fructose, maltose, galactose, glucose, D-mannose, and sorbose), disaccharides (e.g., lactose, sucrose, trehalose, and cellobiose), polysaccharides (e.g., raffinose, melezitose, maltodextrins, dextrans, and starches), and alditols (acyclic polyols; e.g., mannitol, xylitol, maltitol, lactitol, xylitol sorbitol, pyranosyl sorbitol, and myoinsitol). Preferred carbohydrates include non-reducing sugars, such as sucrose, trehalose, and raffinose.
  • Examples of antioxidants that may be included in the particle formulation include, but are not limited to, methionine, ascorbic acid, sodium thiosulfate, catalase, platinum, ethylenediaminetetraacetic acid (EDTA), citric acid, cysteins, thioglycerol, thioglycolic acid, thiosorbitol, butylated hydroxanisol, butylated hydroxyltoluene, and propyl gallate.
  • Examples of amino acids that may be included in the particle formulation include, but are not limited to, arginine, methionine, glycine, histidine, alanine, L-leucine, glutamic acid, iso-leucine, L-threonine, 2-phenylamine, valine, norvaline, praline, phenylalanine, trytophan, serine, asparagines, cysteine, tyrosine, lysine, and norleucine. Preferred amino acids include those that readily oxidize, e.g., cysteine, methionine, and trytophan.
  • Examples of buffers that may be included in the particle formulation include, but are not limited to, citrate, histidine, succinate, phosphate, maleate, tris, acetate, carbohydrate, and gly-gly. Preferred buffers include citrate, histidine, succinate, and tris. It is to be understood that buffers can be added to the solution before formation of the particles, for example, by spray drying. However, after the dry particle formation is prepared, the buffer component no longer serves as a buffer in the dried particles. For ease of reference herein, when referring to buffer components, the term buffer is used.
  • Examples of inorganic compounds that may be included in the particle formulation include, but are not limited to, NaCl, Na2SO4, NaHCO3, KCl, KH2PO4, CaCl2, and MgCl2.
  • In addition, the particle formulation may include other excipients, such as but not limited to surfactants and salts. Examples of surfactants include, but are not limited to, Polysorbate 20, Polysorbate 80, PLURONIC®, F68, and sodium docecyl sulfate (SDS). Examples of other excipients include, but are not limited to, mannitol and glycine. Examples of salts include, but are not limited to, sodium chloride, calcium chloride, and magnesium chloride.
  • In one embodiment, the particle formulation comprises, for example, exenatide peptide, sucrose (carbohydrate), methionine (antioxidant), and sodium citrate/citric acid.
  • All components included in the particle formulation are typically acceptable for pharmaceutical use in mammals, in particular, in humans.
  • Particle size distribution of the dry particle powder can be well controlled (0.1 micron −20 micron), for example, by using the methods of spray drying or lyophilization to prepare the particle formulations. The process parameters for formation of the dry powder are optimal to produce particles with desired particle size distribution, density, and surface area.
  • The selected excipients and stabilizers in the particle formulation may provide, for example, the following functions: density modification of the dry powder; preservation of the peptide chemical stability; maintenance of the peptide's physical stability (e.g., high glass transition temperature, and avoiding phase to phase transition); producing homogenous dispersions in suspension; and modification of hydrophobicity and/or hydrophilicity to manipulate dry powder solubility in selected solvents.
  • See U.S. Patent Publication No. 2008/0260840, incorporated herein by reference in its entirety, for detailed methods of producing particle formulations.
  • In summary, GLP-1 receptor agonists can be formulated into dried powders in solid state, which preserves maximum chemical and biological stability of proteins or peptides. The particle formulation offers long term storage stability at high temperature, and therefore, allows delivery to a subject of stable and biologically effective peptide for extended periods of time.
  • Although the particle formulations described above are with reference to GLP-1 receptor agonists, such particle formulations can also be formed with any other suitable agents, such as other suitable beneficial polypeptides, including suitable anticancer polypeptides, antibodies and the like, described in detail below.
  • Suspension formulations for use with the present invention can be produced using particle formulations as described above. See U.S. Patent Publication No. 2008/0260840, incorporated herein by reference in its entirety, for detailed methods of producing such suspension formulations. In one aspect of the present invention, the suspension formulation includes a suspension vehicle to provide a stable environment in which the GLP-1 receptor agonist particle formulation (or other suitable particle formulation) is dispersed. The particle formulations are chemically and physically stable (as described above) in the suspension vehicle. The suspension vehicle typically comprises one or more polymers and one or more solvents that form a solution of sufficient viscosity to uniformly suspend the particles comprising the GLP-1 receptor agonist or other suitable agent. In addition to the GLP-1 receptor agonist, the suspension formulations can be used with any suitable agents, such as other suitable beneficial polypeptides, including suitable anticancer polypeptides, antibodies and the like, described in detail below.
  • The viscosity of the suspension vehicle is typically sufficient to prevent the particle formulation from settling during storage and use in a method of delivery, for example, in an implantable, drug delivery device. The suspension vehicle is biodegradable in that the suspension vehicle disintegrates or breaks down over a period of time in response to a biological environment. The disintegration of the suspension vehicle may occur by one or more physical or chemical degradative processes, such as by enzymatic action, oxidation, reduction, hydrolysis (e.g., proteolysis), displacement (e.g., ion exchange), or dissolution by solubilization, emulsion or micelle formation. After the suspension vehicle disintegrates, components of the suspension vehicle are absorbed or otherwise dissipated by the body and surrounding tissue of the patient.
  • The solvent in which the polymer is dissolved may affect characteristics of the suspension formulation, such as the behavior of the particle formulation during storage. A solvent may be selected in combination with a polymer so that the resulting suspension vehicle exhibits phase separation upon contact with the aqueous environment. In some embodiments, the solvent may be selected in combination with the polymer so that the resulting suspension vehicle exhibits phase separation upon contact with the aqueous environment having less than approximately about 10% water.
  • The solvent may be an acceptable solvent that is not miscible with water. The solvent may also be selected so that the polymer is soluble in the solvent at high concentrations, such as at a polymer concentration of greater than about 30%. However, typically particles comprising the GLP-1 receptor agonists are substantially insoluble in the solvent. Examples of solvents useful in the practice of the present invention include, but are not limited to, lauryl alcohol, benzyl benzoate, benzyl alcohol, lauryl lactate, decanol (also called decyl alcohol), ethyl hexyl lactate, and long chain (C8 to C24) aliphatic alcohols, esters, or mixtures thereof. The solvent used in the suspension vehicle may be “dry,” in that it has a low moisture content. Preferred solvents for use in formulation of the suspension vehicle include lauryl lactate, lauryl alcohol, benzyl benzoate, and combinations thereof.
  • Examples of polymers for formulation of the suspension vehicles include, but are not limited to, a polyester (e.g., polylactic acid or polylacticpolyglycolic acid), pyrrolidone polymer (e.g., polyvinylpyrrolidone (PVP) having a molecular weight ranging from approximately 2,000 to approximately 1,000,000), ester or ether of an unsaturated alcohol (e.g., vinyl acetate), polyoxyethylenepolyoxypropylene block copolymer, or mixtures thereof. In one embodiment, the polymer is PVP having a molecular weight of 2,000 to 1,000,000. In a preferred embodiment the polymer is polyvinylpyrrolidone K-17 (typically having an approximate average molecular weight range of 7,900-10,800). Polyvinylpyrrolidone can be characterized by its K-value (e.g., K-17), which is a viscosity index. The polymer used in the suspension vehicle may include one or more different polymers or may include different grades of a single polymer. The polymer used in the suspension vehicle may also be dry or have a low moisture content.
  • Generally speaking, a suspension vehicle according to the present invention may vary in composition based on the desired performance characteristics. In one embodiment, the suspension vehicle may comprise about 40% to about 80% (w/w) polymer(s) and about 20% to about 60% (w/w) solvent(s). Preferred embodiments of a suspension vehicle include vehicles formed of polymer(s) and solvent(s) combined at the following ratios: about 25% solvent and about 75% polymer; about 50% solvent and about 50% polymer; about 75% solvent and about 25% polymer.
  • The suspension vehicle may exhibit Newtonian behavior. The suspension vehicle is typically formulated to provide a viscosity that maintains a uniform dispersion of the particle formulation for a predetermined period of time. This helps facilitate making a suspension formulation tailored to provide controlled delivery of the insulinotropic peptide at a desired rate. The viscosity of the suspension vehicle may vary depending on the desired application, the size and type of the particle formulation, and the loading of the particle formulation in the suspension vehicle. The viscosity of the suspension vehicle may be varied by altering the type or relative amount of the solvent or polymer used.
  • The suspension vehicle may have a viscosity ranging from about 100 poise to about 1,000,000 poise, preferably from about 1,000 poise to about 100,000 poise. The viscosity may be measured at a selected temperature, for example, 33° C., at a shear rate of 10−4/sec, using a parallel plate rheometer. In some embodiments, the viscosity of the suspension vehicle ranges from approximately 5,000 poise to approximately 50,000 poise, such as about 7,000 poise to about 40,000 poise, about 8,000 poise to about 20,000 poise, about 9,000 poise to about 25,000 poise, about 10,000 poise to about 20,000 poise, and the like. In preferred embodiments, the viscosity range is between about 12,000 to about 18,000 poise at 33° C.
  • The suspension vehicle may exhibit phase separation when contacted with the aqueous environment; however, typically the suspension vehicle exhibits substantially no phase separation as a function of temperature. For example, at a temperature ranging from approximately 0° C. to approximately 70° C. and upon temperature cycling, such as cycling from 4° C. to 37° C. to 4° C., the suspension vehicle typically exhibits no phase separation.
  • The suspension vehicle may be prepared by combining the polymer and the solvent under dry conditions, such as in a dry box. The polymer and solvent may be combined at an elevated temperature, such as from approximately 40° C. to approximately 70° C., and allowed to liquefy and form the single phase. The ingredients may be blended under vacuum to remove air bubbles produced from the dry ingredients. The ingredients may be combined using a conventional mixer, such as a dual helix blade or similar mixer, set at a speed of approximately 40 rpm. However, higher speeds may also be used to mix the ingredients. Once a liquid solution of the ingredients is achieved, the suspension vehicle may be cooled to room temperature. Differential scanning calorimetry (DSC) may be used to verify that the suspension vehicle is a single phase. Further, the components of the vehicle (e.g., the solvent and/or the polymer) may be treated to substantially reduce or substantially remove peroxides (e.g., by treatment with methionine; see, e.g., U.S. Patent Application Publication No. 2007-0027105, incorporated herein by reference in its entirety).
  • The particle formulation, comprising a GLP-1 receptor agonist, or other suitable agent, is added to the suspension vehicle to form a suspension formulation. The suspension formulation may be prepared by dispersing the particle formulation in the suspension vehicle. The suspension vehicle may be heated and the particle formulation added to the suspension vehicle under dry conditions. The ingredients may be mixed under vacuum at an elevated temperature, such as from about 40° C. to about 70° C. The ingredients may be mixed at a sufficient speed, such as from about 40 rpm to about 120 rpm, and for a sufficient amount of time, such as about 15 minutes, to achieve a uniform dispersion of the particle formulation in the suspension vehicle. The mixer may be a dual helix blade or other suitable mixer. The resulting mixture may be removed from the mixer, sealed in a dry container to prevent water from contaminating the suspension formulation, and allowed to cool to room temperature before further use, for example, loading into an implantable, drug delivery device, unit dose container, or multiple-dose container.
  • The suspension formulation typically has an overall moisture content of less than about 10 wt %, preferably less than about 5 wt %, and more preferably less than about 4 wt %.
  • The suspension formulations of the present invention are exemplified herein below with reference to exenatide and GLP-1(7-36)amide as representative GLP-1 receptor agonists (see, Example 3 and Example 4). These examples are not intended to be limiting.
  • In summary, the components of the suspension vehicle provide biocompatibility. Components of the suspension vehicle offer suitable chemico-physical properties to form stable suspensions of, for example, dry powder particle formulations. These properties include, but are not limited to, the following: viscosity of the suspension; purity of the vehicle; residual moisture of the vehicle; density of the vehicle; compatibility with the dry powders; compatibility with implantable devices; molecular weight of the polymer; stability of the vehicle; and hydrophobicity and hydrophilicity of the vehicle. These properties can be manipulated and controlled, for example, by variation of the vehicle composition and manipulation of the ratio of components used in the suspension vehicle.
  • 3.0.0 DELIVERY OF SUSPENSION FORMULATIONS
  • The suspension formulations described herein may be used in an implantable, drug delivery device to provide sustained delivery of a compound over an extended period of time, such as over weeks, months, or up to about one year. Such an implantable drug delivery device is typically capable of delivering the compound at a desired flow rate over a desired period of time. The suspension formulation may be loaded into the implantable, drug delivery device by conventional techniques.
  • The suspension formulation may be delivered, for example, using an osmotically, mechanically, electromechanically, or chemically driven drug delivery device. The active agent in the suspension formulation is delivered at a flow rate that is therapeutically effective to the subject in need of treatment.
  • The active agent, such as GLP-1(7-36)amide, exenatide, or other suitable beneficial agent, may be delivered over a period ranging from more than about one week to about one year or more, preferably for about one month to about a year or more, more preferably for about three months to about a year or more. The implantable, drug delivery device may include a reservoir having at least one orifice through which the agent is delivered. The suspension formulation may be stored within the reservoir. In one embodiment, the implantable, drug delivery device is an osmotic delivery device, wherein delivery of the drug is osmotically driven. Some osmotic delivery devices and their component parts have been described, for example, the DUROS® delivery device or similar devices (see, e.g., U.S. Pat. Nos. 5,609,885; 5,728,396; 5,985,305; 5,997,527; 6,113,938; 6,132,420; 6,156,331; 6,217,906; 6,261,584; 6,270.787; 6,287,295; 6,375,978; 6,395,292; 6,508,808; 6,544,252; 6,635,268; 6,682,522; 6,923,800; 6,939,556; 6,976,981; 6,997,922; 7,014,636; 7,207,982; 7,112,335; 7,163,688; U.S. Patent Publication Nos. 2005-0175701, 2007-0281024, and 2008-0091176, all of which are incorporated herein by reference in their entireties).
  • The DUROS® delivery device typically consists of a cylindrical reservoir which contains the osmotic engine, piston, and drug formulation. The reservoir is capped at one end by a controlled-rate water-permeable membrane and capped at the other end by a diffusion moderator through which drug formulation is released from the drug reservoir. The piston separates the drug formulation from the osmotic engine and utilizes a seal to prevent the water in the osmotic engine compartment from entering the drug reservoir. The diffusion moderator is designed, in conjunction with the drug formulation, to prevent body fluid from entering the drug reservoir through the orifice.
  • The DUROS® device releases a therapeutic agent at a predetermined rate based on the principle of osmosis. Extracellular fluid enters the DUROS® device through a semi-permeable membrane directly into a salt engine that expands to drive the piston at a slow and even delivery rate. Movement of the piston forces the drug formulation to be released through the orifice or exit port at a predetermined sheer rate. In one embodiment, the reservoir of the DUROS® device is loaded with a suspension formulation comprising, for example, GLP-1(7-36)amide or exenatide, wherein the device is capable of delivering the suspension formulation to a subject over an extended period of time (e.g., about one, about two, about three, about six, or about 12 months) at a predetermined, therapeutically effective delivery rate.
  • Other implantable, drug delivery devices may be used in the practice of the present invention and may include regulator-type implantable pumps that provide constant flow, adjustable flow, or programmable flow of the compound, such as those available from Codman & Shurtleff, Inc. (Raynham, Mass.), Medtronic, Inc. (Minneapolis, Minn.), and Tricumed Medinzintechnik GmbH (Germany).
  • Implantable devices, for example, the DUROS® device, provide the following advantages for administration of the formulations of the present invention: true zero-order release of the insulinotropic peptide pharmacokinetically; long-term release period time (e.g., up to about 12 months); and reliable delivery and dosing of the GLP-1 receptor agonist or other suitable beneficial agent.
  • FIG. 2 depicts a representative osmotic delivery device useful in the practice of the present invention. In FIG. 2, an osmotic delivery device 10 is shown comprising a reservoir 12. A piston assembly 14 is positioned in the lumen of the reservoir and divides the lumen into two chambers. In this example, the chamber 16 contains a beneficial agent formulation, such as a GLP-1 receptor agonist (e.g., GLP-1 (7-36)amide or exenatide) formulation, an anticancer agent, or the like and the chamber 20 contains an osmotic agent formulation. A semi-permeable membrane 18 is positioned at a distal end of the reservoir, adjacent the chamber 20 containing the osmotic agent formulation. A diffusion moderator 22 is positioned in mating relationship at a distal end of the reservoir 12, adjacent the chamber 16 containing the beneficial agent formulation. The diffusion moderator 22 includes a delivery orifice 24. The diffusion moderator 22 may be any suitable flow device having a delivery orifice. In this embodiment, the flow path 26 is formed between a threaded diffusion moderator 22 and threads 28 formed on the interior surface of the reservoir 12. In alternative embodiments, the diffusion moderator can, for example, (i) be press-fit (or friction fit) through an opening and contacting a smooth interior surface of the reservoir, or (ii) comprise two pieces with an outer shell constructed and arranged for positioning in an opening, an inner core inserted in the outer shell, and a fluid channel having a spiral shape defined between the outer shell and the inner core (e.g., U.S. Patent Publication No. 2007-0281024, incorporated herein by reference in its entirety).
  • Fluid is imbibed into the chamber 20 through the semi-permeable membrane 18. The beneficial agent formulation is dispensed from the chamber 16 through the delivery orifice 24 in the diffusion moderator 22. The piston assembly 14 engages and seals against the interior wall of the reservoir 12, thereby isolating the osmotic agent formulation in chamber 20 and fluid imbibed through the semi-permeable membrane 18 from the beneficial agent formulation in chamber 16. At steady-state, the beneficial agent formulation is expelled through the delivery orifice 24 in the diffusion moderator 22 at a rate corresponding to the rate at which external fluid is imbibed into the chamber 20 through the semi-permeable membrane 18.
  • The semi-permeable membrane 18 may be in the form of a plug that is resiliently engaged in sealing relationship with the interior surface of the reservoir 12. In FIG. 2, it is shown to have ridges that serve to frictionally engage the semi-permeable membrane 18 with the interior surface of the reservoir 12.
  • The amount of beneficial agent employed in the delivery device of the invention is that amount necessary to deliver a therapeutically effective amount of the agent to achieve the desired therapeutic result. In practice, this will vary depending upon such variables, for example, as the particular agent, the site of delivery, the severity of the condition, and the desired therapeutic effect. Typically, for an osmotic delivery device, the volume of a beneficial agent chamber comprising the beneficial agent formulation is between about 100 μl to about 1000 μl, more preferably between about 120 μl and about 500 μl, more preferably between about 150 μl and about 200 μl.
  • Typically, the osmotic delivery device is implanted within the subject, for example, subcutaneously. The device(s) can be inserted in either or both arms (e.g., in the inside, outside, or back of the upper arm) or into the abdomen. Preferred locations in the abdomen are under the abdominal skin in the area extending below the ribs and above the belt line. To provide a number of locations for insertion of one or more osmotic delivery devices within the abdomen, the abdominal wall can be divided into 4 quadrants as follows: the upper right quadrant extending 5-8 centimeters below the right ribs and about 5-8 centimeters to the right of the midline, the lower right quadrant extending 5-8 centimeters above the belt line and 5-8 centimeters to the right of the midline, the upper left quadrant extending 5-8 centimeters below the left ribs and about 5-8 centimeters to the left of the midline, and the lower left quadrant extending 5-8 centimeters above the belt line and 5-8 centimeters to the left of the midline. This provides multiple available locations for implantation of one or more devices on one or more occasions.
  • The suspension formulation may also be delivered from a drug delivery device that is not implantable or implanted, for example, an external pump such as a peristaltic pump used for subcutaneous delivery in a hospital setting.
  • The suspension formulations of the present invention may also be used in infusion pumps, for example, the ALZET® osmotic pumps which are miniature, infusion pumps for the continuous dosing of laboratory animals (e.g., mice and rats).
  • The suspension formulations of the present invention may also be used in the form of injections to provide highly concentrated bolus doses of biologically active agents, such as the GLP-1 receptor agonists, anti-cancer agents, etc.
  • 4.0.0 ANTICANCER AGENTS
  • The GLP-1 receptor agonists, such as GLP-1(7-36)amide and exenatide, can be delivered to a patient as a single modality treatment or in combination with other beneficial agents, including anticancer agents as described below, chemotherapeutic drugs, anticancer antibodies, antisense molecules, siRNA, and the like.
  • For example, one useful combination is with a tyrosine kinase inhibitor, such as SUTENT®, NEXAVAR®, BIBF 1120, ZD1839 (gefitinib), erlotinib, TYKERB™, and the like.
  • mTOR inhibitors, such as rapamycin (sirolimus), AZD8055, NVP-BEZ235, deforolimus, everolimus, temsirolimus, GSK1059615, WYE354, KU0063794, XL765 (all available from Selleck Chemicals) will also find use in a combination treatment.
  • Other drugs for use in combination with the GLP-1 receptor agonists (e.g., exenatide and GLP-1(7-36)amide), are those that cause hypoxia in tumor tissues, such as metformin, and drugs that inhibit the hypoxia inducible factor 1 such as CCAA/enhancer binding protein a, PX-478, resveratrol, and the various small molecule inhibitors described in Jones et al., Mol. Cancer. Ther. (2006) 5:2193-2202.
  • Also useful are drugs that inhibit IGF-1, such as octreonide acetate and tyrosine kinase inhibitors, that serve to block IGF-1 receptor signaling.
  • VEGF-inhibitors, such as anti-VEGF antibodies including bevacizumab) (AVASTIN®, as well as prolactin, sunitinib and sorafenib, may also be used in combination with the GLP-1 receptor agonists.
  • Another useful combination therapy is the use of a sugar analog, such as 2DG, subsequent to reducing glucose availability to the cancer cells using GLP-1 receptor agonists, such as exenatide and GLP-1(7-36)amide.
  • Cell cycle blockers will also find use herein, such as a cyclin-dependent kinase (cdk)-inhibitor, e.g., olomoucin, butyrolactone-I, n-butyrate, upregulators of cdk activity, e.g., flavopiridol, Chalcones (1,3-diphenylpropen-1-ones) and derivatives thereof.
  • The histone deacetylase (HDAC) enzyme SIRT-1 and other related sirtuin proteins, analogs and derivatives thereof will also find use herein.
  • Also useful are peptides that induce cell apoptosis, such TRAIL, antagonists or antibodies against integrin αvβ3, anti-survivin antibodies and antagonists of survivin, and numerous pro-apoptotic peptides, well known in the art, such as described in Ellerby et al., Nat. Med. (1999) 5:1032-1038.
  • Examples of cytokines which can be administered in a combination treatment include G-CSF, GM-CSF, M-CSF, IL-1α, IL-1β, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-10, IL-12, IL-18, IL-21, IL-23, IFN-α, IFN-β, IFN-γ, IFN-λ, MIP-1α, MIP-1β, TGF-β, TNFα, and TNF-β.
  • Examples of chemokines which can be administered include BCA-1/BLC, BRAK, Chemokine CC-2, CTACK, CXCL-16, ELC, ENA, ENA-70, ENA-74, ENA-78, Eotaxin, Exodus-2, Fractalkine, GCP-2, GRO, GRO alpha (MGSA), GRO-beta, GRO-gamma, HCC-1, HCC-4, 1-309, IP-10, 1-TAC, LAG-1, LD78-beta, LEC/NCC-4, LL-37, Lymphotactin, MCP, MCAF (MCP-1), MCP-2, MCP-3, MCP-4, MDC, MDC, MDC-2, MDC-4, MEC/CCL28, MIG, MIP, MIP-1 alpha, MIP-1 beta, MIP-1 delta, MIP-3/MPIF-1, MIP-3 alpha, MIP-3 bet, MIP-4 (PARC), MIP-5, NAP-2, PARC PF-4, RANTES, RANTES-2, SDF-1 alpha, SDF-1 beta, TARC, and TECK.
  • Examples of growth factors which can be delivered include Human Amphiregulin, Human Angiogenesis Proteins, Human ACE, Human Angiogenin, Human Angiopoietin, Human Angiostatin, Human Endostatin, Human Betacellulin, Human BMP, Human BMP-13/CDMP-2, Human BMP-14/CDMP-1, Human BMP-2, Human BMP-3, Human BMP-4, Human BMP-5, Human BMP-6, Human BMP-7, Human BMP-8, Human BMP-9, Human Colony Stimulating Factors, Human flt3-Ligand, Human G-CSF, Human GM-CSF, Human M-CSF, Human Connective Tissue Growth Factor, Human Cripto-1, Human Cryptic, Human ECGF, Human EGF, Human EG-VEGF, Human Erythropoietin, Human Fetuin, Human FGF, Human FGF-1, Human FGF-10, Human FGF-16, Human FGF-17, Human FGF-18, Human FGF-19, Human FGF-2, Human FGF-20, Human FGF-3, Human FGF-4, Human FGF-5, Human FGF-6, Human FGF-7/KGF, Human FGF-8, Human FGF-9, Human FGF-acidic, Human FGF-basic, Human GDF-11, Human GDF-15, Human Growth Hormone Releasing Factor, Human HB-EGF, Human Heregulin, Human HGF, Human IGF, Human IGF-1, Human IGF-11, Human Inhibin, Human KGF, Human LCGF, Human LIF, Human Miscellaneous Growth Factors, Human MSP, Human Myostatin, Human Myostatin Propeptide, Human Nerve Growth Factor, Human Oncostatin M, Human PD-ECGF, Human PDGF, Human PDGF (AA Homodimer), Human PDGF (AB Heterodimer), Human PDGF (BB Homodimer), Human PDGF (CC Homodimer), Human PLGF, Human PLGF-1, Human PLGF-2, Human SCF, Human SMDF, Human Stem Cell Growth Factor, Human SCGF-alpha, Human SCGF-beta, Human Thrombopoietin, Human Transforming Growth Factor, Human TGF-alpha, and Human TGF-beta.
  • In some embodiments, chemotherapeutic agents used in the methods of the invention are selected from antimetabolites; enzyme inhibitors including topoisomerase I and II inhibitors, tyrosine and serine/threonine kinase inhibitors and COX2 inhibitors, tubulin binders, proteasome inhibitors, anticancer alkylating agents including bifunctional and monofunctional alkylating agents and methylating agents, anticancer antibiotics, anticancer antibodies and active fragments and fusions thereof and antibody-drug conjugates, bisphosphonates, antiestrogens and antiandrogens, anticancer cytokines, anticancer enzymes, immunomodulatory agents, anticancer peptides, anticancer retinoids, anticancer steroids and related agents, anticancer phototherapeutics, normal tissue protectors and antihormonal agents including aromatase inhibitors.
  • Antimetabolites may include folate analogs, which inhibit dihydrofolate reductase resulting in DNA breaks by blocking purine and thymidylate synthesis. Examples of folate analogs include methotrexate (FOLEX™), trimetrexate (NEUTREXIN®) and pemetrexed (ALIMTA®). Other anitmetabolites are nucleoside analogs that disrupt DNA or RNA synthesis, such as purine or pyrimidine analogs. Examples of purine analogs include allopurinol (ZYLOPRIM®), mercaptopurine (PURINETHOL®), fludarabine (FLUDARA™), thioguanine (6-TG), cladribine (LEUSTATIN®, 2-CdA), and pentostatin (NIPENT®). Examples of pyrimidine analogs include capecitabine (XELODA®), cytarabine (CYTOSAR™), liposomal cytarabine (DEPOCYT®), floxuridine (FUDR™), fluororouracil (ADRUCIL®), gemcitabine (GEMZAR®), and clofarabine (CLOLAR®), decitabine (DACOGEN®) and azacitadine (VIDAZA®).
  • Topoisomerase II inhibitors bind to topoisomerase II and DNA, preventing the resealing of DNA strands during replication, and leading to DNA strand breaks, such as epipodophyllotoxins. Examples of epipodophyllotoxins include etoposide (VEPESID®, ETOPOPHOS®) and teniposide (VUMON®, VM26™). Alternatively, topoisomerase II inhibitors, such as anthracycline antibiotics, intercalate between DNA base pairs leading to free radicals and also topoisomerase II inhibition. Examples of anthracyclines include daunorubicin (DANOIJXOME®, CERUBIDINE™), liposomal daunorubicin (DAUNOXOME®), doxorubicin (ADRIAMYCIN™, RUBEX™), liposomal doxorubicin (DOXIL™), epirubicin (ELLENCE™), valrubicin (VALSTAR®), and idarubicin (IDAMYCIN™). Mitoxantrone (NOVANTRONE®) also inhibits topoisomerase II and is an anticancer therapeutic.
  • Topoisomerase I inhibitors bind to topoisomerase I and DNA, preventing DNA strand breaks, such as, e.g., camptothecins, including irinotecan (CAMPTOSAR®) and topotecan (HYCAMTIN®).
  • Anticancer kinase inhibitors inhibit phosphorylation of a protein or small molecule messenger in a an intracellular signaling pathway in malignant cells or vascular or stromal cells, such as, e.g., imatinib mseylate (GLEEVEC®), gefitinib (IRESSA®) or erlotinib (TARCEVA®), sorafenib (NEXAVAR®), sunitinib (SUTENT®), nilotinib (TASIGN®), everolimus (AFINITOR®), lapatinib (TYKERB®), dasatinib (SPRYCEL®), BRAF inhibitors such as GSK218436 (GlaxoSmithKline, London UK) and vemurafenib (Plexxikon Inc., CA) and MEK inhibitors.
  • Tubulin binders include agents that bind to microtubules, shift the microtubules toward polymerization, and are active in the M phase, such as taxanes including docetaxel (TAXOTERE®) and paclitaxel (TAXOL®) and epothilones including ixabepilone (IXEMPRA®) and eribulin mesylate. Other tubulin binders act by inhibiting polymerization and mitotic spindle formation, and are active in the S phase, such as, e.g., vinca alkaloids, including vinblastine (VELBAN®), vincristine (ONCOVIN™), and vinorelbine (NAVELBINE®). Other tubulin binders include ILX-651 (TASIDOTIN™) and estramustine (EMCYT®), which inhibit microtubule assembly and disassembly.
  • Proteasome inhibitors block the trypsin-like, chymotrypsin-like and/or peptidylglutamyl peptide hydrolyzing-like protease activities in nuclear and cytoplasmic proteasomes. Examples of proteasome inhibitors include bortezomib (VELCADE®).
  • Anticancer alkylating agents are reactive molecules that bind to DNA and interfere with DNA replication. These agents include, but are not limited to, alkyl sulfonates such as busulfan (MYLERAN®), platinum analogs such as carboplatin (PARAPLATIN®), cisplatin (PLATINOL®-AQ, and oxaliplatin (ELOXATIN®), nitrosoureas such as carmustine (BICNU®), lomustine (CCNU™, CEENU®), and streptozocin (ZANOSAle), nitrogen mustards including chlorambucil (LEUKERAN®), uracil mustard, cyclophosphamide (CYTOXAN®), ifosfamide (IFEX®), meclorethamine (MUSTARGEN®), and melphalan (ALKERAN®, L-PAM), bendamustine (TREANDA®), triazenes such as dacarbazine (DTIC-DOME®), procarbazine (MATULANE®), temozolomide (TEMODAR®), ethylenimines including hexamethylamine (HEXALEN®), and thiotepa (THIOPLEX®), hydroxyurea (HYDREA®, arsenic trioxide (TRISENOX®), mitomycin C (MUTAMYCIN®, MITOZYTREX™) and trabectedin (YONDELIS®).
  • Anticancer antibiotics act by a variety of mechanisms including inhibition of protein synthesis generation of oxygen free radicals in the vicinity of DNA and other mechanisms. Examples of anticancer antibiotics include actinomycin D (COSMEGEN®), bleomycin sulfate (BLENOXANE®) and plicamycin (MITHRACIN™).
  • Anticancer antibodies bind to specific molecular targets on cells or in the extracellular space. Anticancer antibodies act by neutralizing the activity of the target, attracting immune cells to the target cell or by being directly or indirectly cytotoxic toward the target cell. Anticancer antibodies include, but are not limited to, anti-CD52 antibodies such as alemtuzumab (CAMPATH®); anti-VEGF antibodies including bevacizumab (AVASTIN®); anti-CD33 antibodies, including gemtuzumab ozogamicin (MYLOTARG®); anti-CD20 antibodies including ibritumomab (ZEVALIN™), rituximab (RITUXAN™), tositumomab (BEXXAR®) and ofatumumab (ARZERRA®); anti-EGFR antibodies such as cetuximab (ERBITUX®) and panitumumab (VECTIBEX®); anti-Her2 antibodies, including trastuzumab (HERCEPTIN®); anti-CTLA4 antibodies including Ipilimumab (YERVOY™); adnectins; and domain antibodies. Active fragments and fusions of these antibodies will also find use herein.
  • Anticancer cytokines include, but are not limited to, aldesleukin (PROLEUKIN®), denileukin diftitox (ONTAK®), GM-CSF (sargramostim, PROKINE™, LEUKINE™), interferon alfa-2b (INTRON®-A), PEGinterferon alpha (PEGASYS® or PEGINTRON®) and consensus interferon (INFERGEN®).
  • Immunomodulatory agents are effective by increasing the response of the immune system of the host to the malignancy. Immunomodulatory agents include, but are not limited to, Bacillus Calmette-Gurerin (BCG Vaccine), levamisole (ERGAMISOL™), thalidomide (THALIDOMID®), sipuleucel-T (PROVENGE®), and lenalidomide (REVLIMID®).
  • Anticancer retinoids include, but are not limited to, aliretinoin (PANRETIN®), bexarotene (TARGRETIN®) and tretinoin (VESANOID®, ATRA™); other agents include octreotide acetate (SANDOSTATIN®).
  • Anticancer enzymes include asparaginase (ELSPAR®), pegademase (ADAGEN®), and pegaspargase (ONCASPAR®).
  • Anticancer steroids and related agents include dexamethasone (DECADRON™), predisone (DELTASONE®), prednisolone (DELTA-CORTEF™) and mitotane (LYSODREN®).
  • Normal tissue protectors include, but are not limited to, amifostine (ETHYOL®), darbepoetin alfa (ARANESP®), dexrazoxane (ZINECARD®), epoetin alfa (EPOGEN®, PROCRIT®), filgrastim (NEUPOGEN®), folinic acid (leucovorin), allopurinol (ALOPRIM®) mesna (MESNEX®), oprelvekin (NEUMEGA®), pegfilgrastim (NEULASTA®), GM-CSF (sargramostim, PROKINE™, LEUKINE®), raloxifene (EVISTA®) and eltrombopag (PROMACTA®).
  • Phototherapeutics are agents that sensitize cells so that exposure to a specific frequency of laser light induces abundant free radical formation and DNA alkylation. These agents include, but are not limited to, porfimer sodium (PHOTOFRIN®).
  • Antihormones include LHRH agonists, which compete with gonadotropin by binding to the hypothalamus causing an initial surge of LH and FSH followed by down regulation by negative feedback, including goserelin (ZOLADEX®), leuprolide (LUPRON® or ELIGARD®), and triptorelin (TRELSTAR®); and antiandrogens, which competitively bind and inhibit the binding of androgens to androgen receptors, such as hicalutamide (CASODEX®), flutamide (EULEXIN™), nilutamide (NILANDRON®), aminoglutethimide (CYTADREN®), and abarelix (PLENAXIS®); and antiestrogens, which competitively bind and inhibit the binding of estrogens to estrogen receptors such as tamoxifen (NOLVADEX®), fluoxymesterone (HALOTESTIN®) and megestrol (MEGACE®), bisphosphonates including pamidronate (AREDIA®) and zoledronate (ZOMETA®), and aromatase inhibitors including anastrozole (ARIMIDEX®), exemestane (AROMASIN®), fulvestrant (FASLODEX®), and letrozole (FEMARA®), androgen biosynthesis inhibitors such as abiraterone acetate (ZITIGA®), androgen signaling inhibitor such as MDV 3100.
  • ATP-competitive inhibitors of c-Met/HGF receptor and/or the nucleophosmin-anaplastic lymphoma kinase (NPM-ALK) include crizotinib, CH5424802 (Chugai Pharmaceutical Co., Ltd., Japan), and AP26113 (ARIAD Pharmaceuticals, Inc., MA).
  • Exemplary agents including beneficial agents and anticancer agents that can be delivered with the GLP-1 receptor agonist compositions described herein include those described above and/or shown in Table 1.
  • TABLE 1
    Antimetabolites
    Folate Anatagonists
    Methotrexate (FOLEX ™)
    Trimetrexate (NEUTREXIN ®)
    Pemetrexed (ALIMTA ®)
    Purine Analogs
    Allopurinol (ZYLOPRIM ®)
    Mercaptopurine (PURINETHOL ®)
    Fludarabine (FLUDARA ™)
    Thioguanine (6-TG)
    Cladribine (LEUSTATIN ®)
    Pentostatin (NIPENT ®)
    Pyrimidine Analogs
    Capecitabine (XELODA ®)
    Cytarabine (CYTOSAR ™)
    Liposomal cytarabine (DEPOCYT ®)
    Floxuridine (FUDR ™)
    Fluorouracil (ADRUCIL ®)
    Gemcitabine (GEMZAR ®)
    Clofarabine (CLOLAR ®)
    Decitabine (DACOGEN ®)
    Azacitadine (VIDAZA ®)
    Enzyme Inhibitors
    COX-2 Inhibitors (CELEBREX ®)
    Topoisomerase II Inhibitors
    Epipodophyllotoxins
    Etoposide (VEPESID ®, ETOPOPHOS ®)
    Teniposide (VUMON ®, VM 26 ™)
    Anthracyclines
    Daunorubicin (CERUBIDINE ™)
    Liposomal Daunorubicin (DAUNOXOME ®)
    Doxorubicin (ADRIAMYCIN ™, RUBEX ™)
    Liposomal Doxorubicin (DOXIL ™)
    Epirubicin (ELLENCE ®)
    Valrubicin (VALSTAR ®)
    Idarubicin (IDAMYCIN ™)
    Mitoxantrone (NOVANTRONE ®)
    Topoisomerase I Inhibitors
    Camptothecins
    Irinotecan (CAMPTOSAR ®)
    Topotecan (HYCAMTIN ®)
    Anticancer Kinase Inhibitors
    Imatinib mesylate (GLEEVEC ®)
    Gefitinib (IRESSA ®)
    Erlotinib (TARCEVA ®)
    Sorafenib (NEXAVAR ®)
    Sunitinib (SUTENT ®)
    Nilotinib (TASIGNA ®)
    Everolimus (AFINITOR ®)
    Lapatinib (TYKERB ®)
    Dasatinib (SPRYCEL ®)
    Antitubulins
    Taxanes
    Docetaxel (TAXOTERE ®)
    Paclitaxel (TAXOL ®)
    Ixabepilone (IXEMPRA ®)
    Cabazitaxel (JEVTANA ®)
    Vinca Alkaloids
    Vinblastine (VELBAN ®)
    Vincristine (ONCOVIN ™)
    Vinorelbine (NAVELBINE ®)
    Vinflunine (JAVLOR ®)
    ILX-651 (TASIDOTIN ™)
    Tasidotin-C-carboxylate
    Estramustine (EMCYT ®)
    Anticancer Phototherapeutics
    Porfimer Sodium (PHOTOFRIN ®)
    Anticancer Antibodies
    Anti-CD52 Antibodies
    Alemtuzumab (CAMPATH ®)
    Anti-CD33 Antibodies
    Gemtuzumab ozogamicin (MYLOTARG ®)
    Anti-CD20 Antibodies
    Ibritumomab (ZEVALIN ®)
    Rituximab (RITUXAN ®)
    Tositumomab (BEXXAR ®)
    Ofatumumab (ARZERRA ®)
    Anti-Her2 Antibodies
    Trastuzumab (HERCEPTIN ®)
    Anti-VEGF
    Bevacizumab (AVASTIN ®)
    Anti-EGFR
    Cetuximab (ERBITUX ®)
    Anticancer Retinoids
    Alitretinoin (PANRETIN ®)
    Bexarotene (TARGRETIN ®)
    Tretinoin (VESANOID ®, ATRA ™)
    Octreotide acetate (SANDOSTATIN ®)
    Normal Tissue Protectors
    Amifostine (ETHYOL ®)
    Darbepoetin alfa (ARANESP ®)
    Dexrazoxane (ZINECARD ®)
    Epoetin alfa (EPOGEN ®, PROCRIT ®)
    Filgrastim (NEUPOGEN ®)
    Folinic Acid (leucovorin)
    Allopurinol (ALOPRIM ®)
    Mesna (MESNEX ®)
    Oprelvekin (rhIL-11) (NEUMEGA ®)
    Pegfilgrastim (NEULASTA ®)
    GM-CSF (sargramostim, PROKINE ™, LEUKINE ®)
    Eltrombopag (PROMACTA ®)
    AMD3100 (plerixafor, MOZOBIL ®)
    Alkylating Agents
    Alkyl Sulfonates
    Busulfan (MYLERAN ®)
    Platinum Analogs
    Carboplatin (PARAPLATIN ®)
    Cisplatin (PLATINOL ®-AQ)
    Oxaliplatin (ELOXATIN ®)
    Nitrosoureas
    Carmustine (BICNU ®)
    Lomustine (CCNU ™, CEENU ®)
    Streptozocin (ZANOSAR ®)
    Nitrogen Mustards
    Chlorambucil (LEUKERAN ®)
    Uracil mustard
    Cyclophosphamide (CYTOXAN ®)
    Ifosfamide (IFEX ®)
    Meclorethamine (MUSTARGEN ®)
    Melphalan (ALKERAN ®, L-PAM)
    Bendamustine (TREANDA ®)
    Triazenes
    Dacarbazine (DTIC-DOME ®)
    Procarbazine (MATULANE ®)
    Temozolomide (TEMODAR ®)
    Ethylenimines
    Hexamethylamine (HEXALEN ®, altretamine, HEXASTAT ™)
    Thiotepa (THIOPLEX ®, TESPA ™)
    Hydroxyurea (HYDREA ®)
    Arsenic trioxide (TRISENOX ®)
    Mitomycin C (MUTAMYCIN ®)
    Trabectedin (YONDELIS ®)
    Anticancer Antibiotics
    Actinomycin D (dactinomycin, COSMEGEN ®)
    Bleomycin sulfate (BLENOXANE ®)
    Plicamycin (MITHRACIN ™)
    Proteasome Inhibitors
    Bortezomib (VELCADE ®)
    Anticancer Anti-hormones
    LHRH Agonists
    Histrelin (VANTAS ®)
    Goserelin (ZOLADEX ®)
    Leuprolide (LUPRON ®, ELIGARD ®)
    Triptorelin (TRELSTAR ®)
    Anti-Androgens
    Bicalutamide (CASODEX ®)
    Flutamide (EULEXIN ™)
    Nilutamide (NILANDRON ®)
    Aminoglutethimide (CYTADREN ®)
    Abarelix (PLENAXIS ®)
    Anti-Estrogens and Aromatase Inhibitors
    Tamoxifen (NOLVADEX ®)
    Raloxifene (EVISTA ®)
    Anastrozole (ARIMIDEX ®)
    Exemestane (AROMASIN ®)
    Fulvestrant (FASLODEX ®)
    Letrozole (FEMARA ®)
    Fluoxymesterone (HALOTESTIN ®)
    Megestrol acetate (MEGACE ®)
    Bisphosphonates
    Pamidronate (AREDIA ®)
    Zoledronate (ZOMETA ®)
    Ibandronate (BONIVA ®)
    Anticancer Enzymes
    Asparaginase (ELSPAR ®)
    Pegademase (ADAGEN ®)
    Pegaspargase (ONCASPAR ®)
    Anticancer Cytokines
    Aldesleukin (rhIL-2) (PROLEUKIN ®)
    Denileukin Diftitox (ONTAK ®)
    Interferon alfa-2b (INTRON ® A)
    Peginterferon alfa-2a (PEGASYS ®)
  • Treatment will depend on the cancer in question. Tests can be performed prior to treatment to specifically tailor a treatment for a patient. Such tests may include genetic or protein marker testing of tumor markers to determine susceptibility or resistance to a particular drug or class of drugs. For example, recently a mutation in von Hippel-Landau (VHL) gene have been found to be associated with a more favorable drug response for drugs such as SUTENT®, NEXAVAR®, and AVASTIN®. Other genetic and protein tests can be performed to link a treatment to an appropriate patient population.
  • The agents described above can be provided in formulations obtained from the manufacturer. Such formulations typically include the active components mixed with a pharmaceutically acceptable vehicle or excipient. The vehicle may contain minor amounts of auxiliary substances such as wetting or emulsifying agents. The formulations may also include ancillary substances, such as pharmacological agents, cytokines, or other biological response modifiers.
  • In other embodiments of the invention, the pharmaceutical composition comprising the agent is a sustained-release formulation, and/or a formulation that is administered using a sustained-release device. Such devices are well known in the art, and include, for example, transdermal patches, and miniature implantable pumps (such as described herein) that can provide for drug delivery over time in a continuous, steady-state fashion at a variety of doses to achieve a sustained-release effect with either a non-sustained-release or a sustained release pharmaceutical composition. For example, polypeptide agents and antibodies described herein are suitable agents for delivery using an osmotic delivery device such as the DUROS® implantable device described above. In this embodiment, two or more such implantable delivery devices can be used, one including the GLP-1 receptor agonist and one or more including one or more additional beneficial agents, such as anticancer polypeptide formulations, antibodies, and the like. See, e.g., U.S. Patent Publication 2009/0202608, incorporated herein by reference in its entirety, for a description of the use of two or more implantable delivery devices.
  • The additional beneficial agents may also be formulated as particle and suspension formulations as described herein, if appropriate. Such particle and suspension formulations are useful with polypeptide agents and antibodies and can be delivered using implantable devices as described above. In addition to the suspension formulations, comprising a suspension vehicle and particle formulation, described above, some polypeptide agents (e.g., leuprolide acetate) can be directly dissolved or dispersed in a vehicle for delivery from implantable devices. For example, some polypeptides (e.g., leuprolide acetate) can be dissolved in non-aqueous polar aprotic solvents (e.g., dimethylsulfoxide) to provide peptide formulations (see, e.g., U.S. Pat. Nos. 5,932,547; 6,235,712; 5,981,489, incorporated herein by reference in their entireties). The use of one such formulation in an implantable osmotic delivery device is described below in Example 5. Other examples of peptide formulations include, but are not limited to, non-aqueous protic peptide formulations (see, e.g., U.S. Pat. No. 6,066,619, incorporated herein by reference in its entirety) and aqueous formulations of peptides (see, e.g., U.S. Pat. No. 6,068,850, incorporated herein by reference in its entirety).
  • Other suitable routes of administration for the beneficial agents include parenteral administration, such as subcutaneous (s.c.), intraperitoneal (i.p.), intramuscular (i.m.), intravenous (i.v.), or infusion, oral (p.o.) and pulmonary, nasal, topical, transdermal, and suppositories. Where the composition is administered via pulmonary delivery, the therapeutically effective dose is adjusted such that the soluble level of the agent in the bloodstream, is equivalent to that obtained with a therapeutically effective dose that is administered parenterally, for example s.c., i.p., i.m., or i.v. In some embodiments of the invention, the pharmaceutical composition comprising the beneficial agent is administered by i.m. or s.c. injection, particularly by i.m. or s.c. injection locally to the region where the GLP-1 receptor agonist is administered.
  • One or more therapeutically effective dose of the additional beneficial agent, such as an anticancer agent will be administered. By “therapeutically effective dose or amount” of each of these agents is intended an amount that when administered in combination with the other agents, brings about a positive therapeutic response with respect to treatment of an individual with cancer. Of particular interest is an amount of these agents that provides an anti-tumor effect, as defined herein. In certain embodiments, multiple therapeutically effective doses of the additional beneficial agent will be provided.
  • The additional beneficial agents can be administered prior to, concurrent with, or subsequent to administration of the GLP-1 receptor agonist. For example, initial treatment with a chemotherapeutic agent can be performed, followed by implantation of a delivery device including the GLP-1 receptor agonist formulation or vice versa. Moreover, the additional beneficial agent may be administered over the time that the GLP-1 receptor agonist formulation is also being delivered. By “concurrent therapy” is intended administration to a subject such that the therapeutic effect of the combination of the substances is caused in the subject undergoing therapy.
  • 5.0.0 USES
  • The GLP-1 receptor agonists, e.g., exenatide and GLP-1(7-36)amide, optionally in combination with other beneficial agents, can be used to treat various cancers. In particular, as explained above, cancer cells are known to exhibit increased glycolysis as compared to normal cells. An advantage of the present invention is that inhibiting glucose availability to cancer cells by using a GLP-1 receptor agonist, such as exenatide and GLP-1(7-36)amide, effectively reduces the amount of energy metabolites such as ATP and NADH produced, thereby starving the cancer cell of energy.
  • Any number of cancers can benefit from the delivery of GLP-1 receptor agonists. For example, tumors or cancers such as hemangiomas, neufibromatosis, breast, colorectal, lung, brain and CNS, renal, gynecological (e.g. ovarian, fallopian, cervical, peritoneal), hematological (lymphoma, multiple myeloma, leukemia), neuroendocrine, mesothelioma, melanoma, prostate, esophagus, liver, gastric, rectal, carcinoid tumors; head and neck, squamous cell carcinoma, sarcomas, pancreas, colon, thymoma, thyroid, small intestine, bladder, testicular, bile duct, gall bladder, kidney, gastrointestinal stromal tumors, endometrial cancers and choriocarcinoma. A list of cancers that may benefit from delivery of the GLP-1 receptor agonists is shown in Table 2.
  • TABLE 2
    Acute Lymphoblastic Leukemia, Adult
    Acute Lymphoblastic Leukemia, Childhood
    Acute Myeloid Leukemia, Adult
    Acute Myeloid Leukemia, Childhood
    Adrenocortical Carcinoma
    Adrenocortical Carcinoma, Childhood
    AIDS-Related Cancers
    AIDS-Related Lymphoma
    Anal Cancer
    Appendix Cancer
    Atypical Teratoid/Rhabdoid Tumor, Childhood, Central Nervous System
    Basal Cell Carcinoma, see Skin Cancer (Non-melanoma)
    Bladder Cancer
    Bladder Cancer, Childhood
    Bone Cancer, Osteosarcoma and Malignant Fibrous Histiocytoma
    Brain Stem Glioma, Childhood
    Brain Tumor, Adult
    Brain Tumor, Brain Stem Glioma, Childhood
    Brain Tumor, Central Nervous System Atypical Teratoid/Rhabdoid
    Tumor, Childhood
    Brain Tumor, Central Nervous System Embryonal Tumors, Childhood
    Brain Tumor, Cerebellar Astrocytoma, Childhood
    Brain Tumor, Cerebral Astrocytoma/Malignant Glioma, Childhood
    Brain Tumor, Craniopharyngioma, Childhood
    Brain Tumor, Ependymoblastoma, Childhood
    Brain Tumor, Ependymoma, Childhood
    Brain Tumor, Medulloblastoma, Childhood
    Brain Tumor, Medulloepithelioma, Childhood
    Brain Tumor, Pineal Parenchymal Tumors of Intermediate
    Differentiation, Childhood
    Brain Tumor, Supratentorial Primitive Neuroectodermal Tumors and
    Pineoblastoma, Childhood
    Brain Tumor, Visual Pathway and Hypothalamic Glioma, Childhood
    Brain and Spinal Cord Tumors, Childhood (Other)
    Breast Cancer
    Breast Cancer and Pregnancy
    Breast Cancer, Childhood
    Breast Cancer, Male
    Bronchial Tumors, Childhood
    Burkitt Lymphoma
    Carcinoid Tumor, Childhood
    Carcinoid Tumor, Gastrointestinal
    Carcinoma of Unknown Primary
    Central Nervous System Embryonal Tumors, Childhood
    Central Nervous System Lymphoma, Primary
    Cerebral Astrocytoma/Malignant Glioma, Childhood
    Cervical Cancer
    Cervical Cancer, Childhood
    Childhood Cancers
    Chordoma, Childhood
    Chronic Lymphocytic Leukemia
    Chronic Myelogenous Leukemia
    Chronic Myeloproliferative Disorders
    Colon Cancer
    Colorectal Cancer, Childhood
    Cutaneous T-Cell Lymphoma, see Mycosis Fungoides and Sézary
    Syndrome
    Ependymoma, Childhood
    Esophageal Cancer
    Esophageal Cancer, Childhood
    Ewing Family of Tumors
    Extracranial Germ Cell Tumor, Childhood
    Extragonadal Germ Cell Tumor
    Extrahepatic Bile Duct Cancer
    Eye Cancer, Intraocular Melanoma
    Eye Cancer, Retinoblastoma
    Gallbladder Cancer
    Gastrointestinal Carcinoid Tumor
    Gastrointestinal Stromal Tumor (GIST)
    Gastrointestinal Stromal Cell Tumor, Childhood
    Germ Cell Tumor, Extracranial, Childhood
    Germ Cell Tumor, Extragonadal
    Germ Cell Tumor, Ovarian
    Gestational Trophoblastic Tumor
    Glioma, Adult
    Glioma, Childhood Brain Stem
    Glioma, Childhood Cerebral Astrocytoma
    Hairy Cell Leukemia
    Head and Neck Cancer
    Hepatocellular (Liver) Cancer, Adult (Primary)
    Hepatocellular (Liver) Cancer, Childhood (Primary)
    Histiocytosis, Langerhans Cell
    Hodgkin Lymphoma, Adult
    Hodgkin Lymphoma, Childhood
    Hypopharyngeal Cancer
    Hypothalamic and Visual Pathway Glioma, Childhood
    Islet Cell Tumors (Endocrine Pancreas)
    Kaposi Sarcoma
    Kidney (Renal Cell) Cancer
    Kidney Cancer, Childhood
    Laryngeal Cancer
    Laryngeal Cancer, Childhood
    Lip and Oral Cavity Cancer
    Liver Cancer, Adult (Primary)
    Liver Cancer, Childhood (Primary)
    Malignant Fibrous Histiocytoma of Bone and Osteosarcoma
    Mesothelioma, Adult Malignant
    Mesothelioma, Childhood
    Metastatic Squamous Neck Cancer with Occult Primary
    Mouth Cancer
    Multiple Endocrine Neoplasia Syndrome, Childhood
    Multiple Myeloma/Plasma Cell Neoplasm
    Mycosis Fungoides
    Myelodysplastic Syndromes
    Myelodysplastic/Myeloproliferative Diseases
    Nasal Cavity and Paranasal Sinus Cancer
    Nasopharyngeal Cancer
    Nasopharyngeal Cancer, Childhood
    Neuroblastoma
    Non-Hodgkin Lymphoma, Adult
    Non-Hodgkin Lymphoma, Childhood
    Non-Small Cell Lung Cancer
    Oral Cancer, Childhood
    Oral Cavity Cancer, Lip tongue and mouth
    Oropharyngeal Cancer
    Ovarian Cancer, Childhood
    Ovarian Epithelial Cancer
    Ovarian Germ Cell Tumor
    Ovarian Low Malignant Potential Tumor
    Pancreatic Cancer
    Pancreatic Cancer, Childhood
    Pancreatic Cancer, Islet Cell Tumors
    Papillomatosis, Childhood
    Parathyroid Cancer
    Penile Cancer
    Pharyngeal Cancer
    Pheochromocytoma
    Pineoblastoma and Supratentorial Primitive Neuroectodermal Tumors,
    Childhood
    Pituitary Tumor
    Pleuropulmonary Blastoma
    Prostate Cancer
    Rectal Cancer
    Respiratory Tract Carcinoma Involving the NUT Gene
    on Chromosome 15
    Rhabdomyosarcoma, Childhood
    Salivary Gland Cancer
    Salivary Gland Cancer, Childhood
    Sarcoma, Ewing Family of Tumors
    Sézary Syndrome
    Skin Cancer (Non-melanoma)
    Skin Cancer, Childhood
    Skin Cancer (Melanoma)
    Skin Carcinoma, Merkel Cell
    Small Cell Lung Cancer
    Small Intestine Cancer
    Soft Tissue Sarcoma, Adult
    Soft Tissue Sarcoma, Childhood
    Squamous Neck Cancer with Occult Primary, Metastatic
    Stomach (Gastric) Cancer
    Stomach (Gastric) Cancer, Childhood
    Testicular Cancer
    Throat Cancer
    Thymoma and Thymic Carcinoma
    Thymoma and Thymic Carcinoma, Childhood
    Thyroid Cancer
    Thyroid Cancer, Childhood
    Transitional Cell Cancer of the Renal Pelvis and Ureter
    Trophoblastic Tumor, Gestational
    Unusual Cancers of Childhood
    Ureter and Renal Pelvis, Transitional Cell Cancer
    Urethral Cancer
    Uterine Cancer, Endometrial
    Uterine Sarcoma
    Vaginal Cancer
    Vaginal Cancer, Childhood
    Vulvar Cancer
    Waldenström Macroglobulinemia
    Wilms Tumor
  • In some embodiments, the GLP-1 receptor agonists, are used in the treatment of hematological tumors and/or solid tumors. In a preferred embodiment, the GLP-1 receptor agonists, for example, exenatide and GLP-1(7-36)amide, are used in the treatment of solid tumors.
  • The GLP-1 receptor agonists are delivered in order to provide a positive therapeutic response. By “positive therapeutic response” it is intended the individual undergoing the combination treatment of a GLP-1 receptor agonist, such as exenatide and GLP-1(7-36)amide, and an additional beneficial agent exhibits an improvement in one or more symptoms of the cancer for which the individual is undergoing therapy. Therefore, for example, a positive therapeutic response refers to one or more of the following improvements in the disease: (1) reduction in tumor size; (2) reduction in the number of cancer cells; (3) inhibition (i.e., slowing to some extent, preferably halting) of tumor growth; (4) inhibition (i.e., slowing to some extent, preferably halting) of cancer cell infiltration into peripheral organs; (5) inhibition (i.e., slowing to some extent, preferably halting) of tumor metastasis; and (6) some extent of relief from one or more symptoms associated with the cancer. Such therapeutic responses may be further characterized as to degree of improvement. Thus, for example, an improvement may be characterized as a complete response. By “complete response” is documentation of the disappearance of all symptoms and signs of all measurable or evaluable disease confirmed by physical examination, laboratory, nuclear and radiographic studies (i.e., CT (computer tomography) and/or MRI (magnetic resonance imaging)), and other non-invasive procedures repeated for all initial abnormalities or sites positive at the time of entry into the study. Alternatively, an improvement in the disease may be categorized as stabilization of the disease or may be a partial response. By “partial response” is intended a reduction of greater than 50% in the sum of the products of the perpendicular diameters of one or more measurable lesions when compared with pretreatment measurements (for patients with evaluable response only, partial response does not apply).
  • In one embodiment, the GLP-1 receptor agonist is delivered in a suspension formulation, administered using an osmotic delivery device as described above. Examples of target rates of delivery for suspension formulations of the present invention, comprising GLP-1 receptor agonists, include, but are not limited to: suspension formulations comprising particle formulations comprising GLP-1 (e.g., GLP-1(7-36)amide), between about 20 μg/day and about 900 μg/day, preferably between about 100 μg/day and about 600 μg/day, for example, at about 480 μg/day; and suspension formulations comprising particle formulations comprising exenatide, between about 5 μg/day and about 320 μg/day, preferably between about 5 μg/day and about 160 μg/day, for example, at about 10 μg/day to about 20 μg/day, such as 10, 20, 40, 60, 80, 100, 120 μg/day, or any integers between the above ranges. An exit sheer rate of the suspension formulation from the osmotic delivery device is determined such that the target daily target delivery rate of the GLP-1 receptor agonist is reasonably achieved by substantially continuous, uniform delivery of the suspension formulation from the osmotic delivery device. Examples of exit sheer rates include, but are not limited to, about 1 to about 1×104 reciprocal second, preferably about 4×10−2 to about 6×104 reciprocal second, more preferably 5×10−3 to 1×10−3 reciprocal second.
  • As explained above, a subject being treated with the GLP-1 receptor agonist formulations of the present invention may also benefit from co-treatment with other beneficial agents, including anticancer agents described above, as well as antidiabetic agents.
  • Additional beneficial agents that can be delivered include, but are not limited to, pharmacologically beneficial peptides proteins, polypeptides, genes, gene products, other gene therapy agents, or other small molecules. The additional beneficial agents are useful for the treatment of a variety of conditions including but not limited to hemophilia and other blood disorders, growth disorders, diabetes, leukemia and lymphoma, hepatitis, renal failure, bacterial infection, viral infection (e.g., infection by HIV, HCV, etc.), hereditary diseases such as cerbrosidase deficiency and adenosine deaminase deficiency, hypertension, septic shock, autoimmune diseases (e.g., Graves disease, systemic lupus erythematosus and rheumatoid arthritis), shock and wasting disorders, cystic fibrosis, lactose intolerance, Crohn's disease, inflammatory bowel disease, Alzheimer's disease, metabolic disorders (such as obesity), and cancers.
  • The polypeptides may include but are not limited to the following: glucagon-like peptide 2 (GLP-2), cholecystokinin (CCK), CCK octapeptide, growth hormone, somatostatin; somatropin, somatotropin, somatotropin analogs, somatomedin-C, somatotropin plus an amino acid, somatotropin plus a protein; follicle stimulating hormone; luteinizing hormone, luteinizing hormone-releasing hormone (LHRH), LHRH analogs/agonists such as leuprolide, nafarelin and goserelin, LHRH antagonists; growth hormone releasing factor; calcitonin; colchicine; gonadotropins such as chorionic gonadotropin; antiandrogens such as flutamide, nilutamide and cytoprerone; aromatase inhibitors such as exemastane, letrozole and anastrazole; selective estrogen receptor modulators such as raloxifene, lasoxifene; oxytocin, octreotide; vasopressin; adrenocorticotrophic hormone; epidermal growth factor; fibroblast growth factor; platelet-derived growth factor; transforming growth factor; nerve growth factor; prolactin; cosyntropin; lypressin polypeptides such as thyrotropin releasing hormone; thyroid stimulation hormone; secretin; leptin; adiponectin; amylin, amylin analogs (e.g., pramlintide acetate); pancreozymin; enkephalin; glucagon; endocrine agents secreted internally and distributed by way of the bloodstream; carbohydrases, nucleases, lipase, proteases, amylase, or the like.
  • Further beneficial agents that may be delivered include but are not limited to the following: alpha antitrypsin; factor VII; factor IX, thrombin and other coagulation factors; insulin; peptide hormones; adrenal cortical stimulating hormone, thyroid stimulating hormone and other pituitary hormones; erythropoietin; growth factors such as granulocyte-colony stimulating factor, granulocyte-macrophage colony stimulating factor, thrombopoietin, insulin-like growth factor 1; tissue plasminogen activator; CD4; 1-deamino-8-D-arginine vasopressin; interleukin-1 receptor antagonist; tumor necrosis factor, tumor necrosis factor receptor; tumor suppresser proteins; pancreatic enzymes; lactase; cytokines, including lymphokines, chemokines or interleukins such as interleukin-1, interleukin-2 and other members of the interleukin family (e.g., IL-1, 6, 12, 15, 17, 18, 32); cytotoxic proteins; superoxide dismutase; endocrine agents secreted internally and distributed in an animal by way of the bloodstream; recombinant antibodies, antibody fragments, humanized antibodies, single chain antibodies, monoclonal antibodies; avimers; or the like.
  • Further, the beneficial agents that may be administered include, but are not limited to, organic compounds including those compounds that transport across a vessel. Examples of beneficial agents that may be used in the practice of the present invention include, but are not limited to, the following: hypnotics and sedatives such as pentobarbital sodium, phenobarbital, secobarbital, thiopental, amides and ureas exemplified by diethylisovaleramide and alpha-bromo-isovaleryl urea, urethanes, or disulfanes; heterocyclic hypnotics such as dioxopiperidines, and glutarimides; antidepressants such as isocarboxazid, nialamide, phenelzine, imipramine, tranylcypromine, pargyline; tranquilizers such as chloropromazine, promazine, fluphenazine reserpine, deserpidine, meprobamate, benzodiazepines such as chlordiazepoxide; tricyclic antidepressants; anticonvulsants such as primidone, diphenylhydantoin, ethltoin, pheneturide, ethosuximide; muscle relaxants and anti-parkinson agents such as mephenesin, methocarbomal, trihexylphenidyl, biperiden, levo-dopa, also known as L-dopa and L-beta-3-4-dihydroxyphenylalanine; analgesics such as morphine, codeine, meperidine, nalorphine; antipyretics and anti-inflammatory agents such as aspirin, salicylamide, sodium salicylamide, naproxin, ibuprofen, acetaminophen; local anesthetics such as procaine, lidocaine, naepaine, piperocaine, tetracaine, dibucane; antispasmodics and antiulcer agents such as atropine, scopolamine, methscopolamine, oxyphenonium, papaverine, prostaglandins such as PGE1, PGE2, PGF1alpha, PGF2alpha, PGA; anti-microbials such as penicillin, tetracycline, oxytetracycline, chlorotetracycline, chloramphenicol, sulfonamides, bacitracin, chlorotetracycline, levofloxacin, erythromycin; anti-fungals such as Amphotericin B; anti-malarials such as 4-aminoquinolines, 8-aminoquinolines and pyrimethamine; hormonal agents such as prednisolone, cortisone, cortisol and triamcinolone, androgenic steroids (for example, methyltestosterone, fluoxmesterone), estrogenic steroids (for example, 17-beta-estradiol and ethinyl estradiol), progestational steroids (for example, 17-alpha-hydroxyprogesterone acetate, 19-nor-progesterone, norethindrone); sympathomimetic drugs such as epinephrine, amphetamine, ephedrine, norepinephrine; cardiovascular drugs such as procainamide, amyl nitrate, nitroglycerin, dipyridamole, sodium nitrate, mannitol nitrate; diuretics such as acetazolamide, chlorothiazide, flumethiazide; antiparasitic agents such as bephenium hydroxynaphthoate, dichlorophen, enitabas, dapsone; anti-neoplastic agents such as mechloroethamine, uracil mustard, 5-fluorouracil, 6-thioguanine, procarbazine, paclitaxel, docetaxel, carboplatin, gemcitabine, oxaliplatin, fludarabine, ara-C, camptothecin, bortezomib, methrotrexate, capecitabine, doxorubicin, vincristine, cyclophosphamide, etoposide; VEGF/EGF inhibitors (for example, small molecules and antibodies); VEGF/EGF receptor inhibitors; hypoglycemic drugs such as insulin related compounds (for example, isophane insulin suspension, protamine zinc insulin suspension, globin zinc insulin, extended insulin zinc suspension) tolbutamide, acetohexamide, tolazamide, chlorpropamide; nutritional agents such as vitamins, essential amino acids, and essential fats; eye drugs such as pilocarpine base, pilocarpine hydrochloride, pilocarpine nitrate; antiviral drugs such as disoproxil fumarate, aciclovir, cidofovir, docosanol, famciclovir, fomivirsen, foscarnet, ganciclovir, idoxuridine, penciclovir, trifluridine, tromantadine, valaciclovir, valganciclovir, vidarabine, amantadine, arbidol, oseltamivir, peramivir, rimantadine, zanamivir, abacavir, didanosine, emtricitabine, lamivudine, stavudine, zalcitabine, zidovudine, tenofovir, efavirenz, delavirdine, nevirapine, loviride, amprenavir, atazanavir, darunavir, fosamprenavir, indinavir, lopinavir, nelfinavir, ritonavir, saquinavir, tipranavir, enfuvirtide, adefovir, fomivirsen, imiquimod, inosine, podophyllotoxin, ribavirin, viramidine, fusion inhibitors specifically targeting viral surface proteins or viral receptors (for example, gp-41 inhibitor (T-20), CCR-5 inhibitor, enfuvirtide (FUZEON®)); anti-nausea (such as scopolamine, dimenhydrinate, granisetron, dolasetron, palonesetron, metaclopramide, ondansetron); iodoxuridine, hydrocortisone, eserine, phospholine, iodide, as well as other beneficial agents.
  • Numerous peptides, proteins, or polypeptides that are useful in the practice of the present invention are described herein. In addition to the peptides, proteins, or polypeptides described, modifications of these peptides, proteins, or polypeptides are also known to one of skill in the art and can be used in the practice of the present invention following the guidance presented herein. Such modifications include, but are not limited to, amino acid analogs, amino acid mimetics, analog polypeptides, or derivative polypeptides. Further, the beneficial agents disclosed herein may be formulated singly or in combination (e.g., mixtures).
  • Peptide YY (PYY) inhibits gut motility and blood flow (Laburthe, M., Trends Endocrinol Metab. 1(3):168-74 (1990), mediates intestinal secretion (Cox, H. M., et al., Br J Pharmacol 101(2):247-52 (1990); Playford, R. J., et al., Lancet 335(8705):1555-7 (1990)), stimulate net absorption (MacFayden, R. J., et al., Neuropeptides 7(3):219-27 (1986)), and two major in vivo variants (PYY and PYY3-36) have been identified (e.g., Eberlein, G. A., et al., Peptides 10 (4), 797-803 (1989)). The sequence of PYY, as well as analogs and derivatives thereof, including PYY3-36, are known in the art (e.g., U.S. Pat. Nos. 5,574,010 and 5,552,520). For ease of reference herein, the family of PYY polypeptides, PYY derivatives, variants and analogs are referred to collectively as PYY.
  • GIP is an insulinotropic peptide hormone (Efendic, S., Horm Metab Res. (2004) 36:742-746) and is secreted by the mucosa of the duodenum and jejunum in response to absorbed fat and carbohydrate that stimulate the pancreas to secrete insulin. GIP stimulates insulin secretion from pancreatic beta cells in the presence of glucose (Tseng et al., PATAS (1993) 90:1992-1996). GIP circulates as a biologically active 42-amino acid peptide. GIP is also known as glucose-dependent insulinotropic protein. The sequence of GIP, as well as peptide analogs and peptide derivatives thereof, are known in the art (see, e.g., Meier J. J., Diabetes Metab Res Rev. (2005) 21(2):91-117; Efendic S., Horm Metab Res. (2004) 36(11-12):742-746). For ease of reference herein, the family of GIP polypeptides, GIP derivatives, variants and analogs are referred to collectively as GIP.
  • Oxyntomodulin is a naturally occurring 37 amino acid peptide hormone found in the colon that has been found to suppress appetite and facilitate weight loss (Wynne K, et al., Int J Obes (Lond) 30(12):1729-36 (2006)). The sequence of oxyntomodulin, as well as analogs and derivatives thereof, are known in the art (e.g., U.S. Patent Publication Nos. 2005-0070469 and 2006-0094652). For ease of reference herein, the family of oxyntomodulin polypeptides, oxyntomodulin derivatives, variants and analogs are referred to collectively as oxyntomodulin.
  • Amylin, as well as analogs and derivatives thereof: are known in the art (e.g., U.S. Pat. Nos. 5,686,411, 5,814,600, 5,998,367, 6,114,304, 6,410,511, 6,608,029, and 6,610,824). For ease of reference herein, the family of amylin polypeptides, amylin derivatives, variants and analogs are referred to collectively as amylin.
  • The cDNA sequence encoding the human leptin protein hormone is known (e.g., Masuzaki, H., et al. (Diabetes 44: 855-858, 1995)). Leptin, as well as analogs and derivatives thereof, are known in the art (e.g., U.S. Pat. Nos. 5,521,283, 5,525,705, 5,532,336, 5,552,522, 5,552,523, 5,552,524, 5,554,727, 5,559,208, 5,563,243, 5,563,244, 5,563,245, 5,567,678, 5,567,803, 5,569,743, 5,569,744, 5,574,133, 5,580,954, 5,594,101, 5,594,104, 5,605,886, 5,691,309, and 5,719,266; P.C.T. International Patent Publication Nos. WO96/22308, WO96/31526, WO96/34885, 97/46585, WO97/16550, and WO 97/20933; European Patent Publication No. EP 0 741 187). For ease of reference herein, the family of leptin polypeptides, leptin derivatives, variants and analogs are referred to collectively as leptin.
  • Further, oligonucleotides (e.g., RNA, DNA, alternative backbones) may be used as beneficial agents in the practice of the present invention. In one embodiment therapeutic RNA molecules may include, but are not limited to, small nuclear RNAs (snRNAs), and small interfering RNA strands (siRNA) for use in RNA interference (RNAi) inhibition of gene expression. RNAi inhibition typically occurs at the stage of translation or by hindering the transcription of specific genes. RNAi targets include, but are not limited to, RNA from viruses and genes with roles in regulating development and genome maintenance.
  • The beneficial agents can also be in various forms including, but not limited to, the following: uncharged molecules; components of molecular complexes; and pharmacologically acceptable salts such as hydrochloride, hydrobromide, sulfate, laurates, palmatates, phosphate, nitrate, borate, acetate, maleate, tartrate, oleates, or salicylates. For acidic drugs, salts of metals, amines or organic cations, for example, quaternary ammonium, can be employed. Furthermore, simple derivatives of the drug such as esters, ethers, amides and the like that have solubility characteristics suitable for the purpose of the invention can also be used herein. The formulation used can have been in various art known forms such as solution, dispersion, paste, cream, particle, granule, tablet, emulsions, suspensions, powders and the like. In addition to the one or more beneficial agents, the beneficial agent formulation may optionally include pharmaceutically acceptable carriers and/or additional ingredients such as antioxidants, stabilizing agents, buffers, and permeation enhancers.
  • The amount of beneficial agent used is that amount necessary to deliver a therapeutically effective amount of the agent to achieve the desired therapeutic result. In practice, this will vary depending upon such variables, for example, as the particular agent, the site of delivery, the severity of the condition, and the desired therapeutic effect. Beneficial agents and their dosage unit amounts are known to the prior art in Goodman & Gilman's The Pharmacological Basis of Therapeutics, 11th Ed., (2005), McGraw Hill; Remington's Pharmaceutical Sciences, 18th Ed., (1995), Mack Publishing Co.; and Martin's Physical Pharmacy and Pharmaceutical Sciences, 1.00 edition (2005), Lippincott Williams & Wilkins.
  • The additional beneficial agent can be delivered using any of the various delivery techniques outlined above, including without limitation parenterally (including by subcutaneous, intravenous, intramedullary, intraarticular, intramuscular, or intraperitoneal injection) rectally, topically, transdermally, intranasally, by inhalation, or orally (for example, in capsules, suspensions, or tablets). In certain embodiments, the agent is in a sustained-release formulation, or administered using a sustained-release device. Such devices are well known in the art, and include, for example, transdermal patches, and miniature implantable pumps (such as the DUROS® delivery device described herein) that can provide for drug delivery over time in a continuous, steady-state fashion at a variety of doses to achieve a sustained-release effect with a non-sustained-release pharmaceutical composition. If an osmotic delivery device is used, the volume of a beneficial agent chamber comprising the beneficial agent formulation is between about 50 μl to about 1000 μl, more preferably between about 100 μl and about 500 μl, more preferably between about 150 μl and about 200 μl. Moreover, two or more such devices can be used, one including the GLP-1 receptor agonist and one or more including one or more additional beneficial agents, such as an antidiabetic compound. See, e.g., U.S. Patent Publication 2009/0202608, incorporated herein by reference in its entirety, for a description of the use of two or more implantable delivery devices.
  • An example of a cancer treatment using delivery of an anticancer agent from a first osmotic delivery device and delivery of a GLP-1 receptor agonist from a second osmotic delivery device is presented below in Example 5. In the example, the cancer is prostate cancer, the anticancer agent is leuprolide acetate and the GLP-1 receptor agonist is exenatide.
  • Other objects may be apparent to one of ordinary skill upon reviewing the following specification and claims.
  • 6.0.0 EXPERIMENTAL
  • The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the devices, methods, and formulae of the present invention, and are not intended to limit the scope of what the inventor regards as the invention. Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperature, etc.) but some experimental errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, molecular weight is weight average molecular weight, temperature is in degrees Centigrade, and pressure is at or near atmospheric.
  • The compositions produced according to the present invention meet the specifications for content and purity required of pharmaceutical products.
  • Example 1 Exenatide Particle Formulations
  • This example describes making exenatide particle formulations.
  • A. Formulation 1
  • Exenatide (0.25 g) was dissolved in 50 mM sodium citrate buffer at pH 6.04. The solution was dialyzed with a formulation solution containing sodium citrate buffer, sucrose, and methionine. The formulated solution was then spray dried using Buchi 290 with 0.7 mm nozzle, outlet temperature of 75° C., atomization pressure of 100 Psi, solid content of 2%, and flow rate of 2.8 mL/min. The dry powder contained 21.5% of exenatide with 4.7% residual moisture and 0.228 g/ml density.
  • B. Formulations 2 and 3
  • Two additional formulations of exenatide were prepared essentially by the method just described. Following here in Table 3 is a summary of the weight percentages (wt %) of the components of the Formulations 1, 2 and 3.
  • TABLE 3
    Particle Particle Particle
    Formulation 1 Formulation 2 Formulation 3
    Component (wt %) (wt %) (wt %)
    Exenatide 21.5 11.2 50.0
    Sodium Citrate* 63.6 74.7 28.4
    Citric Acid* 7.1 9.1 3.6
    Sucrose 3.9 2.5 9.0
    Methionine 3.9 2.5 9.0
    *Sodium Citrate/Citric Acid formed the citrate buffer in the pre-spray drying process for preparation of this particle formulation.
  • Example 2 GLP-1 (7-36)amide Dry Powder
  • This example describes making a GLP-1(7-36)amide particle formulation. GLP-1(7-36)amide (1.5 g) was dissolved in 5 mM sodium citrate buffer at pH 4. The solution was dialyzed with a formulation solution containing sodium citrate buffer and methionine. The formulated solution was then spray dried using Buchi 290 with 0.7 mm nozzle, outlet temperature of 70° C., atomization pressure of 100 Psi, solid content of 1.5%, and flow rate of 5 mL/min. The dry powder contained 90% of GLP-1(7-36)amide.
  • Example 3 Exenatide Suspension Formulation
  • This example describes making suspension formulations comprising a suspension vehicle and an exenatide particle formulation.
  • A. Suspension Formulation of 20 wt % Exenatide Particles
  • An exenatide particle formulation was generated by spray-drying, and contained 20 wt % exenatide, 32 wt % sucrose, 16 wt % methionine and 32 wt % citrate buffer.
  • A suspension vehicle was formed by dissolving the polymer polyvinylpyrrolidone in the solvent benzyl benzoate at approximately a 50/50 ratio by weight. The vehicle viscosity was approximately 12,000 to 18,000 poise when measured at 33° C. Particles containing the peptide exenatide were dispersed throughout the vehicle at a concentration of 10% particles by weight.
  • B. Suspension Formulations of Particle Formulations 1, 2, and 3
  • A suspension vehicle was formed by dissolving the polymer polyvinylpyrrolidone K-17 (typically having an approximate average molecular weight range of 7,900-10,800) in the solvent benzyl benzoate heated to approximately 65° C. under a dry atmosphere and reduced pressure at approximately a 50/50 ratio by weight. The vehicle viscosity was approximately 12,000 to 18,000 poise when measured at 33° C. Particle formulations 1-3, described in Example 1, were dispersed throughout the vehicle at the concentrations (by weight percent) shown in Table 4.
  • TABLE 4
    Suspension Suspension Suspension
    Formulation 1 Formulation 2 Formulation 3
    Component (wt %) (wt %) (wt %)
    Particle Formulation 1 21.40
    Particle Formulation 2 11.73
    Particle Formulation 3 10.05
    Polyvinylpyrrolidone 39.30 44.13 44.98
    Benzyl Benzoate 39.30 44.13 44.98
  • Example 4 GLP-1(7-36)amide Formulation
  • This example describes making a suspension formulation comprising a suspension vehicle and an GLP-1(7-36)amide particle formulation. A GLP-1(7-36)amide particle formulation was generated by spray-drying, and contained 90 wt % GLP-1, 5 wt % methionine and 5 wt % citrate buffer.
  • A suspension vehicle containing the polymer polyvinylpyrrolidone was dissolved in the solvent benzyl benzoate at approximately a 50/50 ratio by weight. The vehicle viscosity was approximately 12,000 to 18,000 poise when measured at 33° C. Particles containing the peptide GLP-1(7-36)amide were dispersed throughout the vehicle at a concentration of 33% particles by weight.
  • Example 5 Co-Treatment of Prostate Cancer using Leuprolide Acetate and Exenatide
  • Leuprolide acetate, an LHRH agonist, acts as a potent inhibitor of gonadotropin secretion when given continuously and in therapeutic doses. Animal and human studies indicate that following an initial stimulation, chronic administration of leuprolide acetate results in suppression of testicular steroidogenesis. This effect is reversible upon discontinuation of drug therapy. Administration of leuprolide acetate has resulted in inhibition of the growth of certain hormone-dependent tumors (prostatic tumors in Noble and Dunning male rats and DMBA-induced mammary tumors in female rats) as well as atrophy of the reproductive organs. In humans, administration of leuprolide acetate results in an initial increase in circulating levels of luteinizing hormone (LH) and follicle stimulating hormone (FSH), leading to a transient increase in levels of the gonadal steroids (testosterone and dihydrotestosterone in males). However, continuous administration of leuprolide acetate results in decreased level of LH and FSH. In males, testosterone is reduced to castrate levels. These decreases occur within two to six weeks after initiation of treatment, and castrate levels of testosterone in prostatic cancer patients have been demonstrated for multiyear periods. Leuprolide acetate is not active when given orally.
  • An implantable device containing leuprolide acetate for the treatment of prostate cancer is assembled as described in U.S. Pat. No. 5,728,396, incorporated herein by reference in its entirety. The device includes the following components:
  • Reservoir (Titanium, Ti6A 14V alloy) (4 mm outside diameter, 3 mm inside diameter)
  • Piston (C-Flex®)
  • Lubricant (silicone medical fluid)
    Compressed osmotic engine (76.4% NaCl, 15.5% sodium carboxymethyl cellulose, 6% povidone, 0.5% Mg Stearate, 1.6% water)
    PEG 400 (8 mg added to osmotic engine to fill air spaces)
    Membrane plug (polyurethane polymer, injection molded to desired shape)
    Back diffusion Regulating Outlet (polyethylene)
    Drug formulation (1) 0.150 g of 60% water and 40% leuprolide acetate; or (2) leuprolide acetate dissolved in DMSO to a measured content of 65 mg leuprolide.
  • To assemble the device, the piston and inner diameter of the reservoir are lightly lubricated. The piston is inserted about 0.5 cm into the reservoir at the membrane end. PEG 400 is added into the reservoir. Two osmotic engine tablets (40 mg each) are then inserted into the reservoir from the membrane end. After insertion, the osmotic engine is flush with the end of the reservoir. The membrane plug is inserted by lining up the plug with the reservoir and pushing gently until the retaining features of the plug are fully engaged in the reservoir. Formulation is loaded into a syringe which is then used to fill the reservoir from the outlet end by injecting formulation into the open tube until the formulation is about 3 mm from the end. The filled reservoir is centrifuged (outlet end “up”) to remove any air bubbles that have been trapped in the formulation during filling. The outlet is screwed into the open end of the reservoir until completely engaged. As the outlet is screwed in, excess formulation exits out of the orifice ensuring a uniform fill.
  • These devices deliver about 0.35 μL/day leuprolide formulation containing on average 150 μg leuprolide in the amount delivered per day. They provide delivery of leuprolide at this rate for at least one year. The devices can achieve approximately 70% steady-state delivery by day 14.
  • Exenatide suspension formulations are produced as described in Example 1 and loaded into an implantable delivery device as above. Two implantable devices, one including an exenatide formulation and one including a leuprolide formulation are implanted under local anesthetic and by means of an incision in a patient suffering from advanced prostatic cancer. Implantation can be accomplished using, for example, an implanter device. See e.g., U.S. Pat. No. 6,190,350, incorporated herein by reference in its entirety. After an appropriate period of time, the implantable delivery devices are removed under local anesthetic. New devices may be inserted at that time.
  • 7.0.0 FURTHER EXEMPLARY EMBODIMENTS OF THE PRESENT INVENTION
  • Embodiments of the present invention include, but are not limited to, the following:
  • 1. A method of treating cancer in a subject in need of such treatment, comprising: administering a GLP-1 receptor agonist to said subject.
  • 2. The method of embodiment 1, wherein the GLP-1 receptor agonist is a small molecule.
  • 3. The method of embodiment 1, wherein the GLP-1 receptor agonist is a peptide, polypeptide or protein.
  • 4. The method of embodiment 3, wherein the GLP-1 receptor agonist is a glucagon-like peptide-1 (GLP-1), a derivative of GLP-1, or an analog of GLP-1.
  • 5. The method of embodiment 4, wherein the GLP-1 receptor agonist is GLP(7-36)amide comprising the sequence of SEQ ID NO:1.
  • 6. The method of embodiment 3, wherein the GLP-1 receptor agonist is exenatide, a derivative of exenatide, or an analog of exenatide.
  • 7. The method of embodiment 6, wherein the GLP-1 receptor agonist is synthetic exenatide peptide comprising the sequence of SEQ ID NO:2.
  • 8. The method of embodiment 4, wherein the GLP-1 receptor agonist is selected from the group consisting of liraglutide, albiglutide, semaglutide and taspoglutide.
  • 9. The method of embodiment 6, wherein the GLP-1 receptor agonist is lixisenatide.
  • 10. The method of any one of embodiments 1-9, wherein the GLP-1 receptor agonist is provided in a suspension formulation comprising: (a) a particle formulation comprising said GLP-1 receptor agonist; and (b) a vehicle formulation, wherein the particle formulation is dispersed in the vehicle.
  • 11. The method of embodiment 10, wherein (a) the particle formulation additionally comprises a disaccharide, methionine and a buffer and (b) the vehicle formulation is a non-aqueous, single-phase suspension vehicle comprising one or more pyrrolidone polymers and one or more solvents selected from the group consisting of lauryl lactate, lauryl alcohol, benzyl benzoate, and mixtures thereof; wherein the suspension vehicle exhibits viscous fluid characteristics, and the particle formulation is dispersed in the vehicle.
  • 12. The method of embodiment 11, wherein the buffer is selected from the group consisting of citrate, histidine, succinate, and mixtures thereof.
  • 13. The method of embodiment 12, wherein the buffer is citrate.
  • 14. The method of embodiment 11, wherein the disaccharide is selected from the group consisting of lactose, sucrose, trehalose, cellobiose, and mixtures thereof.
  • 15. The method of embodiment 11, wherein the particle formulation is a spray dried preparation of particles.
  • 16. The method of embodiment 11, wherein the solvent is selected from the group consisting of lauryl lactate, benzyl benzoate, and mixtures thereof.
  • 17. The method of embodiment 16, wherein the solvent consists essentially of benzyl benzoate.
  • 18. The method of embodiment 11, wherein the pyrrolidone polymer consists essentially of polyvinylpyrrolidone.
  • 19. The method of embodiment 11, wherein the vehicle consists essentially of a pyrrolidone polymer and benzyl benzoate.
  • 20. The method of embodiment 19, wherein the vehicle is about 50% solvent and about 50% polymer.
  • 21. The method of embodiment 11, wherein the suspension formulation has an overall moisture content of less than or equal to about 10 wt %.
  • 22. The method of any one of embodiments 1-21, wherein the GLP-1 receptor agonist is delivered using an implantable osmotic delivery device.
  • 23. The method of embodiment 22, wherein the osmotic delivery device provides continuous delivery of the GLP-1 receptor agonist for a period of at least one month.
  • 24. The method of any one of embodiments 1-9, wherein the GLP-1 receptor agonist is provided in an injectable formulation.
  • 25. The method of any one of embodiments 1-24, wherein a beneficial agent in addition to the GLP-1 receptor agonist is delivered to said subject.
  • 26. The method of embodiment 25, wherein the additional beneficial agent is an anticancer agent.
  • 27. The method of embodiment 26, wherein the anticancer agent is a chemotherapeutic agent.
  • 28. The method of embodiment 26, wherein the anticancer agent is an anticancer antibody.
  • 29. The method of any one of embodiments 25-28, wherein the additional beneficial agent is an antidiabetic agent.
  • 30. The method of any one of embodiments 25-29, wherein the additional beneficial agent is delivered using an implantable osmotic delivery device.
  • 31. The method of embodiment 30, wherein the osmotic delivery device provides continuous delivery of the GLP-1 receptor agonist for a period of at least one month.
  • 32. The method of either one of embodiments 30 or 31, wherein the additional beneficial agent is a luteinizing hormone-releasing hormone (LHRH) agonist.
  • 33. The method of any one of embodiments 25-29, wherein the additional beneficial agent is provided in an injectable formulation.
  • 34. The method of any one of embodiments 25-29, wherein the additional beneficial agent is provided in an oral formulation.
  • 35. The method of embodiment 25, wherein the additional beneficial agent is GIP.
  • 36. The method of any one of embodiments 25-35, wherein the additional beneficial agent is delivered prior to the GLP-1 receptor agonist.
  • 37. The method of any one of embodiments 25-35, wherein the additional beneficial agent is delivered subsequent to the GLP-1 receptor agonist.
  • 38. The method of any one of embodiments 25-35, wherein the additional beneficial agent is delivered concurrent with the GLP-1 receptor agonist.
  • As is apparent to one of skill in the art, various modification and variations of the above embodiments can be made without departing from the spirit and scope of this invention. Such modifications and variations are within the scope of this invention.

Claims (20)

1. A method of treating cancer in a subject in need of such treatment, comprising administering a GLP-1 receptor agonist to the subject.
2. The method of claim 1, wherein the GLP-1 receptor agonist is a peptide, polypeptide or protein.
3. The method of claim 2, wherein the GLP-1 receptor agonist is a glucagon-like peptide-1 (GLP-1), a derivative of GLP-1, or an analog of GLP-1.
4. The method of claim 3, wherein the GLP-1 receptor agonist is GLP(7-36)amide comprising the sequence of SEQ ID NO:1.
5. The method of claim 3, wherein the GLP-1 receptor agonist is selected from the group consisting of liraglutide, albiglutide, semaglutide and taspoglutide.
6. The method of claim 2, wherein the GLP-1 receptor agonist is exenatide, a derivative of exenatide, or an analog of exenatide.
7. The method of claim 6, wherein the GLP-1 receptor agonist is synthetic exenatide peptide comprising the sequence of SEQ ID NO:2.
8. The method of claim 6, wherein the GLP-1 receptor agonist is lixisenatide.
9. The method of claim 1, wherein the GLP-1 receptor agonist is provided in a suspension formulation comprising:
(a) a particle formulation comprising the GLP-1 receptor agonist; and
(b) a vehicle formulation,
wherein the particle formulation is dispersed in the vehicle.
10. The method of claim 9, wherein (a) the particle formulation additionally comprises a disaccharide, methionine and a buffer and (b) the vehicle formulation is a non-aqueous, single-phase suspension vehicle comprising one or more pyrrolidone polymers and one or more solvents selected from the group consisting of lauryl lactate, lauryl alcohol, benzyl benzoate, and mixtures thereof;
wherein the suspension vehicle exhibits viscous fluid characteristics, and the particle formulation is dispersed in the vehicle.
11. The method of claim 10, wherein the buffer is selected from the group consisting of citrate, histidine, succinate, and mixtures thereof.
12. The method of claim 10, wherein the disaccharide is selected from the group consisting of lactose, sucrose, trehalose, cellobiose, and mixtures thereof.
13. The method of claim 10, wherein the particle formulation is a spray dried preparation of particles.
14. The method of claim 10, wherein the vehicle consists essentially of polyvinylpyrrolidone and benzyl benzoate.
15. The method of claim 14, wherein the vehicle is about 50% solvent and about 50% polymer.
16. The method of claim 10, wherein the suspension formulation has an overall moisture content of less than or equal to about 10 wt %.
17. The method of claim 9, wherein the CLP-1 receptor agonist is delivered using an implantable osmotic delivery device.
18. The method of claim 1, wherein the GLP-1 receptor agonist is provided in an injectable formulation.
19. The method of claim 1, wherein one or more beneficial agent in addition to the GLP-1 receptor agonist is delivered to the subject, and the beneficial agent is an anticancer agent, a chemotherapeutic agent, or an antidiabetic agent.
20. The method of claim 19, wherein the additional beneficial agent is delivered using an implantable osmotic delivery device.
US13/372,326 2011-02-16 2012-02-13 Compositions, Devices and Methods of Use Thereof for the Treatment of Cancers Abandoned US20120208755A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US13/372,326 US20120208755A1 (en) 2011-02-16 2012-02-13 Compositions, Devices and Methods of Use Thereof for the Treatment of Cancers
US14/525,201 US20150057227A1 (en) 2011-02-16 2014-10-27 Compositions, devices and methods of use thereof for the treatment of cancers
US15/597,788 US10159714B2 (en) 2011-02-16 2017-05-17 Compositions, devices and methods of use thereof for the treatment of cancers

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161443628P 2011-02-16 2011-02-16
US13/372,326 US20120208755A1 (en) 2011-02-16 2012-02-13 Compositions, Devices and Methods of Use Thereof for the Treatment of Cancers

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/525,201 Continuation US20150057227A1 (en) 2011-02-16 2014-10-27 Compositions, devices and methods of use thereof for the treatment of cancers

Publications (1)

Publication Number Publication Date
US20120208755A1 true US20120208755A1 (en) 2012-08-16

Family

ID=46637352

Family Applications (3)

Application Number Title Priority Date Filing Date
US13/372,326 Abandoned US20120208755A1 (en) 2011-02-16 2012-02-13 Compositions, Devices and Methods of Use Thereof for the Treatment of Cancers
US14/525,201 Abandoned US20150057227A1 (en) 2011-02-16 2014-10-27 Compositions, devices and methods of use thereof for the treatment of cancers
US15/597,788 Active US10159714B2 (en) 2011-02-16 2017-05-17 Compositions, devices and methods of use thereof for the treatment of cancers

Family Applications After (2)

Application Number Title Priority Date Filing Date
US14/525,201 Abandoned US20150057227A1 (en) 2011-02-16 2014-10-27 Compositions, devices and methods of use thereof for the treatment of cancers
US15/597,788 Active US10159714B2 (en) 2011-02-16 2017-05-17 Compositions, devices and methods of use thereof for the treatment of cancers

Country Status (4)

Country Link
US (3) US20120208755A1 (en)
EP (1) EP2675469A4 (en)
CA (1) CA2823721C (en)
WO (1) WO2012112626A2 (en)

Cited By (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015093854A1 (en) * 2013-12-17 2015-06-25 주식회사 카엘젬백스 Composition for treating prostate cancer
WO2016061583A1 (en) * 2014-10-17 2016-04-21 University Of Virginia Patent Foundation Compositions and methods for treating pituitary tumors
US9526763B2 (en) 2005-02-03 2016-12-27 Intarcia Therapeutics Inc. Solvent/polymer solutions as suspension vehicles
US9540419B2 (en) 2012-05-11 2017-01-10 Gemvax & Kael Co., Ltd. Anti-inflammatory peptides and composition comprising the same
US9539200B2 (en) 2005-02-03 2017-01-10 Intarcia Therapeutics Inc. Two-piece, internal-channel osmotic delivery system flow modulator
US9572889B2 (en) 2008-02-13 2017-02-21 Intarcia Therapeutics, Inc. Devices, formulations, and methods for delivery of multiple beneficial agents
US9670261B2 (en) 2012-12-21 2017-06-06 Sanofi Functionalized exendin-4 derivatives
US9682127B2 (en) 2005-02-03 2017-06-20 Intarcia Therapeutics, Inc. Osmotic delivery device comprising an insulinotropic peptide and uses thereof
US9694053B2 (en) 2013-12-13 2017-07-04 Sanofi Dual GLP-1/glucagon receptor agonists
US9724293B2 (en) 2003-11-17 2017-08-08 Intarcia Therapeutics, Inc. Methods of manufacturing viscous liquid pharmaceutical formulations
US9730984B2 (en) 2012-05-11 2017-08-15 Gemvax & Kael Co., Ltd. Composition for preventing or treating rheumatoid arthritis
US9751926B2 (en) 2013-12-13 2017-09-05 Sanofi Dual GLP-1/GIP receptor agonists
US9750788B2 (en) 2013-12-13 2017-09-05 Sanofi Non-acylated exendin-4 peptide analogues
US9758561B2 (en) 2014-04-07 2017-09-12 Sanofi Dual GLP-1/glucagon receptor agonists derived from exendin-4
US9771406B2 (en) 2014-04-07 2017-09-26 Sanofi Peptidic dual GLP-1/glucagon receptor agonists derived from exendin-4
US9775904B2 (en) 2014-04-07 2017-10-03 Sanofi Exendin-4 derivatives as peptidic dual GLP-1/glucagon receptor agonists
US9789165B2 (en) 2013-12-13 2017-10-17 Sanofi Exendin-4 peptide analogues as dual GLP-1/GIP receptor agonists
EP3160491A4 (en) * 2014-06-25 2018-01-17 GlaxoSmithKline LLC Pharmaceutical compositions
US9889085B1 (en) 2014-09-30 2018-02-13 Intarcia Therapeutics, Inc. Therapeutic methods for the treatment of diabetes and related conditions for patients with high baseline HbA1c
US9907838B2 (en) 2013-04-19 2018-03-06 Gemvax & Kael Co., Ltd. Composition and methods for treating ischemic damage
US9932381B2 (en) 2014-06-18 2018-04-03 Sanofi Exendin-4 derivatives as selective glucagon receptor agonists
US9937240B2 (en) 2014-04-11 2018-04-10 Gemvax & Kael Co., Ltd. Peptide having fibrosis inhibitory activity and composition containing same
CN108030997A (en) * 2016-08-03 2018-05-15 尼尔·希夫拉杰·戴维 Adj sp medicine delivery implantable device
CN108066762A (en) * 2016-11-07 2018-05-25 赫兰 The purposes of GLP-1 receptor stimulating agents and biguanides
US9982029B2 (en) 2015-07-10 2018-05-29 Sanofi Exendin-4 derivatives as selective peptidic dual GLP-1/glucagon receptor agonists
US10034922B2 (en) 2013-11-22 2018-07-31 Gemvax & Kael Co., Ltd. Peptide having angiogenesis inhibitory activity and composition containing same
WO2018165462A1 (en) * 2017-03-08 2018-09-13 Intarcia Therapeutics, Inc Apparatus and methods for administration of a nauseogenic compound from a drug delivery device
USD835783S1 (en) 2016-06-02 2018-12-11 Intarcia Therapeutics, Inc. Implant placement guide
US10159714B2 (en) 2011-02-16 2018-12-25 Intarcia Therapeutics, Inc. Compositions, devices and methods of use thereof for the treatment of cancers
US10231923B2 (en) 2009-09-28 2019-03-19 Intarcia Therapeutics, Inc. Rapid establishment and/or termination of substantial steady-state drug delivery
US10383926B2 (en) 2013-06-07 2019-08-20 Gemvax & Kael Co., Ltd. Biological markers useful in cancer immunotherapy
USD860451S1 (en) 2016-06-02 2019-09-17 Intarcia Therapeutics, Inc. Implant removal tool
US10463708B2 (en) 2014-12-23 2019-11-05 Gemvax & Kael Co., Ltd. Peptide for treating ocular diseases and composition for treating ocular diseases comprising same
US10501517B2 (en) 2016-05-16 2019-12-10 Intarcia Therapeutics, Inc. Glucagon-receptor selective polypeptides and methods of use thereof
US10527170B2 (en) 2006-08-09 2020-01-07 Intarcia Therapeutics, Inc. Osmotic delivery systems and piston assemblies for use therein
US10561703B2 (en) 2013-06-21 2020-02-18 Gemvax & Kael Co., Ltd. Method of modulating sex hormone levels using a sex hormone secretion modulator
US10662223B2 (en) 2014-04-30 2020-05-26 Gemvax & Kael Co., Ltd. Composition for organ, tissue, or cell transplantation, kit, and transplantation method
US10758592B2 (en) 2012-10-09 2020-09-01 Sanofi Exendin-4 derivatives as dual GLP1/glucagon agonists
US10806797B2 (en) 2015-06-05 2020-10-20 Sanofi Prodrugs comprising an GLP-1/glucagon dual agonist linker hyaluronic acid conjugate
US10835582B2 (en) 2015-02-27 2020-11-17 Gemvax & Kael Co. Ltd. Peptide for preventing hearing loss, and composition comprising same
US10835580B2 (en) 2017-01-03 2020-11-17 Intarcia Therapeutics, Inc. Methods comprising continuous administration of a GLP-1 receptor agonist and co-administration of a drug
US10888605B2 (en) 2017-08-24 2021-01-12 Novo Nordisk A/S GLP-1 compositions and uses thereof
US10898540B2 (en) 2016-04-07 2021-01-26 Gem Vax & KAEL Co., Ltd. Peptide having effects of increasing telomerase activity and extending telomere, and composition containing same
US10925639B2 (en) 2015-06-03 2021-02-23 Intarcia Therapeutics, Inc. Implant placement and removal systems
US10967000B2 (en) 2012-07-11 2021-04-06 Gemvax & Kael Co., Ltd. Cell-penetrating peptide, conjugate comprising same and composition comprising same
US11015179B2 (en) 2015-07-02 2021-05-25 Gemvax & Kael Co., Ltd. Peptide having anti-viral effect and composition containing same
USD933219S1 (en) 2018-07-13 2021-10-12 Intarcia Therapeutics, Inc. Implant removal tool and assembly
US11173291B2 (en) 2020-03-20 2021-11-16 The Regents Of The University Of California Implantable drug delivery devices for localized drug delivery
US11246913B2 (en) 2005-02-03 2022-02-15 Intarcia Therapeutics, Inc. Suspension formulation comprising an insulinotropic peptide
US11318191B2 (en) 2020-02-18 2022-05-03 Novo Nordisk A/S GLP-1 compositions and uses thereof
US11338119B2 (en) * 2020-03-20 2022-05-24 The Regents Of The University Of California Implantable drug delivery devices for localized drug delivery
US11344526B2 (en) 2020-03-20 2022-05-31 The Regents Of The University Of California Implantable drug delivery devices for localized drug delivery
US11439802B2 (en) * 2018-11-19 2022-09-13 Biora Therapeutics, Inc. Ingestible device for delivery of therapeutic agent to the gastrointestinal tract

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9592407B2 (en) 2012-09-13 2017-03-14 Chang Gung Memorial Hospital Pharmaceutical composition and uses thereof
WO2016094228A1 (en) * 2014-12-07 2016-06-16 Nano Precision Medical, Inc. Implantable drug delivery device
EP3297654B1 (en) 2015-05-22 2021-07-07 The Board of Trustees of the Leland Stanford Junior University Treatment of post-bariatric hypoglycemia with exendin(9-39)
US10653753B2 (en) 2016-03-04 2020-05-19 Eiger Biopharmaceuticals, Inc. Treatment of hyperinsulinemic hypoglycemia with exendin-4 derivatives
AU2017361539B2 (en) 2016-11-21 2023-06-29 Eiger Biopharmaceuticals, Inc. Buffered formulations of exendin (9-39)
GB201714777D0 (en) * 2017-09-14 2017-11-01 Univ London Queen Mary Agent
HRP20230929T1 (en) * 2018-10-26 2023-11-24 Novo Nordisk A/S Stable semaglutide compositions and uses thereof
JP7410952B2 (en) 2018-12-21 2024-01-10 ノヴォ ノルディスク アー/エス Process of spray drying of GLP-1 peptide
WO2022034489A1 (en) * 2020-08-11 2022-02-17 Auxilla Pharmaceuticals And Research Llp A non-aqueous suspension of anticancer agent
KR20230129468A (en) * 2021-01-08 2023-09-08 스타트온 테라퓨틱스, 인크. Stable solutions of immunomodulatory imide compounds for parenteral use

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090042781A1 (en) * 2004-06-28 2009-02-12 Novo Nordisk A/S Methods for Treating Diabetes
US20090202608A1 (en) * 2008-02-13 2009-08-13 Alessi Thomas R Devices, formulations, and methods for delivery of multiple beneficial agents
WO2009109927A2 (en) * 2008-03-05 2009-09-11 Tel Hashomer Medical Research Infrastructure And Services Ltd. Glp-1 receptor agonists and related active pharmaceutical ingredients for treatment of cancer
US20100092566A1 (en) * 2008-10-15 2010-04-15 Alessi Thomas R Highly concentrated drug particles, formulations, suspensions and uses thereof

Family Cites Families (684)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR640907A (en) 1927-06-25 1928-07-24 Automatic flow limiter
US2168437A (en) 1935-04-10 1939-08-08 Kenneth O Buercklin Injection device
US2110208A (en) 1937-02-12 1938-03-08 U S Standard Products Company Antigen preparations
US2531724A (en) 1948-09-20 1950-11-28 Edmund D Cevasco Infant bath mat
US3025991A (en) 1960-05-23 1962-03-20 Carron Products Co Bottle stopper
NL280825A (en) 1962-07-11
GB1049104A (en) 1963-05-11 1966-11-23 Prodotti Antibiotici Spa Pharmaceutical compositions for oral or parenteral administration comprising tetracycline antibiotics
US3122162A (en) 1963-06-20 1964-02-25 Asa D Sands Flow control device
BE744162A (en) 1969-01-16 1970-06-15 Fuji Photo Film Co Ltd ENCAPSULATION PROCESS
US3632768A (en) 1969-10-02 1972-01-04 Upjohn Co Therapeutic composition and method for treating infections with actinospectacin
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
DE2010115A1 (en) 1970-03-04 1971-09-16 Farbenfabriken Bayer Ag, 5090 Leverkusen Process for the production of micro-granules
US3625214A (en) 1970-05-18 1971-12-07 Alza Corp Drug-delivery device
US3732865A (en) 1971-01-13 1973-05-15 Alza Corp Osmotic dispenser
US4034756A (en) 1971-01-13 1977-07-12 Alza Corporation Osmotically driven fluid dispenser
US3995631A (en) 1971-01-13 1976-12-07 Alza Corporation Osmotic dispenser with means for dispensing active agent responsive to osmotic gradient
US4211771A (en) 1971-06-01 1980-07-08 Robins Ronald K Treatment of human viral diseases with 1-B-D-ribofuranosyl-1,2,4-triazole-3-carboxamide
JPS523342B2 (en) 1972-01-26 1977-01-27
BE795516A (en) 1972-02-17 1973-08-16 Ciba Geigy PREPARATIONS OF OILY AND INJECTABLE PEPTIDES AND PROCESS FOR THEIR PREPARATION
US3797492A (en) 1972-12-27 1974-03-19 Alza Corp Device for dispensing product with directional guidance member
US3995632A (en) 1973-05-04 1976-12-07 Alza Corporation Osmotic dispenser
GB1413186A (en) 1973-06-27 1975-11-12 Toyo Jozo Kk Process for encapsulation of medicaments
DE2528516A1 (en) 1974-07-05 1976-01-22 Sandoz Ag NEW GALENIC PREPARATION
JPS523653A (en) 1975-06-27 1977-01-12 Fuji Photo Film Co Ltd Process for producing fine polymer particles
US3987790A (en) 1975-10-01 1976-10-26 Alza Corporation Osmotically driven fluid dispenser
US4008719A (en) 1976-02-02 1977-02-22 Alza Corporation Osmotic system having laminar arrangement for programming delivery of active agent
US4078060A (en) 1976-05-10 1978-03-07 Richardson-Merrell Inc. Method of inducing an estrogenic response
US4203439A (en) 1976-11-22 1980-05-20 Alza Corporation Osmotic system with volume amplifier for increasing amount of agent delivered therefrom
US4111203A (en) 1976-11-22 1978-09-05 Alza Corporation Osmotic system with means for improving delivery kinetics of system
US4111201A (en) 1976-11-22 1978-09-05 Alza Corporation Osmotic system for delivering selected beneficial agents having varying degrees of solubility
US4111202A (en) 1976-11-22 1978-09-05 Alza Corporation Osmotic system for the controlled and delivery of agent over time
USD258837S (en) 1977-10-17 1981-04-07 Gambro Dialysatoren Gmbh And Co. Kg Dialyzer cartridge
USD259458S (en) 1978-06-09 1981-06-09 Fuller Charles R Support pad for an infant
US4243030A (en) 1978-08-18 1981-01-06 Massachusetts Institute Of Technology Implantable programmed microinfusion apparatus
US4305927A (en) 1979-02-05 1981-12-15 Alza Corporation Method for the management of intraocular pressure
US4373527B1 (en) 1979-04-27 1995-06-27 Univ Johns Hopkins Implantable programmable medication infusion system
US4310516A (en) 1980-02-01 1982-01-12 Block Drug Company Inc. Cosmetic and pharmaceutical vehicle thickened with solid emulsifier
US4384975A (en) 1980-06-13 1983-05-24 Sandoz, Inc. Process for preparation of microspheres
AU546785B2 (en) 1980-07-23 1985-09-19 Commonwealth Of Australia, The Open-loop controlled infusion of diabetics
US4350271A (en) 1980-08-22 1982-09-21 Alza Corporation Water absorbing fluid dispenser
US4376118A (en) 1980-10-06 1983-03-08 Miles Laboratories, Inc. Stable nonaqueous solution of tetracycline salt
US4389330A (en) 1980-10-06 1983-06-21 Stolle Research And Development Corporation Microencapsulation process
PH19942A (en) 1980-11-18 1986-08-14 Sintex Inc Microencapsulation of water soluble polypeptides
US4340054A (en) 1980-12-29 1982-07-20 Alza Corporation Dispenser for delivering fluids and solids
US4444498A (en) 1981-02-27 1984-04-24 Bentley Laboratories Apparatus and method for measuring blood oxygen saturation
US4455145A (en) 1981-07-10 1984-06-19 Alza Corporation Dispensing device with internal drive
AU561343B2 (en) 1981-10-19 1987-05-07 Genentech Inc. Human immune interferon by recombinant dna
EP0079143A3 (en) 1981-10-20 1984-11-21 Adnovum Ag Pseudoplastic gel transfer
EP0080879B1 (en) 1981-11-28 1986-10-01 Sunstar Kabushiki Kaisha Pharmaceutical composition containing interferon in stable state
US5004689A (en) 1982-02-22 1991-04-02 Biogen, Massachusetts DNA sequences, recombinant DNA molecules and processes for producing human gamma interferon-like polypeptides in high yields
US4439196A (en) 1982-03-18 1984-03-27 Merck & Co., Inc. Osmotic drug delivery system
US4455143A (en) 1982-03-22 1984-06-19 Alza Corporation Osmotic device for dispensing two different medications
US6936694B1 (en) 1982-05-06 2005-08-30 Intermune, Inc. Manufacture and expression of large structural genes
DE3220116A1 (en) 1982-05-28 1983-12-01 Dr. Karl Thomae Gmbh, 7950 Biberach MICROBIOLOGICALLY MANUFACTURED (ALPHA) AND SS INTERFERONES, DNA SEQUENCES CODING FOR THESE INTERFERONES, MICROORGANISMS CONTAINING THIS GENETIC INFORMATION, AND METHOD FOR THE PRODUCTION THEREOF
US4530840A (en) 1982-07-29 1985-07-23 The Stolle Research And Development Corporation Injectable, long-acting microparticle formulation for the delivery of anti-inflammatory agents
US4753651A (en) 1982-08-30 1988-06-28 Alza Corporation Self-driven pump
US4966843A (en) 1982-11-01 1990-10-30 Cetus Corporation Expression of interferon genes in Chinese hamster ovary cells
US4552561A (en) 1982-12-23 1985-11-12 Alza Corporation Body mounted pump housing and pump assembly employing the same
US4673405A (en) 1983-03-04 1987-06-16 Alza Corporation Osmotic system with instant drug availability
US4639244A (en) 1983-05-03 1987-01-27 Nabil I. Rizk Implantable electrophoretic pump for ionic drugs and associated methods
US4765989A (en) 1983-05-11 1988-08-23 Alza Corporation Osmotic device for administering certain drugs
US4783337A (en) 1983-05-11 1988-11-08 Alza Corporation Osmotic system comprising plurality of members for dispensing drug
DE3320583A1 (en) 1983-06-08 1984-12-13 Dr. Karl Thomae Gmbh, 7950 Biberach NEW GALENIC PREPARATION FORMS OF ORAL ANTIDIABETICS AND METHOD FOR THE PRODUCTION THEREOF
JPS6058915A (en) 1983-09-12 1985-04-05 Fujisawa Pharmaceut Co Ltd Lipid microcapsule preparation containing medicament
US4594108A (en) 1983-09-19 1986-06-10 The Dow Chemical Company Highly pseudoplastic polymer solutions
US5385738A (en) 1983-10-14 1995-01-31 Sumitomo Pharmaceuticals Company, Ltd. Sustained-release injection
US4840896A (en) 1983-11-02 1989-06-20 Integrated Genetics, Inc. Heteropolymeric protein
US4923805A (en) 1983-11-02 1990-05-08 Integrated Genetics, Inc. Fsh
US5639639A (en) 1983-11-02 1997-06-17 Genzyme Corporation Recombinant heterodimeric human fertility hormones, and methods, cells, vectors and DNA for the production thereof
US4855238A (en) 1983-12-16 1989-08-08 Genentech, Inc. Recombinant gamma interferons having enhanced stability and methods therefor
MX9203641A (en) 1983-12-16 1992-07-01 Genentech Inc RECOMBINANT GAMMA INTERFERONS THAT HAVE IMPROVED STABILITY AND BIOTECHNOLOGICAL METHODS FOR THEIR OBTAINING.
US4851228A (en) 1984-06-20 1989-07-25 Merck & Co., Inc. Multiparticulate controlled porosity osmotic
US5231176A (en) 1984-08-27 1993-07-27 Genentech, Inc. Distinct family DNA encoding of human leukocyte interferons
US5120832A (en) 1984-08-27 1992-06-09 Genentech, Inc. Distinct family of human leukocyte interferons
US4927687A (en) 1984-10-01 1990-05-22 Biotek, Inc. Sustained release transdermal drug delivery composition
US5411951A (en) 1984-10-04 1995-05-02 Monsanto Company Prolonged release of biologically active somatotropin
IE58110B1 (en) 1984-10-30 1993-07-14 Elan Corp Plc Controlled release powder and process for its preparation
FI90990C (en) 1984-12-18 1994-04-25 Boehringer Ingelheim Int Recombinant DNA molecule, transformed host organism, and method for producing interferon
US4655462A (en) 1985-01-07 1987-04-07 Peter J. Balsells Canted coiled spring and seal
JPS61189230A (en) 1985-02-19 1986-08-22 Nippon Kayaku Co Ltd Etoposide preparation
US4609374A (en) 1985-04-22 1986-09-02 Alza Corporation Osmotic device comprising means for governing initial time of agent release therefrom
ATE78262T1 (en) 1985-06-11 1992-08-15 Ciba Geigy Ag HYBRID INTERFERONS.
US4845196A (en) 1985-06-24 1989-07-04 G. D. Searle & Co. Modified interferon gammas
US4847079A (en) 1985-07-29 1989-07-11 Schering Corporation Biologically stable interferon compositions comprising thimerosal
IE59361B1 (en) 1986-01-24 1994-02-09 Akzo Nv Pharmaceutical preparation for obtaining a highly viscous hydrogel or suspension
DE3607835A1 (en) 1986-03-10 1987-09-24 Boehringer Ingelheim Int HYBRID INTERFERONS, THEIR USE AS MEDICINAL PRODUCTS AND AS INTERMEDIATE PRODUCTS FOR THE PRODUCTION OF ANTIBODIES AND THE USE THEREOF AND METHOD FOR THEIR PRODUCTION
US4865845A (en) 1986-03-21 1989-09-12 Alza Corporation Release rate adjustment of osmotic or diffusional delivery devices
US4737437A (en) 1986-03-27 1988-04-12 East Shore Chemical Co. Light sensitive diazo compound, composition and method of making the composition
US5614492A (en) 1986-05-05 1997-03-25 The General Hospital Corporation Insulinotropic hormone GLP-1 (7-36) and uses thereof
US7138486B2 (en) 1986-05-05 2006-11-21 The General Hospital Corporation Insulinotropic hormone derivatives and uses thereof
US5118666A (en) 1986-05-05 1992-06-02 The General Hospital Corporation Insulinotropic hormone
US6849708B1 (en) 1986-05-05 2005-02-01 The General Hospital Corporation Insulinotropic hormone and uses thereof
US5120712A (en) 1986-05-05 1992-06-09 The General Hospital Corporation Insulinotropic hormone
US4755180A (en) 1986-06-16 1988-07-05 Alza Corporation Dosage form comprising solubility regulating member
DE3636123A1 (en) 1986-10-23 1988-05-05 Rentschler Arzneimittel ORAL ADMINISTRATIVE PREPARATIONS CONTAINING SINGLE DOSE FROM 10 TO 240 MG DIHYDROPYRIDINE
ZA878295B (en) 1986-11-06 1988-05-03 Amarillo Cell Culture Co. Inc. Treatment of immuno-resistant disease
CA1320905C (en) 1986-11-06 1993-08-03 Joseph M. Cummins Treatment of immuno-resistant disease
DE3642096A1 (en) 1986-12-10 1988-06-16 Boehringer Ingelheim Int HORSE (GAMMA) INTERFERON
US5371089A (en) 1987-02-26 1994-12-06 Senetek, Plc Method and composition for ameliorating the adverse effects of aging
US5278151A (en) 1987-04-02 1994-01-11 Ocular Research Of Boston, Inc. Dry eye treatment solution
JPH0720866B2 (en) 1987-05-15 1995-03-08 三生製薬株式会社 Transdermal preparation containing eperisone or tolperisone or their salts
US4940465A (en) 1987-05-27 1990-07-10 Felix Theeuwes Dispenser comprising displaceable matrix with solid state properties
US4892778A (en) 1987-05-27 1990-01-09 Alza Corporation Juxtaposed laminated arrangement
US5938654A (en) 1987-06-25 1999-08-17 Alza Corporation Osmotic device for delayed delivery of agent
US5023088A (en) 1987-06-25 1991-06-11 Alza Corporation Multi-unit delivery system
US4874388A (en) 1987-06-25 1989-10-17 Alza Corporation Multi-layer delivery system
US4915949A (en) 1987-07-13 1990-04-10 Alza Corporation Dispenser with movable matrix comprising a plurality of tiny pills
US4897268A (en) 1987-08-03 1990-01-30 Southern Research Institute Drug delivery system and method of making the same
DE3879031T2 (en) 1987-08-08 1993-06-24 Akzo Nv CONCEPTUAL IMPLANT.
US4915954A (en) 1987-09-03 1990-04-10 Alza Corporation Dosage form for delivering a drug at two different rates
US5756450A (en) 1987-09-15 1998-05-26 Novartis Corporation Water soluble monoesters as solubilisers for pharmacologically active compounds and pharmaceutical excipients and novel cyclosporin galenic forms
US4886668A (en) 1987-09-24 1989-12-12 Merck & Co., Inc. Multiparticulate controlled porosity osmotic pump
GB8723846D0 (en) 1987-10-10 1987-11-11 Danbiosyst Ltd Bioadhesive microsphere drug delivery system
AU2810189A (en) 1987-10-30 1989-05-23 Stolle Research & Development Corporation Low residual solvent microspheres and microencapsulation process
US4917895A (en) 1987-11-02 1990-04-17 Alza Corporation Transdermal drug delivery device
US4915366A (en) 1988-04-25 1990-04-10 Peter J. Balsells Outside back angle canted coil spring
US5203849A (en) 1990-03-20 1993-04-20 Balsells Peter J Canted coil spring in length filled with an elastomer
US4934666A (en) 1988-04-25 1990-06-19 Peter J. Balsells Coiled spring electromagnetic shielding gasket
US4964204A (en) 1988-04-25 1990-10-23 Peter J. Balsells Method for making a garter-type axially-resilient coil spring
US4830344A (en) 1988-04-25 1989-05-16 Peter J. Balsells Canted-coil spring with turn angle and seal
US5160122A (en) 1990-03-20 1992-11-03 Peter J. Balsells Coil spring with an elastomer having a hollow coil cross section
US5117066A (en) 1988-04-25 1992-05-26 Peter J. Balsells Retaining and locking electromagnetic gasket
US5072070A (en) 1989-12-01 1991-12-10 Peter J. Balsells Device for sealing electromagnetic waves
US4961253A (en) 1988-04-25 1990-10-09 Peter J. Balsells Manufacturing method for canted-coil spring with turn angle and seal
EP0339544B1 (en) 1988-04-25 1993-09-22 Peter J. Balsells Garter spring with canted back angle located on outside diameter
US5079388A (en) 1989-12-01 1992-01-07 Peter J. Balsells Gasket for sealing electromagnetic waves
US4876781A (en) 1988-04-25 1989-10-31 Peter J. Balsells Method of making a garter-type axially resilient coiled spring
US4826144A (en) 1988-04-25 1989-05-02 Peter J. Balsells Inside back angle canted coil spring
US4893795A (en) 1988-08-15 1990-01-16 Peter J. Balsells Radially loaded canted coiled spring with turn angle
US4974821A (en) 1988-04-25 1990-12-04 Peter J. Balsells Canted-coil spring with major axis radial loading
US5108078A (en) 1988-04-25 1992-04-28 Peter J. Balsells Canted-coil spring loaded while in a cavity
US4907788A (en) 1988-04-25 1990-03-13 Peter J. Balsells Dual concentric canted-coil spring apparatus
US5024842A (en) 1988-04-28 1991-06-18 Alza Corporation Annealed coats
US5160743A (en) 1988-04-28 1992-11-03 Alza Corporation Annealed composition for pharmaceutically acceptable drug
US5006346A (en) 1988-04-28 1991-04-09 Alza Corporation Delivery system
US4931285A (en) 1988-04-28 1990-06-05 Alza Corporation Aqueous based pharmaceutical coating composition for dosage forms
JP2827287B2 (en) 1988-07-05 1998-11-25 武田薬品工業株式会社 Sustained release microcapsules containing water-soluble drugs
JP2794022B2 (en) 1988-11-11 1998-09-03 三生製薬株式会社 Transdermal preparation containing bunazosin or its salts
US5059423A (en) 1988-12-13 1991-10-22 Alza Corporation Delivery system comprising biocompatible beneficial agent formulation
US5728088A (en) 1988-12-13 1998-03-17 Alza Corporation Osmotic system for delivery of fluid-sensitive somatotropins to bovine animals
US5034229A (en) 1988-12-13 1991-07-23 Alza Corporation Dispenser for increasing feed conversion of hog
US5110596A (en) 1988-12-13 1992-05-05 Alza Corporation Delivery system comprising means for delivering agent to livestock
US5057318A (en) 1988-12-13 1991-10-15 Alza Corporation Delivery system for beneficial agent over a broad range of rates
US4969884A (en) 1988-12-28 1990-11-13 Alza Corporation Osmotically driven syringe
US5234424A (en) 1988-12-28 1993-08-10 Alza Corporation Osmotically driven syringe
US4976966A (en) 1988-12-29 1990-12-11 Alza Corporation Delayed release osmotically driven fluid dispenser
IL92344A0 (en) 1989-01-04 1990-07-26 Gist Brocades Nv Microencapsulation of bioactive substances in biocompatible polymers,microcapsules obtained and pharmaceutical preparation comprising said microcapsules
US5288479A (en) 1989-01-17 1994-02-22 Sterling Drug, Inc. Extrudable elastic oral pharmaceutical gel compositions and metered dose dispensers containing them and method of making and method of use thereof
US5906816A (en) 1995-03-16 1999-05-25 University Of Florida Method for treatment of autoimmune diseases
US5705363A (en) 1989-03-02 1998-01-06 The Women's Research Institute Recombinant production of human interferon τ polypeptides and nucleic acids
US5219572A (en) 1989-03-17 1993-06-15 Pitman-Moore, Inc. Controlled release delivery device for macromolecular proteins
AU620253B2 (en) 1989-05-01 1992-02-13 Alkermes Controlled Therapeutics, Inc. Process for producing small particles of biologically active molecules
US5019400A (en) 1989-05-01 1991-05-28 Enzytech, Inc. Very low temperature casting of controlled release microspheres
EP0471036B2 (en) 1989-05-04 2004-06-23 Southern Research Institute Encapsulation process
US5133974A (en) 1989-05-05 1992-07-28 Kv Pharmaceutical Company Extended release pharmaceutical formulations
US5126142A (en) 1989-07-18 1992-06-30 Alza Corporation Dispenser comprising ionophore
US5439688A (en) 1989-07-28 1995-08-08 Debio Recherche Pharmaceutique S.A. Process for preparing a pharmaceutical composition
US5225205A (en) 1989-07-28 1993-07-06 Debiopharm S.A. Pharmaceutical composition in the form of microparticles
AU6402490A (en) 1989-08-28 1991-04-08 Pharmaceutical Delivery Systems, Inc. Bioerodible polymers useful for the controlled release of therapeutic agents
US5112614A (en) 1989-09-14 1992-05-12 Alza Corporation Implantable delivery dispenser
US5290558A (en) 1989-09-21 1994-03-01 Osteotech, Inc. Flowable demineralized bone powder composition and its use in bone repair
SE465950B (en) 1989-10-23 1991-11-25 Medinvent Sa Combination of an aggregate particle size, crystalline or freeze-dried drug with a pseudoplastic gel for preparation of an injectable preparation as well as a process for its preparation
US5707644A (en) 1989-11-04 1998-01-13 Danbiosyst Uk Limited Small particle compositions for intranasal drug delivery
US5312335A (en) 1989-11-09 1994-05-17 Bioject Inc. Needleless hypodermic injection device
ES2046039T3 (en) 1989-11-13 1994-01-16 Becton Dickinson France STORAGE BOTTLE CONTAINING A COMPONENT OF A MEDICINAL SOLUTION.
JPH03236317A (en) 1989-12-06 1991-10-22 Sansei Seiyaku Kk Dopamine derivative-containing percutaneous
US5030216A (en) 1989-12-15 1991-07-09 Alza Corporation Osmotically driven syringe
US5733572A (en) 1989-12-22 1998-03-31 Imarx Pharmaceutical Corp. Gas and gaseous precursor filled microspheres as topical and subcutaneous delivery vehicles
USD326718S (en) 1989-12-26 1992-06-02 Minnesota Mining And Manufacturing Co. Blood sensor cassette
US5545618A (en) 1990-01-24 1996-08-13 Buckley; Douglas I. GLP-1 analogs useful for diabetes treatment
US5213809A (en) 1990-01-24 1993-05-25 Alza Corporation Delivery system comprising means for controlling internal pressure
US5223266A (en) 1990-01-24 1993-06-29 Alza Corporation Long-term delivery device with early startup
US5126147A (en) 1990-02-08 1992-06-30 Biosearch, Inc. Sustained release dosage form
US5478564A (en) 1990-02-22 1995-12-26 Teva Pharmaceutical Industries, Ltd. Preparation of microparticles for controlled release of water-soluble substances
US5122128A (en) 1990-03-15 1992-06-16 Alza Corporation Orifice insert for a ruminal bolus
US5120306A (en) 1990-03-21 1992-06-09 Gosselin Leon F Direct delivery of anti-inflammatories to the proximal small bowel
US5213810A (en) 1990-03-30 1993-05-25 American Cyanamid Company Stable compositions for parenteral administration and method of making same
US5207752A (en) 1990-03-30 1993-05-04 Alza Corporation Iontophoretic drug delivery system with two-stage delivery profile
US5324280A (en) 1990-04-02 1994-06-28 Alza Corporation Osmotic dosage system for delivering a formulation comprising liquid carrier and drug
US5091188A (en) 1990-04-26 1992-02-25 Haynes Duncan H Phospholipid-coated microcrystals: injectable formulations of water-insoluble drugs
US5290271A (en) 1990-05-14 1994-03-01 Jernberg Gary R Surgical implant and method for controlled release of chemotherapeutic agents
US5374620A (en) 1990-06-07 1994-12-20 Genentech, Inc. Growth-promoting composition and its use
US5234693A (en) 1990-07-11 1993-08-10 Alza Corporation Delivery device with a protective sleeve
US5180591A (en) 1990-07-11 1993-01-19 Alza Corporation Delivery device with a protective sleeve
US5234692A (en) 1990-07-11 1993-08-10 Alza Corporation Delivery device with a protective sleeve
US5234695A (en) 1990-07-24 1993-08-10 Eastman Kodak Company Water dispersible vitamin E composition
USD329278S (en) 1990-10-01 1992-09-08 Gallup Allen I Infant's bathing support
US5300302A (en) 1990-10-04 1994-04-05 Nestec S.A. Pharmaceutical composition in gel form in a dispensing package
US5529914A (en) 1990-10-15 1996-06-25 The Board Of Regents The Univeristy Of Texas System Gels for encapsulation of biological materials
US5151093A (en) 1990-10-29 1992-09-29 Alza Corporation Osmotically driven syringe with programmable agent delivery
US5122377A (en) 1990-11-19 1992-06-16 A.H. Robins, Company, Incorporated Oral delivery system for veterinary drugs
IT1243390B (en) 1990-11-22 1994-06-10 Vectorpharma Int PHARMACEUTICAL COMPOSITIONS IN THE FORM OF PARTICLES SUITABLE FOR THE CONTROLLED RELEASE OF PHARMACOLOGICALLY ACTIVE SUBSTANCES AND PROCEDURE FOR THEIR PREPARATION.
US5161806A (en) 1990-12-17 1992-11-10 Peter J. Balsells Spring-loaded, hollow, elliptical ring seal
GB9027422D0 (en) 1990-12-18 1991-02-06 Scras Osmotically driven infusion device
PT99989A (en) 1991-01-09 1994-05-31 Alza Corp BIODEGRADABLE AND COMPOSITION DEVICES FOR DIFFUSIBLE LIBERATION OF AGENTS
US5443459A (en) 1991-01-30 1995-08-22 Alza Corporation Osmotic device for delayed delivery of agent
NL9100160A (en) 1991-01-30 1992-08-17 Texas Instruments Holland INJECTOR.
US5861166A (en) 1991-03-12 1999-01-19 Alza Corporation Delivery device providing beneficial agent stability
CA2102507A1 (en) 1991-05-07 1992-11-08 Judith P. Kitchell A controlled, sustained release delivery system for smoking cessation
US5113938A (en) 1991-05-07 1992-05-19 Clayton Charley H Whipstock
US5137727A (en) 1991-06-12 1992-08-11 Alza Corporation Delivery device providing beneficial agent stability
EP0520119A1 (en) 1991-06-17 1992-12-30 Spirig Ag Pharmazeutische Präparate New oral diclofenac composition
US5190765A (en) 1991-06-27 1993-03-02 Alza Corporation Therapy delayed
US5252338A (en) 1991-06-27 1993-10-12 Alza Corporation Therapy delayed
HU222501B1 (en) 1991-06-28 2003-07-28 Endorecherche Inc. Controlled release pharmaceutical composition containing mpa or mga and process for its preparation
DE4122217C2 (en) 1991-07-04 1997-02-13 Merz & Co Gmbh & Co Process for the preparation of mechanically stable, well decomposing compressed products from small active substance-containing moldings
US5288214A (en) 1991-09-30 1994-02-22 Toshio Fukuda Micropump
YU87892A (en) 1991-10-01 1995-12-04 Eli Lilly And Company Lilly Corporate Center INJECTIBLE LONG TERM RELEASE FORMULATIONS AND PROCEDURES FOR THEIR OBTAINING AND USE
WO1993006821A1 (en) 1991-10-04 1993-04-15 Yoshitomi Pharmaceutical Industries, Ltd. Sustained-release tablet
WO1993006819A1 (en) 1991-10-10 1993-04-15 Alza Corporation Osmotic drug delivery devices with hydrophobic wall materials
US5288502A (en) 1991-10-16 1994-02-22 The University Of Texas System Preparation and uses of multi-phase microspheres
US5318780A (en) 1991-10-30 1994-06-07 Mediventures Inc. Medical uses of in situ formed gels
US5236707A (en) 1991-11-08 1993-08-17 Dallas Biotherapeutics, Inc. Stabilization of human interferon
DE4137649C2 (en) 1991-11-15 1997-11-20 Gerhard Dingler Component
AU3136293A (en) 1991-11-15 1993-06-15 Isp Investments Inc. Pharmaceutical tablet with pvp having an enhanced drug dissolution rate
US5200195A (en) 1991-12-06 1993-04-06 Alza Corporation Process for improving dosage form delivery kinetics
US5580578A (en) 1992-01-27 1996-12-03 Euro-Celtique, S.A. Controlled release formulations coated with aqueous dispersions of acrylic polymers
US5223265A (en) 1992-01-10 1993-06-29 Alza Corporation Osmotic device with delayed activation of drug delivery
US5658593A (en) 1992-01-16 1997-08-19 Coletica Injectable compositions containing collagen microcapsules
US5676942A (en) 1992-02-10 1997-10-14 Interferon Sciences, Inc. Composition containing human alpha interferon species proteins and method for use thereof
US5456679A (en) 1992-02-18 1995-10-10 Alza Corporation Delivery devices with pulsatile effect
US5209746A (en) 1992-02-18 1993-05-11 Alza Corporation Osmotically driven delivery devices with pulsatile effect
US5308348A (en) 1992-02-18 1994-05-03 Alza Corporation Delivery devices with pulsatile effect
US5573934A (en) 1992-04-20 1996-11-12 Board Of Regents, The University Of Texas System Gels for encapsulation of biological materials
JP3267972B2 (en) 1992-02-28 2002-03-25 コラーゲン コーポレイション High concentration homogenized collagen composition
US5221278A (en) 1992-03-12 1993-06-22 Alza Corporation Osmotically driven delivery device with expandable orifice for pulsatile delivery effect
US5656297A (en) 1992-03-12 1997-08-12 Alkermes Controlled Therapeutics, Incorporated Modulated release from biocompatible polymers
US5540912A (en) 1992-03-30 1996-07-30 Alza Corporation Viscous suspensions of controlled-release drug particles
EP0633907A1 (en) 1992-03-30 1995-01-18 Alza Corporation Additives for bioerodible polymers to regulate degradation
US6197346B1 (en) 1992-04-24 2001-03-06 Brown Universtiy Research Foundation Bioadhesive microspheres and their use as drug delivery and imaging systems
FR2690622B1 (en) 1992-04-29 1995-01-20 Chronotec Programmable ambulatory infusion pump system.
US5314685A (en) 1992-05-11 1994-05-24 Agouron Pharmaceuticals, Inc. Anhydrous formulations for administering lipophilic agents
US5711968A (en) 1994-07-25 1998-01-27 Alkermes Controlled Therapeutics, Inc. Composition and method for the controlled release of metal cation-stabilized interferon
JP2651320B2 (en) 1992-07-16 1997-09-10 田辺製薬株式会社 Method for producing sustained-release microsphere preparation
US5413672A (en) 1992-07-22 1995-05-09 Ngk Insulators, Ltd. Method of etching sendust and method of pattern-etching sendust and chromium films
US5512293A (en) 1992-07-23 1996-04-30 Alza Corporation Oral sustained release drug delivery device
US5609885A (en) 1992-09-15 1997-03-11 Alza Corporation Osmotic membrane and delivery device
GB9223146D0 (en) 1992-11-05 1992-12-16 Scherer Corp R P Vented capsule
DE69224386T2 (en) 1992-11-06 1998-06-18 Texas Instruments Inc Device for subcutaneous insertion of a needle
DE69327542T2 (en) 1992-11-17 2000-07-06 Yoshitomi Pharmaceutical ANTIPSYCHOTIC-CONTAINING MICROBALL FOR DELAYED RELEASE AND METHOD FOR THEIR PRODUCTION
USD342855S (en) 1992-11-20 1994-01-04 Butler Ii George D Combined infant cushion and cover
US5260069A (en) 1992-11-27 1993-11-09 Anda Sr Pharmaceuticals Inc. Pulsatile particles drug delivery system
SE9203594D0 (en) 1992-11-30 1992-11-30 Christer Nystroem DISPERSA SYSTEM MEDICINAL PRODUCT
PT674506E (en) 1992-12-02 2001-01-31 Alkermes Inc MICROSPHERES CONTAINING GROWTH HORMONE WITH CONTROLLED LIBERATION
TW333456B (en) 1992-12-07 1998-06-11 Takeda Pharm Ind Co Ltd A pharmaceutical composition of sustained-release preparation the invention relates to a pharmaceutical composition of sustained-release preparation which comprises a physiologically active peptide.
PT686045E (en) 1993-02-23 2001-04-30 Genentech Inc STABILIZATION BY EXPIPIENTS OF POLYPEPTIDES TREATED WITH ORGANIC SOLVENTS
US5368588A (en) 1993-02-26 1994-11-29 Bettinger; David S. Parenteral fluid medication reservoir pump
US5981719A (en) 1993-03-09 1999-11-09 Epic Therapeutics, Inc. Macromolecular microparticles and methods of production and use
AU6410494A (en) 1993-03-17 1994-10-11 Alza Corporation Device for the transdermal administration of alprazolam
US5514110A (en) 1993-03-22 1996-05-07 Teh; Eutiquio L. Automatic flow control device
US6284727B1 (en) 1993-04-07 2001-09-04 Scios, Inc. Prolonged delivery of peptides
NZ250844A (en) 1993-04-07 1996-03-26 Pfizer Treatment of non-insulin dependant diabetes with peptides; composition
TW360548B (en) 1993-04-08 1999-06-11 Powderject Res Ltd Products for therapeutic use
NZ247516A (en) 1993-04-28 1995-02-24 Bernard Charles Sherman Water dispersible pharmaceutical compositions comprising drug dissolved in solvent system comprising at least one alcohol and at least one surfactant
US5424286A (en) 1993-05-24 1995-06-13 Eng; John Exendin-3 and exendin-4 polypeptides, and pharmaceutical compositions comprising same
US5639477A (en) 1993-06-23 1997-06-17 Alza Corporation Ruminal drug delivery device
WO1995001167A2 (en) 1993-06-25 1995-01-12 Alza Corporation Incorporating poly-n-vinyl amide in a transdermal system
US5498255A (en) 1993-08-17 1996-03-12 Alza Corporation Osmotic device for protracted pulsatile delivery of agent
US5385887A (en) 1993-09-10 1995-01-31 Genetics Institute, Inc. Formulations for delivery of osteogenic proteins
JP2700141B2 (en) 1993-09-17 1998-01-19 富士化学工業株式会社 Calcium hydrogen phosphate, its production method and excipient using the same
US5747065A (en) 1993-09-29 1998-05-05 Lee; Eun Soo Monoglyceride/lactate ester permeation enhancer for oxybutynin
US6913767B1 (en) 1993-10-25 2005-07-05 Genentech, Inc. Compositions for microencapsulation of antigens for use as vaccines
US5650173A (en) 1993-11-19 1997-07-22 Alkermes Controlled Therapeutics Inc. Ii Preparation of biodegradable microparticles containing a biologically active agent
EP2283821A1 (en) 1993-11-19 2011-02-16 Alkermes, Inc. Preparation of biodegradable microparticles containing a biologically active agent
ATE288270T1 (en) 1993-11-19 2005-02-15 Janssen Pharmaceutica Nv MICRO-ENCAPSULED 1,2-BENZAZOLE
JPH07196479A (en) 1994-01-04 1995-08-01 Unitika Ltd Method for producing microcapsule
USD358644S (en) 1994-01-18 1995-05-23 Bio Medic Data Systems, Inc. Transponder implanter
US6241734B1 (en) 1998-08-14 2001-06-05 Kyphon, Inc. Systems and methods for placing materials into bone
US5540665A (en) 1994-01-31 1996-07-30 Alza Corporation Gas driven dispensing device and gas generating engine therefor
NZ279953A (en) 1994-02-04 1998-02-26 Scotia Lipidteknik Ab Lipid-polar solvent bilayer carrier preparation (eg liposomes) comprising digalactosyldiacylglycerols as the bilayer material
US5697975A (en) 1994-02-09 1997-12-16 The University Of Iowa Research Foundation Human cerebral cortex neural prosthetic for tinnitus
US5458888A (en) 1994-03-02 1995-10-17 Andrx Pharmaceuticals, Inc. Controlled release tablet formulation
WO1995024212A1 (en) 1994-03-07 1995-09-14 Imperial College Of Science, Technology & Medicine The use of interferon subtypes in the preparation of medicaments to treat viral infections
ZA953078B (en) 1994-04-28 1996-01-05 Alza Corp Effective therapy for epilepsies
DE69431533T2 (en) 1994-06-13 2003-10-02 Alza Corp DOSAGE FORM FOR ADMINISTRATION OF LIQUID MEDICINAL FORMULATION
NL9401150A (en) 1994-07-12 1996-02-01 Nederland Ptt Method for presenting on a receiving side a first number of video signals originating from a transmitting side, as well as a system, as well as a transmitter, as well as a network, and also a receiver.
ES2153485T3 (en) 1994-07-13 2001-03-01 Alza Corp COMPOSITION AND PROCEDURE THAT INCREASES THE PERCUTANEOUS DIFFUSION BY ELECTROTRANSPORT OF A SUBSTANCE.
US5633011A (en) 1994-08-04 1997-05-27 Alza Corporation Progesterone replacement therapy
US5574008A (en) 1994-08-30 1996-11-12 Eli Lilly And Company Biologically active fragments of glucagon-like insulinotropic peptide
US5512549A (en) 1994-10-18 1996-04-30 Eli Lilly And Company Glucagon-like insulinotropic peptide analogs, compositions, and methods of use
US5595759A (en) 1994-11-10 1997-01-21 Alza Corporation Process for providing therapeutic composition
ATE232089T1 (en) 1994-11-10 2003-02-15 Univ Kentucky Res Found CONTROLLED RELEASE IMPLANTABLE REFILLABLE DEVICE FOR ADMINISTERING DRUGS IMMEDIATELY TO AN INTERNAL PART OF THE BODY
FR2731150B1 (en) 1995-03-03 1997-04-18 Oreal USE OF AMPHIPHILIC COMPOUNDS AS A THICKENING AGENT FOR NON-AQUEOUS MEDIA
US5844017A (en) 1995-03-06 1998-12-01 Ethicon, Inc. Prepolymers of absorbable polyoxaesters containing amines and/or amido groups
US5859150A (en) 1995-03-06 1999-01-12 Ethicon, Inc. Prepolymers of absorbable polyoxaesters
US6100346A (en) 1995-03-06 2000-08-08 Ethicon, Inc. Copolymers of polyoxaamides
US5595751A (en) 1995-03-06 1997-01-21 Ethicon, Inc. Absorbable polyoxaesters containing amines and/or amido groups
US6147168A (en) 1995-03-06 2000-11-14 Ethicon, Inc. Copolymers of absorbable polyoxaesters
US5648088A (en) 1995-03-06 1997-07-15 Ethicon, Inc. Blends of absorbable polyoxaesters containing amines and/or amide groups
US5962023A (en) 1995-03-06 1999-10-05 Ethicon, Inc. Hydrogels containing absorbable polyoxaamides
US5618552A (en) 1995-03-06 1997-04-08 Ethicon, Inc. Absorbable polyoxaesters
US5700583A (en) 1995-03-06 1997-12-23 Ethicon, Inc. Hydrogels of absorbable polyoxaesters containing amines or amido groups
US5597579A (en) 1995-03-06 1997-01-28 Ethicon, Inc. Blends of absorbable polyoxaamides
US5698213A (en) 1995-03-06 1997-12-16 Ethicon, Inc. Hydrogels of absorbable polyoxaesters
US6403655B1 (en) 1995-03-06 2002-06-11 Ethicon, Inc. Method of preventing adhesions with absorbable polyoxaesters
US5607687A (en) 1995-03-06 1997-03-04 Ethicon, Inc. Polymer blends containing absorbable polyoxaesters
US5464929A (en) 1995-03-06 1995-11-07 Ethicon, Inc. Absorbable polyoxaesters
IL113100A0 (en) 1995-03-23 1995-06-29 Schatz Anat Infant's mattress
US5542682A (en) 1995-03-27 1996-08-06 American Variseal Slant coil spring and seal
US5736159A (en) 1995-04-28 1998-04-07 Andrx Pharmaceuticals, Inc. Controlled release formulation for water insoluble drugs in which a passageway is formed in situ
AU694655B2 (en) 1995-05-02 1998-07-23 Taisho Pharmaceutical Co., Ltd. Composition for oral administration
US5939286A (en) 1995-05-10 1999-08-17 University Of Florida Hybrid interferon tau/alpha polypeptides, their recombinant production, and methods using them
US5922253A (en) 1995-05-18 1999-07-13 Alkermes Controlled Therapeutics, Inc. Production scale method of forming microparticles
US5882676A (en) 1995-05-26 1999-03-16 Alza Corporation Skin permeation enhancer compositions using acyl lactylates
US5718922A (en) 1995-05-31 1998-02-17 Schepens Eye Research Institute, Inc. Intravitreal microsphere drug delivery and method of preparation
WO1996039142A1 (en) 1995-06-06 1996-12-12 F. Hoffmann-La Roche Ag Pharmaceutical composition comprising a proteinase inhibitor and a monoglyceride
US6129761A (en) 1995-06-07 2000-10-10 Reprogenesis, Inc. Injectable hydrogel compositions
US5904935A (en) 1995-06-07 1999-05-18 Alza Corporation Peptide/protein suspending formulations
US5690952A (en) 1995-06-07 1997-11-25 Judy A. Magruder et al. Implantable system for delivery of fluid-sensitive agents to animals
US5747058A (en) 1995-06-07 1998-05-05 Southern Biosystems, Inc. High viscosity liquid controlled delivery system
PL181582B1 (en) 1995-06-07 2001-08-31 Ortho Mcneil Pharm Inc Skin sticking plaster and method of administering sole 17-deacetyl norgestimate or together with estrogen
US6572879B1 (en) 1995-06-07 2003-06-03 Alza Corporation Formulations for transdermal delivery of pergolide
US7833543B2 (en) 1995-06-07 2010-11-16 Durect Corporation High viscosity liquid controlled delivery system and medical or surgical device
US5782396A (en) 1995-08-28 1998-07-21 United States Surgical Corporation Surgical stapler
US5906830A (en) 1995-09-08 1999-05-25 Cygnus, Inc. Supersaturated transdermal drug delivery systems, and methods for manufacturing the same
US5942253A (en) 1995-10-12 1999-08-24 Immunex Corporation Prolonged release of GM-CSF
GB9521125D0 (en) 1995-10-16 1995-12-20 Unilever Plc Cosmetic composition
SE505146C2 (en) 1995-10-19 1997-06-30 Biogram Ab Particles for delayed release
US5766620A (en) 1995-10-23 1998-06-16 Theratech, Inc. Buccal delivery of glucagon-like insulinotropic peptides
GB9521805D0 (en) 1995-10-25 1996-01-03 Cortecs Ltd Solubilisation methods
ES2192221T3 (en) 1995-10-30 2003-10-01 Oleoyl Estrone Developments S MONOESTERS OF STROGEN OLEATE FOR THE TREATMENT OF OBESITY OR OVERWEIGHT.
ATE262920T1 (en) 1995-11-02 2004-04-15 Schering Corp CONTINUOUS, LOW DOSE CYTOKINE INFUSION THERAPY
US5908621A (en) 1995-11-02 1999-06-01 Schering Corporation Polyethylene glycol modified interferon therapy
CA2192782C (en) 1995-12-15 2008-10-14 Nobuyuki Takechi Production of microspheres
CA2192773C (en) 1995-12-15 2008-09-23 Hiroaki Okada Production of sustained-release preparation for injection
AUPN723395A0 (en) 1995-12-19 1996-01-18 Macnaught Medical Pty Limited Lubrication methods
US5980945A (en) 1996-01-16 1999-11-09 Societe De Conseils De Recherches Et D'applications Scientifique S.A. Sustained release drug formulations
EP0985513B1 (en) 1996-01-31 2003-01-02 Sumitomo Bakelite Company Limited Method of producing epoxy resin-encapsulated semiconductor device
US6132420A (en) 1996-02-02 2000-10-17 Alza Corporation Osmotic delivery system and method for enhancing start-up and performance of osmotic delivery systems
US6395292B2 (en) 1996-02-02 2002-05-28 Alza Corporation Sustained delivery of an active agent using an implantable system
RO119929B1 (en) 1996-02-02 2005-06-30 Alza Corporation Device for administering an active agent in a liquid medium
US6156331A (en) 1996-02-02 2000-12-05 Alza Corporation Sustained delivery of an active agent using an implantable system
US6261584B1 (en) 1996-02-02 2001-07-17 Alza Corporation Sustained delivery of an active agent using an implantable system
CA2279349C (en) 1996-02-02 2007-09-25 Rhomed Incorporated Ascorbate-stabilized radiopharmaceutical method and composition
US5807876A (en) 1996-04-23 1998-09-15 Vertex Pharmaceuticals Incorporated Inhibitors of IMPDH enzyme
US6245349B1 (en) 1996-02-23 2001-06-12 éLAN CORPORATION PLC Drug delivery compositions suitable for intravenous injection
KR100611130B1 (en) 1996-03-01 2006-11-30 노보 노르디스크 에이/에스 Use of a pharmaceutical composition comprising an appetite-supressing peptide
JPH09241153A (en) 1996-03-04 1997-09-16 Q P Corp Lipid emulsion for intravenous injection
IL126095A0 (en) 1996-03-08 1999-05-09 Zeneca Ltd Azolobenzazepine derivatives as neurogically active agents
AU728146B2 (en) 1996-03-14 2001-01-04 Immune Response Corporation, The Targeted delivery of genes encoding interferon
US5703200A (en) 1996-03-15 1997-12-30 Ethicon, Inc. Absorbable copolymers and blends of 6,6-dialkyl-1,4-dioxepan-2-one and its cyclic dimer
DE69717263T2 (en) 1996-03-28 2003-07-24 Takeda Chemical Industries Ltd PREPARATION WITH DELAYED RELEASE AND THEIR PRODUCTION
US5660858A (en) 1996-04-03 1997-08-26 Research Triangle Pharmaceuticals Cyclosporin emulsions
US6204022B1 (en) 1996-04-12 2001-03-20 Pepgen Corporation And University Of Florida Low-toxicity human interferon-alpha analogs
US6074673A (en) 1996-04-22 2000-06-13 Guillen; Manuel Slow-release, self-absorbing, drug delivery system
US5976109A (en) 1996-04-30 1999-11-02 Medtronic, Inc. Apparatus for drug infusion implanted within a living body
US5792477A (en) 1996-05-07 1998-08-11 Alkermes Controlled Therapeutics, Inc. Ii Preparation of extended shelf-life biodegradable, biocompatible microparticles containing a biologically active agent
TW487572B (en) 1996-05-20 2002-05-21 Janssen Pharmaceutica Nv Aqueous suspensions of 9-hydroxyrisperidone fatty acid esters
US6214367B1 (en) 1996-06-05 2001-04-10 Ashmont Holdings Limited Injectable compositions
JP2000516912A (en) 1996-06-05 2000-12-19 ロシュ ダイアグノスティクス ゲゼルシャフト ミット ベシュレンクテル ハフツング Exendin analogs, methods for their preparation and formulations containing them
DE29610419U1 (en) 1996-06-14 1996-10-24 Filtertek Sa Gravity infusion device for medical infusions
GB9613858D0 (en) 1996-07-02 1996-09-04 Cortecs Ltd Hydrophobic preparations
US5916582A (en) 1996-07-03 1999-06-29 Alza Corporation Aqueous formulations of peptides
DK0909175T3 (en) 1996-07-03 2003-09-29 Alza Corp Non-aqueous protic peptide formulations
US5932547A (en) 1996-07-03 1999-08-03 Alza Corporation Non-aqueous polar aprotic peptide formulations
AU3601897A (en) 1996-07-15 1998-02-09 Alza Corporation Novel formulations for the administration of fluoxetine
AR008789A1 (en) 1996-07-31 2000-02-23 Bayer Corp PIRIDINES AND SUBSTITUTED BIPHENYLS
WO1998005351A1 (en) 1996-08-08 1998-02-12 Amylin Pharmaceuticals, Inc. Methods for regulating gastrointestinal motility
AU3981097A (en) 1996-08-21 1998-03-06 Alkermes Controlled Therapeutics, Inc. Controlled release microparticles with a hydrophobic material
US6458924B2 (en) 1996-08-30 2002-10-01 Novo Nordisk A/S Derivatives of GLP-1 analogs
US6268343B1 (en) 1996-08-30 2001-07-31 Novo Nordisk A/S Derivatives of GLP-1 analogs
US5984890A (en) 1996-09-27 1999-11-16 American Home Products Corporation Medical device for the placement of solid materials
IN184589B (en) 1996-10-16 2000-09-09 Alza Corp
ES2191834T3 (en) 1996-10-24 2003-09-16 Alza Corp AGENTS THAT FACILITATE THE PERMEATION AND DESTINATIONS FOR COMPOSITIONS, DEVICES AND PROCEDURES OF TRANSDERMAL CONTRIBUTION OF DRUGS.
US5817129A (en) 1996-10-31 1998-10-06 Ethicon, Inc. Process and apparatus for coating surgical sutures
UA65549C2 (en) 1996-11-05 2004-04-15 Елі Ліллі Енд Компані Use of glucagon-like peptides such as glp-1, glp-1 analog, or glp-1 derivative in methods and compositions for reducing body weight
DE19646392A1 (en) 1996-11-11 1998-05-14 Lohmann Therapie Syst Lts Preparation for use in the oral cavity with a layer containing pressure-sensitive adhesive, pharmaceuticals or cosmetics for dosed delivery
US5928666A (en) 1996-11-12 1999-07-27 Cygnus Inc. Crystalline form of estradiol and pharmaceutical formulations comprising same
PT1066081E (en) 1996-11-15 2003-12-31 Alza Corp OSMOTIC DISTRIBUTION SYSTEM AND PROCESS FOR IMPROVING THE START AND PERFORMANCE OF OSMOTIC DISTRIBUTION SYSTEMS
PT949905E (en) 1996-12-20 2001-12-28 Alza Corp DELAYED EFFECT INJECTABLE GEL COMPOSITION AND PROCESS FOR THEIR PREPARATION
GB9626513D0 (en) 1996-12-20 1997-02-05 Bioglan Ireland R & D Ltd A pharmaceutical composition
EP1629849B2 (en) 1997-01-07 2017-10-04 Amylin Pharmaceuticals, LLC Pharmaceutical compositions comprising exendins and agonists thereof
JP2001511128A (en) 1997-01-28 2001-08-07 ファルマシア・アンド・アップジョン・カンパニー Lyophilized product of lipid complex of water-insoluble porphyrin
US5945126A (en) 1997-02-13 1999-08-31 Oakwood Laboratories L.L.C. Continuous microsphere process
ZA981610B (en) 1997-03-24 1999-08-26 Alza Corp Self adjustable exit port.
US5874388A (en) 1997-04-02 1999-02-23 Dow Corning Corporation Lubricant composition for disc brake caliper pin and a disc brake asembly containing the lubricant
US6127520A (en) 1997-04-15 2000-10-03 Regents Of The University Of Michigan Compositions and methods for the inhibition of neurotransmitter uptake of synaptic vesicles
WO1998047487A1 (en) 1997-04-17 1998-10-29 Dumex-Alpharma A/S A novel bioadhesive drug delivery system based on liquid crystals
MY118835A (en) 1997-04-18 2005-01-31 Ipsen Pharma Biotech Sustained release compositions and the process for their preparation
KR20010020342A (en) 1997-04-28 2001-03-15 자끄 사비나 Adenovirus-mediated intratumoral delivery of an angiogenesis antagonist for the treatment of tumors
US20020039594A1 (en) 1997-05-13 2002-04-04 Evan C. Unger Solid porous matrices and methods of making and using the same
US6113947A (en) 1997-06-13 2000-09-05 Genentech, Inc. Controlled release microencapsulated NGF formulation
US6663899B2 (en) 1997-06-13 2003-12-16 Genentech, Inc. Controlled release microencapsulated NGF formulation
SI9700186B (en) 1997-07-14 2006-10-31 Lek, Tovarna Farmacevtskih In Kemicnih Izdelkov, D.D. Novel pharmaceutical preparation with controlled release of active healing substances
MY125849A (en) 1997-07-25 2006-08-30 Alza Corp Osmotic delivery system, osmotic delivery system semipermeable body assembly, and method for controlling delivery rate of beneficial agents from osmotic delivery systems
MY125870A (en) 1997-07-25 2006-08-30 Alza Corp Osmotic delivery system flow modulator apparatus and method
US7157555B1 (en) 1997-08-08 2007-01-02 Amylin Pharmaceuticals, Inc. Exendin agonist compounds
GB9718986D0 (en) 1997-09-09 1997-11-12 Danbiosyst Uk Controlled release microsphere delivery system
US6023802A (en) 1997-09-10 2000-02-15 King; Susan Melton Infant sleeper
US6172046B1 (en) 1997-09-21 2001-01-09 Schering Corporation Combination therapy for eradicating detectable HCV-RNA in patients having chronic Hepatitis C infection
US5989463A (en) 1997-09-24 1999-11-23 Alkermes Controlled Therapeutics, Inc. Methods for fabricating polymer-based controlled release devices
AU750567B2 (en) 1997-09-29 2002-07-25 Novartis Ag Stabilized preparations for use in nebulizers
JPH11100353A (en) 1997-09-29 1999-04-13 Esupo Kk Refined and deodorized liquid ester wax and its composition
US6133429A (en) 1997-10-03 2000-10-17 Becton Dickinson And Company Chromophores useful for the preparation of novel tandem conjugates
USD399821S (en) 1997-11-07 1998-10-20 Motorola, Inc. Front section of a battery charger
WO1999025728A1 (en) 1997-11-14 1999-05-27 Amylin Pharmaceuticals, Inc. Novel exendin agonist compounds
WO1999029863A1 (en) 1997-12-08 1999-06-17 Genentech, Inc. Human interferon-epsilon: a type i interferon
US6368612B1 (en) 1997-12-12 2002-04-09 Biohybrid Technologies Llc Devices for cloaking transplanted cells
WO1999032095A1 (en) 1997-12-22 1999-07-01 Alza Corporation Rate controlling membranes for controlled drug delivery devices
ES2216340T3 (en) 1997-12-29 2004-10-16 Alza Corporation IMPLANTING DEVICE FOR SUBCUTANEOUS IMPLANTS.
ATE260640T1 (en) 1997-12-29 2004-03-15 Alza Corp OSMOTIC ADMINISTRATION SYSTEM WITH PLUG RETENTION MECHANISM
EP1300173B1 (en) 1997-12-29 2006-06-14 Alza Corporation Implanter device for subcutaneous implants
EP1041974B1 (en) 1997-12-30 2006-10-11 Alza Corporation Beneficial agent delivery system with membrane plug
US20040024068A1 (en) 1998-01-23 2004-02-05 Trustees Of Tufts College Antimicrobial compounds
IT1298575B1 (en) 1998-02-06 2000-01-12 Vectorpharma Int PHARMACEUTICAL COMPOSITIONS IN THE FORM OF NANOPARTICLES INCLUDING LIPID SUBSTANCES AND ANTIPHILIC SUBSTANCES AND RELATED PROCESS OF
US6017545A (en) 1998-02-10 2000-01-25 Modi; Pankaj Mixed micellar delivery system and method of preparation
AU759058C (en) 1998-02-13 2005-09-15 Amylin Pharmaceuticals, Inc. Inotropic and diuretic effects of exendin and GLP-1
US6703359B1 (en) 1998-02-13 2004-03-09 Amylin Pharmaceuticals, Inc. Inotropic and diuretic effects of exendin and GLP-1
USD408917S (en) 1998-02-26 1999-04-27 Minnesota Mining And Manufacturing Company Membrane support structure of a flow through cell for blood gas measurement
US6224577B1 (en) 1998-03-02 2001-05-01 Medrad, Inc. Syringes and plungers for use therein
US6056718A (en) 1998-03-04 2000-05-02 Minimed Inc. Medication infusion set
US6245357B1 (en) 1998-03-06 2001-06-12 Alza Corporation Extended release dosage form
US6183461B1 (en) 1998-03-11 2001-02-06 Situs Corporation Method for delivering a medication
US20020136848A1 (en) 1998-03-12 2002-09-26 Fumio Yoshii Lactone-containing resin composition, molded object thereof, and film
US6029361A (en) 1998-03-25 2000-02-29 Ultratech Stepper, Inc. Air-guage nozzle probe structure for microlithographic image focusing
US6074660A (en) 1998-04-20 2000-06-13 Ethicon, Inc. Absorbable polyoxaesters containing amines and/ or amido groups
TW586944B (en) 1998-05-29 2004-05-11 Sumitomo Pharma Controlled release agent having a multi-layer structure
US8626302B2 (en) 1998-06-03 2014-01-07 Spr Therapeutics, Llc Systems and methods to place one or more leads in muscle for providing electrical stimulation to treat pain
CA2334872C (en) 1998-06-12 2014-08-19 Bionebraska, Inc. Use of exendin-4 to treat impaired glucose tolerance
DE69940735D1 (en) 1998-07-17 2009-05-28 Pacira Pharmaceuticals Inc BIODEGRADABLE ARRANGEMENTS FOR THE CONTROLLED RELEASE OF INCLUDED SUBSTANCES
US7390637B2 (en) 1998-07-21 2008-06-24 Human Genome Sciences, Inc. Keratinocyte derived interferon
US6472512B1 (en) 1998-07-21 2002-10-29 Human Genome Sciences, Inc. Keratinocyte derived interferon
US6270700B1 (en) 1998-07-23 2001-08-07 Societe De Conseils De Recherches Et D'applications Scientifiques, Sas Encapsulation of water soluble peptides
USD415073S (en) 1998-08-17 1999-10-12 ScooterBug, Inc. Stroller
US6720407B1 (en) 1998-08-28 2004-04-13 Eli Lilly And Company Method for administering insulinotropic peptides
US6551613B1 (en) 1998-09-08 2003-04-22 Alza Corporation Dosage form comprising therapeutic formulation
US6174547B1 (en) 1999-07-14 2001-01-16 Alza Corporation Dosage form comprising liquid formulation
EP1112060B1 (en) 1998-09-09 2005-12-07 ALZA Corporation Dosage form comprising liquid formulation
US6248112B1 (en) 1998-09-30 2001-06-19 C. R. Bard, Inc. Implant delivery system
US6284725B1 (en) 1998-10-08 2001-09-04 Bionebraska, Inc. Metabolic intervention with GLP-1 to improve the function of ischemic and reperfused tissue
HUP0104993A3 (en) 1998-11-02 2003-02-28 Alza Corp Mountain View Method and device for controlled delivery of active agents
WO2000029206A1 (en) 1998-11-13 2000-05-25 Sensor Technologies Inc. Monodisperse preparations useful with implanted devices
US20030060425A1 (en) 1998-11-24 2003-03-27 Ahlem Clarence N. Immune modulation method using steroid compounds
EP2322545A1 (en) 1998-12-07 2011-05-18 Ipsen Pharma Analogues of GLP-1
CN1161101C (en) 1998-12-17 2004-08-11 阿尔扎有限公司 Conversion of liquid filled gelatin capsules into controlled release systems by multiple coatings
DK1140018T3 (en) 1998-12-23 2004-02-02 Amgen Inc Polyol / oil suspensions for sustained release of proteins
US6433144B1 (en) 1999-01-12 2002-08-13 Viragen, Inc. Compositions of highly-purified natural mixtures of type I Interferon derived from leukocytes and methods
EP1140144A4 (en) 1998-12-31 2002-10-30 Viragen Inc Composition of highly purified natural mixtures of type i interferon derived from leukocytes and methods
DE69906132T2 (en) 1998-12-31 2003-12-18 Alza Corp OSMOTIC ADMINISTRATION SYSTEM WITH SPILLAGE PISTONS
WO2000040273A2 (en) 1999-01-08 2000-07-13 Vical Incorporated Treatment of viral diseases using an interferon omega expressing polynucleotide
US6703225B1 (en) 1999-01-12 2004-03-09 Sumitomo Pharmaceuticals Company, Limited Interferon-α
US7399489B2 (en) 1999-01-14 2008-07-15 Amylin Pharmaceuticals, Inc. Exendin analog formulations
US6902744B1 (en) 1999-01-14 2005-06-07 Amylin Pharmaceuticals, Inc. Exendin agonist formulations and methods of administration thereof
PT1143989E (en) 1999-01-14 2007-03-30 Amylin Pharmaceuticals Inc Exendins for glucagon suppression
KR100858856B1 (en) 1999-02-08 2008-09-17 인타르시아 세라퓨틱스 인코포레이티드 Stable non-aqueous single phase viscous vehicles and formulations utilizing such vehicles
US7919109B2 (en) 1999-02-08 2011-04-05 Intarcia Therapeutics, Inc. Stable non-aqueous single phase viscous vehicles and formulations utilizing such vehicles
US7258869B1 (en) 1999-02-08 2007-08-21 Alza Corporation Stable non-aqueous single phase viscous vehicles and formulations utilizing such vehicle
US6451974B1 (en) 1999-03-17 2002-09-17 Novo Nordisk A/S Method of acylating peptides and novel acylating agents
US6835194B2 (en) 1999-03-18 2004-12-28 Durect Corporation Implantable devices and methods for treatment of pain by delivery of fentanyl and fentanyl congeners
US6541021B1 (en) 1999-03-18 2003-04-01 Durect Corporation Devices and methods for pain management
JP2002540850A (en) 1999-04-05 2002-12-03 ファーマシューティカル ディスカバリー コーポレイション Method for fine powder formation
GB9907658D0 (en) 1999-04-06 1999-05-26 Zeneca Ltd Chemical compounds
CO5241354A1 (en) 1999-04-19 2003-01-31 Schering Corp HCV COMBINATION THERAPY
US6924264B1 (en) 1999-04-30 2005-08-02 Amylin Pharmaceuticals, Inc. Modified exendins and exendin agonists
US6291013B1 (en) 1999-05-03 2001-09-18 Southern Biosystems, Inc. Emulsion-based processes for making microparticles
CA2369594A1 (en) 1999-05-07 2000-11-16 Pharmasol Gmbh Lipid particles on the basis of mixtures of liquid and solid lipids and method for producing same
US6849714B1 (en) 1999-05-17 2005-02-01 Conjuchem, Inc. Protection of endogenous therapeutic peptides from peptidase activity through conjugation to blood components
US6514500B1 (en) 1999-10-15 2003-02-04 Conjuchem, Inc. Long lasting synthetic glucagon like peptide {GLP-!}
US6887470B1 (en) 1999-09-10 2005-05-03 Conjuchem, Inc. Protection of endogenous therapeutic peptides from peptidase activity through conjugation to blood components
US6329336B1 (en) 1999-05-17 2001-12-11 Conjuchem, Inc. Long lasting insulinotropic peptides
US6506724B1 (en) 1999-06-01 2003-01-14 Amylin Pharmaceuticals, Inc. Use of exendins and agonists thereof for the treatment of gestational diabetes mellitus
US6485706B1 (en) 1999-06-04 2002-11-26 Delrx Pharmaceutical Corp. Formulations comprising dehydrated particles of pharma-ceutical agents and process for preparing the same
HUP0201626A3 (en) 1999-06-04 2004-05-28 Alza Corp Mountain View Implantable gel compositions and method of manufacture
US20030059376A1 (en) 1999-06-04 2003-03-27 Libbey Miles A. Formulations comprising dehydrated particles of pharmaceutical agents and process for preparing the same
US20010040326A1 (en) 1999-06-14 2001-11-15 Lord Corporation Resilient member with deformed element and method of forming same
US6833256B1 (en) 1999-06-22 2004-12-21 University Of Maryland Interferon tau mutants and methods for making them
US6528486B1 (en) 1999-07-12 2003-03-04 Zealand Pharma A/S Peptide agonists of GLP-1 activity
USD445975S1 (en) 1999-08-27 2001-07-31 Black & Decker, Inc. Base unit for a hand held vacuum cleaner
SE9903236D0 (en) 1999-09-10 1999-09-10 Astra Ab Method of obtaining microparticles
US6458387B1 (en) 1999-10-18 2002-10-01 Epic Therapeutics, Inc. Sustained release microspheres
US6284283B1 (en) 1999-10-21 2001-09-04 Alkermes Controlled Therapeutics, Inc. Method of producing sub-micron particles of biologically active agents and uses thereof
US6436091B1 (en) 1999-11-16 2002-08-20 Microsolutions, Inc. Methods and implantable devices and systems for long term delivery of a pharmaceutical agent
US7022674B2 (en) 1999-12-16 2006-04-04 Eli Lilly And Company Polypeptide compositions with improved stability
WO2001043528A2 (en) 1999-12-17 2001-06-21 Durect Corporation Devices and methods in intracerebrospinal delivery of morphine-6-glucuronide
EP1328256B1 (en) 1999-12-21 2005-10-19 Alza Corporation Valve for osmotic devices
US6498193B2 (en) 1999-12-22 2002-12-24 Trustees Of Dartmouth College Treatment for complications of type 2 diabetes
US6283949B1 (en) 1999-12-27 2001-09-04 Advanced Cardiovascular Systems, Inc. Refillable implantable drug delivery pump
US6572890B2 (en) 2000-01-13 2003-06-03 Osmotica Corp. Osmotic device containing venlafaxine and an anti-psychotic agent
US6472060B1 (en) 2000-01-19 2002-10-29 Seco Tools Ab Coated body with nanocrystalline CVD coating for enhanced edge toughness and reduced friction
US6844321B2 (en) 2000-01-31 2005-01-18 Novo Nordisk A/S Crystallization of a GLP-1 analogue
US6465425B1 (en) 2000-02-10 2002-10-15 Alkermes Controlled Therapeutics, Inc. Microencapsulation and sustained release of biologically active acid-stable or free sulfhydryl-containing proteins
US6464688B1 (en) 2000-02-15 2002-10-15 Microsolutions, Inc. Osmotic pump delivery system with flexible drug compartment
US6471688B1 (en) 2000-02-15 2002-10-29 Microsolutions, Inc. Osmotic pump drug delivery systems and methods
AU2001252201A1 (en) 2000-03-14 2001-09-24 Amylin Pharmaceuticals, Inc. Effects of glucagon-like peptide-1 (7-36) on antro-pyloro-duodenal motility
US20030211974A1 (en) 2000-03-21 2003-11-13 Brodbeck Kevin J. Gel composition and methods
TWI250874B (en) 2000-03-24 2006-03-11 Nat Health Research Institutes Pharmaceutical compositions for preventing or treating disorders associated with bacterial or viral infection
ATE309822T1 (en) 2000-04-19 2005-12-15 Genentech Inc SUSTAINED RELEASE FORMULATIONS CONTAINING GROWTH HORMONE
WO2001080897A2 (en) 2000-04-21 2001-11-01 Vical Incorporated Compositions and methods for in vivo delivery of polynucleotide-based therapeutics
US6706689B2 (en) 2000-05-19 2004-03-16 Amylin Pharmaceuticals, Inc. Treatment of acute coronary syndrome with GLP-1
US6495164B1 (en) 2000-05-25 2002-12-17 Alkermes Controlled Therapeutics, Inc. I Preparation of injectable suspensions having improved injectability
EP1294757B1 (en) 2000-06-16 2006-11-22 Eli Lilly And Company Glucagon-like peptide-1 analogs
US6479065B2 (en) 2000-08-10 2002-11-12 Alkermes Controlled Therapeutics, Inc. Process for the preparation of polymer-based sustained release compositions
US6547250B1 (en) 2000-08-21 2003-04-15 Westport Research Inc. Seal assembly with two sealing mechanisms for providing static and dynamic sealing
US6824822B2 (en) 2001-08-31 2004-11-30 Alkermes Controlled Therapeutics Inc. Ii Residual solvent extraction method and microparticles produced thereby
WO2002028366A2 (en) 2000-10-06 2002-04-11 Durect Corporation Devices and methods for management of inflammation
CN100420482C (en) 2000-11-03 2008-09-24 精达制药公司 Method for short-term and long-term drug dosimetry
WO2002067895A2 (en) 2000-11-16 2002-09-06 Durect Corporation Implant dosage form and use thereof for the delivery of a cholesterol lowering agent
EP1339449A2 (en) 2000-11-29 2003-09-03 Durect Corporation Devices and methods for controlled delivery from a drug delivery device
US20020165286A1 (en) 2000-12-08 2002-11-07 Hanne Hedeman Dermal anti-inflammatory composition
EP1351984A2 (en) 2000-12-13 2003-10-15 Eli Lilly And Company Amidated glucagon-like peptide-1
EP2186824A3 (en) 2000-12-13 2010-09-22 Eli Lilly & Company Chronic treatment regimen using glucagon-like insulinotropic peptides
BR0116206A (en) 2000-12-14 2003-12-23 Amylin Pharmaceuticals Inc Yy Peptide and yy Peptide Agonists for Treatment of Metabolic Disorders
DK1343480T4 (en) 2000-12-21 2016-04-18 Alrise Biosystems Gmbh Induced phase transition method for the preparation of microparticles containing hydrophobic active agents.
USD472896S1 (en) 2000-12-23 2003-04-08 Andreas Peiker Telephone support
IN188924B (en) 2001-03-01 2002-11-23 Bharat Serums & Vaccines Ltd
US20030009145A1 (en) 2001-03-23 2003-01-09 Struijker-Boudier Harry A.J. Delivery of drugs from sustained release devices implanted in myocardial tissue or in the pericardial space
US6632217B2 (en) 2001-04-19 2003-10-14 Microsolutions, Inc. Implantable osmotic pump
US7144863B2 (en) 2001-06-01 2006-12-05 Eli Lilly And Company GLP-1 formulations with protracted time action
US6514517B2 (en) 2001-06-20 2003-02-04 Ethicon, Inc. Antimicrobial coatings for medical devices
WO2003000282A1 (en) 2001-06-21 2003-01-03 Genentech, Inc. Sustained release formulation
CA2448864C (en) 2001-06-22 2008-04-22 Pfizer Products Inc. Pharmaceutical compositions containing a solid dispersion of a poorly-soluble drug in a matrix and a solubility-enhancing polymer
JP4758607B2 (en) 2001-06-22 2011-08-31 ジョンズ ホプキンズ ユニヴァーシティー スクール オブ メディシン Biodegradable polymer composition and method of use thereof
US7163688B2 (en) 2001-06-22 2007-01-16 Alza Corporation Osmotic implant with membrane and membrane retention means
US20030138403A1 (en) 2001-06-29 2003-07-24 Maxygen Aps Interferon formulations
HUP0401156A2 (en) 2001-07-20 2004-12-28 Intermune, Inc. Methods of treating liver fibrosis
WO2003011892A2 (en) 2001-07-31 2003-02-13 The Government Of The United States Of America As Represented By The Secretary, Department Of Health And Human Services Glp-1 exendin-4 peptide analogs and uses thereof
AU2002322403A1 (en) 2001-08-23 2003-03-10 Eli Lilly And Company Glucagon-like peptide-1 analogs
GB0121709D0 (en) 2001-09-07 2001-10-31 Imp College Innovations Ltd Food inhibition agent
WO2003024357A2 (en) 2001-09-14 2003-03-27 Martin Francis J Microfabricated nanopore device for sustained release of therapeutic agent
ES2333414T3 (en) 2001-09-17 2010-02-22 Durect Corporation DEVICE AND METHOD FOR THE PRECISE DELIVERY OF AN ACTIVE AGENT.
US7459432B2 (en) 2001-09-24 2008-12-02 Imperial College Innovations Ltd. Modification of feeding behavior
IL160965A0 (en) 2001-10-05 2004-08-31 Intermune Inc Method of treating hepatitis virus infection with a multiphasic interferon delivery profile
US7041646B2 (en) 2001-10-05 2006-05-09 Bayer Pharmaceuticals Corporation Methods of treating type 2 diabetes with peptides acting as both GLP-1 receptor agonists and glucagon receptor antagonists
WO2003034960A1 (en) 2001-10-24 2003-05-01 Med-El Elektromedizinische Geräte Ges.m.b.H. Implantable fluid delivery apparatuses and implantable electrode
US20040142902A1 (en) 2001-11-08 2004-07-22 Struijker- Boudier Harry A.J. Implant dosage form and use thereof for the delivery of a cholosterol lowering agent
IL161889A0 (en) 2001-11-09 2005-11-20 Biomedicines Inc Method for treating diseases with omega interferon
BR0206469A (en) 2001-11-14 2004-01-13 Alza Corp Injectable Depot Composition
KR20040058101A (en) 2001-11-14 2004-07-03 알자 코포레이션 Catheter injectable depot compositions and uses thereof
US20030170289A1 (en) 2001-11-14 2003-09-11 Guohua Chen Injectable depot compositions and uses thereof
DE10159217A1 (en) 2001-11-27 2003-06-05 Schering Ag 17alpha-alkyl-17ß-oxy-estratrienes and intermediates for their preparation, use of 17alpha-alkyl-17ß-oxy-estratriene for the preparation of medicaments and pharmaceutical preparations
US20030108608A1 (en) 2001-12-12 2003-06-12 Erik Laridon Thermoplastic articles comprising silver-containing antimicrobials and high amounts of carboxylic acid salts for increased surface-available silver
NZ533062A (en) 2001-12-19 2006-03-31 Alza Corp Formulation and dosage form for the controlled delivery of therapeutic agents
EP2277910A1 (en) 2001-12-21 2011-01-26 Human Genome Sciences, Inc. Albumin fusion proteins
US8058233B2 (en) 2002-01-10 2011-11-15 Oregon Health And Science University Modification of feeding behavior using PYY and GLP-1
US7105489B2 (en) 2002-01-22 2006-09-12 Amylin Pharmaceuticals, Inc. Methods and compositions for treating polycystic ovary syndrome
US20040028733A1 (en) 2002-02-08 2004-02-12 Alkermes Controlled Therapeutics, Inc. Polymer-based compositions for sustained release
US7635463B2 (en) 2002-02-27 2009-12-22 Pharmain Corporation Compositions for delivery of therapeutics and other materials
WO2003072143A1 (en) 2002-02-27 2003-09-04 Pharmain, Ltd. Compositions for delivery of therapeutics and other materials, and methods of making and using the same
GB2386066A (en) 2002-02-28 2003-09-10 Norbrook Lab Ltd Long-acting parasiticidal composition with improved bioavailability comprising a salicylanilide, a further anti-parasitic compound & a polymeric species
GB0204722D0 (en) 2002-02-28 2002-04-17 Norferm Da Method
WO2003087335A2 (en) 2002-04-11 2003-10-23 Medimmune Vaccines, Inc. Preservation of bioactive materials by spray drying
ES2665999T3 (en) 2002-05-31 2018-04-30 Titan Pharmaceuticals, Inc. Implantable polymeric device for sustained release of buprenorphine
US7074423B2 (en) 2002-06-17 2006-07-11 Alza Corporation Osmotic delivery system with early zero order push power engine
US20040001889A1 (en) 2002-06-25 2004-01-01 Guohua Chen Short duration depot formulations
KR101046903B1 (en) 2002-06-26 2011-07-06 인타르시아 세라퓨틱스 인코포레이티드 Minimal bouncing volumetric efficiency piston for osmotic drug delivery systems
US7177526B2 (en) 2002-06-28 2007-02-13 Intel Corporation System and method for improving audio during post-production of video recordings
CN1684663A (en) 2002-07-31 2005-10-19 阿尔萨公司 Injectable multimodal polymer depot compositions and uses thereof
CN1668276A (en) 2002-07-31 2005-09-14 阿尔萨公司 Injectable depot compositions and uses thereof
US20080260838A1 (en) * 2003-08-01 2008-10-23 Mannkind Corporation Glucagon-like peptide 1 (glp-1) pharmaceutical formulations
AU2003267327A1 (en) 2002-09-04 2004-03-29 Ruark Botha Device for securing a blood vessel cannula to a body
WO2004035754A2 (en) 2002-10-17 2004-04-29 Alkermes Controlled Therapeutics, Inc. Ii Microencapsulation and sustained release of biologically active polypeptides
EP1567127A4 (en) 2002-10-17 2007-02-21 Alkermes Inc Method of modifying the release profile of sustained release compositions
US20040209801A1 (en) 2002-10-22 2004-10-21 Brand Stephen J. Treatment of diabetes
CA2504608C (en) 2002-11-06 2013-01-08 Alza Corporation Controlled release depot formulations
US6969702B2 (en) 2002-11-20 2005-11-29 Neuronova Ab Compounds and methods for increasing neurogenesis
US7014636B2 (en) 2002-11-21 2006-03-21 Alza Corporation Osmotic delivery device having a two-way valve and a dynamically self-adjusting flow channel
US7790681B2 (en) 2002-12-17 2010-09-07 Amylin Pharmaceuticals, Inc. Treatment of cardiac arrhythmias with GLP-1 receptor ligands
JP2006512370A (en) 2002-12-19 2006-04-13 アルザ・コーポレーション A stable non-aqueous single phase gel and its formulation for delivery from an implantable device
US7731947B2 (en) 2003-11-17 2010-06-08 Intarcia Therapeutics, Inc. Composition and dosage form comprising an interferon particle formulation and suspending vehicle
GB0300571D0 (en) 2003-01-10 2003-02-12 Imp College Innovations Ltd Modification of feeding behaviour
JP2004238392A (en) 2003-01-14 2004-08-26 Nipro Corp Stabilized proteinic preparation
WO2004075349A2 (en) 2003-02-18 2004-09-02 Medconx, Inc. Male medical device electrical connector with engineered friction fit
AU2004227837A1 (en) 2003-03-31 2004-10-21 Alza Corporation Non-aqueous single phase vehicles and formulations utilizing such vehicles
JP2006521897A (en) 2003-03-31 2006-09-28 アルザ・コーポレーション Osmotic pump with means for dissipating internal pressure
MXPA05010604A (en) 2003-03-31 2005-11-23 Alza Corp Osmotic delivery system and method for decreasing start-up times for osmotic delivery systems.
EP1626752A2 (en) 2003-05-16 2006-02-22 Blue Membranes GmbH Medical implants comprising biocompatible coatings
US20040247672A1 (en) 2003-05-16 2004-12-09 Alkermes Controlled Therapeutics, Inc. Injectable sustained release compositions
WO2005000222A2 (en) 2003-05-30 2005-01-06 Amylin Pharmaceuticals, Inc. Novel methods and compositions for enhanced transmucosal delivery of peptides and proteins
US20050079202A1 (en) 2003-05-30 2005-04-14 Guohua Chen Implantable elastomeric depot compositions and uses thereof
JP4936884B2 (en) 2003-06-03 2012-05-23 ノボ・ノルデイスク・エー/エス Stabilized pharmaceutical peptide composition
US8491571B2 (en) 2003-06-12 2013-07-23 Cordis Corporation Orifice device having multiple channels with varying flow rates for drug delivery
US7454765B2 (en) 2003-07-09 2008-11-18 Samsung Electronics Co., Ltd. Optical disc drive
US7205409B2 (en) 2003-09-04 2007-04-17 Abbott Laboratories Pharmaceutical compositions as inhibitors of dipeptidyl peptidase-IV (DPP-IV)
US9005244B2 (en) 2003-09-30 2015-04-14 Ethicon, Inc. Tissue approximation device
BRPI0414941A (en) 2003-09-30 2006-11-07 Alza Corp osmotically driven active agent dispensing device providing an upward release profile
BRPI0416094A (en) 2003-10-31 2007-01-02 Alza Corp self-retaining quick start membrane plug osmotic pump
WO2005046639A2 (en) 2003-11-06 2005-05-26 Alza Corporation Modular imbibition rate reducer for use with implantable osmotic pump
US20050118206A1 (en) 2003-11-14 2005-06-02 Luk Andrew S. Surfactant-based gel as an injectable, sustained drug delivery vehicle
US20050106214A1 (en) 2003-11-14 2005-05-19 Guohua Chen Excipients in drug delivery vehicles
US20050281879A1 (en) 2003-11-14 2005-12-22 Guohua Chen Excipients in drug delivery vehicles
US7780973B2 (en) 2003-12-15 2010-08-24 Ethicon Endo-Surgery, Inc. Method and device for minimally invasive implantation of biomaterial
US20050216087A1 (en) 2004-01-05 2005-09-29 St. Francis Medical Technologies, Inc. Disk repair structures for positioning disk repair material
US20050175701A1 (en) 2004-02-10 2005-08-11 Alza Corporation Capillary moderator for osmotic delivery system
US8076288B2 (en) 2004-02-11 2011-12-13 Amylin Pharmaceuticals, Inc. Hybrid polypeptides having glucose lowering activity
US20060094652A1 (en) 2004-02-11 2006-05-04 Levy Odile E Hybrid polypeptides with selectable properties
SI2409707T1 (en) 2004-04-15 2015-07-31 Alkermes Pharma Ireland Limited Polymer-based sustained release device
US7456254B2 (en) 2004-04-15 2008-11-25 Alkermes, Inc. Polymer-based sustained release device
WO2005112977A2 (en) 2004-04-23 2005-12-01 Pharmain, Ltd. Compositions for treatment with glucagon-like peptide, and methods of making and using the same
US20050266087A1 (en) 2004-05-25 2005-12-01 Gunjan Junnarkar Formulations having increased stability during transition from hydrophobic vehicle to hydrophilic medium
US7772182B2 (en) 2004-08-05 2010-08-10 Alza Corporation Stable suspension formulations of erythropoietin receptor agonists
JP5244388B2 (en) 2004-08-18 2013-07-24 ウオーターズ・テクノロジーズ・コーポレイシヨン Apparatus and method for generating or carrying fluid under pressure and seal member used in the apparatus
US8268791B2 (en) 2004-08-25 2012-09-18 Aegis Therapeutics, Llc. Alkylglycoside compositions for drug administration
US20060094693A1 (en) 2004-09-21 2006-05-04 Point Therapeutics, Inc. Methods and compositions for treating glucose-associated conditions, metabolic syndrome, dyslipidemias and other conditions
US20080038316A1 (en) 2004-10-01 2008-02-14 Wong Vernon G Conveniently implantable sustained release drug compositions
CN101890167B (en) 2004-10-01 2017-03-01 拉姆斯科股份有限公司 The sustained release pharmaceutical composition that can conveniently implant
US7442682B2 (en) 2004-10-19 2008-10-28 Nitto Denko Corporation Transepithelial delivery of peptides with incretin hormone activities
ATE427759T1 (en) 2004-11-01 2009-04-15 Amylin Pharmaceuticals Inc TREATMENT OF OBESITY AND RELATED DISEASES
US8394765B2 (en) 2004-11-01 2013-03-12 Amylin Pharmaceuticals Llc Methods of treating obesity with two different anti-obesity agents
US7575579B2 (en) 2004-11-18 2009-08-18 Union Surgical, Llc Drill guide tissue protector
US20060142234A1 (en) 2004-12-23 2006-06-29 Guohua Chen Injectable non-aqueous suspension
US20060141040A1 (en) 2004-12-23 2006-06-29 Guohua Chen Injectable non-aqueous suspension
EP1841448A2 (en) 2004-12-30 2007-10-10 Diakine Therapeutics, Inc. Pharmaceutical compositions and methods for restoring beta-cell mass and function
USD587374S1 (en) 2005-01-14 2009-02-24 Edwards Lifesciences Corporation Sensor case
CN101141994B (en) 2005-01-24 2010-10-13 欧加农股份有限公司 Applicator for inserting an implant
US7488316B2 (en) 2005-01-25 2009-02-10 Microchips, Inc. Control of drug release by transient modification of local microenvironments
WO2006083761A2 (en) 2005-02-03 2006-08-10 Alza Corporation Solvent/polymer solutions as suspension vehicles
US20060216242A1 (en) 2005-02-03 2006-09-28 Rohloff Catherine M Suspending vehicles and pharmaceutical suspensions for drug dosage forms
US20060263433A1 (en) 2005-02-03 2006-11-23 Ayer Rupal A Suspension formulation of interferon
US20070032420A1 (en) 2005-02-09 2007-02-08 Entelos, Inc. Treating diabetes with glucagon-like peptide-1 secretagogues
WO2006086769A2 (en) 2005-02-11 2006-08-17 Amylin Pharmaceuticals, Inc. Gip analog and hybrid polypeptides with selectable properties
US8263545B2 (en) 2005-02-11 2012-09-11 Amylin Pharmaceuticals, Inc. GIP analog and hybrid polypeptides with selectable properties
US20090286723A1 (en) 2005-02-11 2009-11-19 Amylin Pharmaceuticals, Inc. Hybrid Polypeptides with Selectable Properties
GB0504857D0 (en) 2005-03-09 2005-04-13 Imp College Innovations Ltd Novel compounds and their effects on feeding behaviour
US7959938B2 (en) 2005-03-15 2011-06-14 Intarcia Therapeutics, Inc. Polyoxaester suspending vehicles for use with implantable delivery systems
WO2006105527A2 (en) 2005-03-31 2006-10-05 Amylin Pharmaceuticals, Inc. Amylin and amylin agonists for treating psychiatric diseases and disorders
ATE509634T1 (en) 2005-04-08 2011-06-15 Amylin Pharmaceuticals Inc PHARMACEUTICAL FORMULATIONS CONTAINING INCRETIN PEPTIDE AND APROTIC-POLAR SOLVENT
WO2006111169A1 (en) 2005-04-21 2006-10-26 Gastrotech Pharma A/S Pharmaceutical preparations of a glp-1 molecule and an anti-emetic drug
US7955305B2 (en) 2005-05-06 2011-06-07 Medtronic Minimed, Inc. Needle inserter and method for infusion device
PL1888031T3 (en) 2005-06-06 2013-04-30 Camurus Ab Glp-1 analogue formulations
US20060280795A1 (en) 2005-06-08 2006-12-14 Dexcel Pharma Technologies, Ltd. Specific time-delayed burst profile delivery system
US9447781B2 (en) 2005-07-22 2016-09-20 University Of Utah Research Foundation Osmotically driven dispense pump and related components for use in high pressure applications
US20070027105A1 (en) 2005-07-26 2007-02-01 Alza Corporation Peroxide removal from drug delivery vehicle
US8389472B2 (en) 2005-08-19 2013-03-05 Amylin Pharmaceuticals, Llc Exendin-4 to treat nonalcoholic steatohepatitis and nonalcoholic fatty liver disease
WO2007024700A2 (en) 2005-08-19 2007-03-01 Amylin Pharmaceuticals, Inc. Exendin for treating diabetes and reducing body weight
PL1965823T3 (en) 2005-11-04 2017-08-31 Glaxosmithkline Llc Methods for administering hypoglycemic agents
US8039432B2 (en) 2005-11-09 2011-10-18 Conjuchem, Llc Method of treatment of diabetes and/or obesity with reduced nausea side effect
JP5096363B2 (en) 2005-12-16 2012-12-12 ネクター セラピューティックス GLP-1 polymer complex
AU2006329836A1 (en) 2005-12-16 2007-07-05 Amylin Pharmaceuticals, Llc Compositions and methods for treating obesity and related metabolic disorders
SI1984009T1 (en) 2006-01-18 2013-02-28 Qps, Llc Pharmaceutical compositions with enhanced stability
CN101432025B (en) 2006-03-21 2012-04-04 安米林药品公司 Peptide-peptidase inhibitor conjugates and methods of using same
EP3255149A3 (en) 2006-05-02 2018-04-18 Intrexon Actobiotics NV Microbial intestinal delivery of obesity related peptides
US8299024B2 (en) 2006-05-12 2012-10-30 Amylin Pharmaceuticals, Llc Methods to restore glycemic control
AU2007266475B2 (en) 2006-05-30 2009-12-03 Intarcia Therapeutics, Inc. Two-piece, internal-channel osmotic delivery system flow modulator
GB0613196D0 (en) 2006-07-03 2006-08-09 Imp Innovations Ltd Novel compounds and their effects on feeding behaviour
CA2657911C (en) 2006-07-11 2012-02-21 Quest Pharmaceutical Services, Llc Pharmaceutical compositions for sustained release delivery of peptides
EP2066337A2 (en) 2006-08-04 2009-06-10 Amylin Pharmaceuticals, Inc. Use of exendins, exendin agonists and glp-1 receptor agonists for altering the concentration of fibrinogen
DK2359808T3 (en) 2006-08-09 2013-08-05 Intarcia Therapeutics Inc Osmotic delivery systems and piston assemblies
MX2009003737A (en) 2006-10-05 2009-06-16 Panacea Biotec Ltd Injectable depot composition and it's process of preparation.
WO2008061355A1 (en) 2006-11-24 2008-05-29 Matregen Corp. Glp-1 depot systems, and methods of manufacture and uses thereof
TWI428346B (en) 2006-12-13 2014-03-01 Imp Innovations Ltd Novel compounds and their effects on feeding behaviour
WO2008086086A2 (en) 2007-01-05 2008-07-17 Indiana University Research And Technology Corporation Glucagon analogs exhibiting enhanced solubility in physiological ph buffers
USD555589S1 (en) 2007-01-23 2007-11-20 Intec, Inc. Battery charger and game controller cradle
US20090022727A1 (en) 2007-01-26 2009-01-22 Alza Corp. Injectable, nonaqueous suspension with high concentration of therapeutic agent
EP1958576A3 (en) 2007-02-13 2009-08-05 Arthrex Inc Double cut shaver
US8262667B1 (en) 2007-02-23 2012-09-11 Holmed Corporation Multi-diameter implant forceps
MX2009011123A (en) 2007-04-23 2009-11-02 Intarcia Therapeutics Inc Suspension formulations of insulinotropic peptides and uses thereof.
WO2008134425A1 (en) 2007-04-27 2008-11-06 Cedars-Sinai Medical Center Use of glp-1 receptor agonists for the treatment of gastrointestinal disorders
WO2009100216A1 (en) 2008-02-08 2009-08-13 Qps Llc Composition for sustained release delivery of proteins or peptides
US9186502B2 (en) 2008-02-14 2015-11-17 Enteromedics Inc. Treatment of excess weight by neural downregulation in combination with compositions
US20090234392A1 (en) 2008-03-13 2009-09-17 Depuy Spine, Inc. Method for inserting a spinal fixation element using implants having guide tabs
JP5792057B2 (en) 2008-04-01 2015-10-07 モサメディックス・ビー.ブイ.MosaMedix B.V. Compositions and methods for reducing scar formation in wound healing
EP2300099A1 (en) 2008-04-04 2011-03-30 Enteromedics Inc. Methods and systems for glucose regulation
US20110263496A1 (en) 2008-05-21 2011-10-27 Amylin Pharmaceuticals, Inc. Exendins to lower cholesterol and triglycerides
MX2011002836A (en) 2008-09-17 2011-04-28 Chiasma Inc Pharmaceutical compositions and related methods of delivery.
CN102202646A (en) 2008-09-30 2011-09-28 Endo药物方法有限公司 Implantable device for the delivery of risperidone and methods of use thereof
WO2010068467A1 (en) 2008-11-25 2010-06-17 Attenuex Technologies, Inc. Implant with high vapor pressure medium
US20100298840A1 (en) 2009-05-22 2010-11-25 Schwartz Lyman D Phimosis Treatment Device and Method
USD638478S1 (en) 2009-07-06 2011-05-24 Performance Designed Products Llc Video game controller accessory
US20110022165A1 (en) 2009-07-23 2011-01-27 Edwards Lifesciences Corporation Introducer for prosthetic heart valve
US9358064B2 (en) 2009-08-07 2016-06-07 Ulthera, Inc. Handpiece and methods for performing subcutaneous surgery
USD789539S1 (en) 2015-07-09 2017-06-13 Spinal Surgical Strategies, Llc Fusion cage
ES2650667T3 (en) * 2009-09-28 2018-01-19 Intarcia Therapeutics, Inc Rapid establishment and / or termination of substantial steady state drug administration
JP5411366B2 (en) * 2009-12-16 2014-02-12 ノヴォ ノルディスク アー/エス Double acylated GLP-1 derivative
AR079344A1 (en) 2009-12-22 2012-01-18 Lilly Co Eli PEPTIDAL ANALOG OF OXINTOMODULIN, PHARMACEUTICAL COMPOSITION THAT UNDERSTANDS AND USES TO PREPARE A USEFUL MEDICINAL PRODUCT TO TREAT NON-INSULINED INDEPENDENT DIABETES AND / OR OBESITY
JO2976B1 (en) 2009-12-22 2016-03-15 ايلي ليلي اند كومباني Oxyntomodulin peptide analogue ‎
WO2011156407A2 (en) 2010-06-09 2011-12-15 Amylin Pharmaceuticals, Inc. Glp-1 receptor agonists to treat pancre-atitis
USD669589S1 (en) 2010-08-31 2012-10-23 Koninklijke Philips Electronics N.V. Blood system cartridge
US20120208755A1 (en) 2011-02-16 2012-08-16 Intarcia Therapeutics, Inc. Compositions, Devices and Methods of Use Thereof for the Treatment of Cancers
EP2729493B1 (en) 2011-07-04 2020-06-10 IP2IPO Innovations Limited Novel compounds and their effects on feeding behaviour
USD678889S1 (en) 2011-10-04 2013-03-26 Avision Inc. Scanner docking station
ES2699655T3 (en) 2012-04-19 2019-02-12 Removaid As Tool to remove an implanted article under the skin
WO2013180126A1 (en) 2012-05-29 2013-12-05 国立大学法人高知大学 Artery visualization device and artery imaging device
US20140058425A1 (en) 2012-08-27 2014-02-27 Amir Porat Manually operated surgical devices with operative portions formed of a see-through material
US9332995B2 (en) 2012-09-25 2016-05-10 Russo Inventions, Llc Bone-harvesting tool
US9241722B2 (en) 2012-10-08 2016-01-26 Warsaw Orthopedic, Inc. Surgical pin guide and methods of use
WO2014078832A1 (en) 2012-11-19 2014-05-22 Endo Pharmaceuticals Solutions Inc. Implantable drug delivery compositions and methods of treatment thereof
USD731630S1 (en) 2014-04-08 2015-06-09 Bootz Manufacturing Company Shower pan
USD724740S1 (en) 2014-06-05 2015-03-17 Deka Products Limited Partnership Enclosure for a peritoneal dialysis device
US9889085B1 (en) 2014-09-30 2018-02-13 Intarcia Therapeutics, Inc. Therapeutic methods for the treatment of diabetes and related conditions for patients with high baseline HbA1c
MA44390A (en) 2015-06-03 2019-01-23 Intarcia Therapeutics Inc IMPLANT PLACEMENT AND REMOVAL SYSTEMS
USD789540S1 (en) 2016-01-29 2017-06-13 Veterinary Implants Direct, Llc. Orthopedic plate
US9931133B2 (en) 2016-03-24 2018-04-03 A.M. Surgical, Inc. Compact endoscopic surgical device and method of use thereof
WO2017200943A1 (en) 2016-05-16 2017-11-23 Intarcia Therapeutics, Inc. Glucagon-receptor selective polypeptides and methods of use thereof
US10835580B2 (en) 2017-01-03 2020-11-17 Intarcia Therapeutics, Inc. Methods comprising continuous administration of a GLP-1 receptor agonist and co-administration of a drug

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090042781A1 (en) * 2004-06-28 2009-02-12 Novo Nordisk A/S Methods for Treating Diabetes
US20090202608A1 (en) * 2008-02-13 2009-08-13 Alessi Thomas R Devices, formulations, and methods for delivery of multiple beneficial agents
WO2009109927A2 (en) * 2008-03-05 2009-09-11 Tel Hashomer Medical Research Infrastructure And Services Ltd. Glp-1 receptor agonists and related active pharmaceutical ingredients for treatment of cancer
US20100092566A1 (en) * 2008-10-15 2010-04-15 Alessi Thomas R Highly concentrated drug particles, formulations, suspensions and uses thereof

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
Jordan et al ("Guidelines for Antiemetic Treatment of Chemotherapy-Induced Nausea and Vomiting: Past, Present and Future Recommendations (2007) The Oncologist 12(9) 1143-1150) *
Quianzon, Celeste C. et al "Lixisenatide - Once-daily Glucagon-like Peptide-1 Receptor Agonist in the Management of Type 2 Diabetes (2011) US Endocrinology, Volume 7(2), pages 104-109. *
Ratner et al ("Dose-dependent effects of the one-daily GLP-1 receptor agonist lixisenatide in patients with Type 2 diabetes inadequately controlled with metformin: a randomized, double-blind, placebo-controlled trial" (September 2010) Diabetic Medicine 27(9): 1024-1032). *
Sequence Listings for (WO2009109927) at WIPO Patentscope Website, http://patentscope.wipo.int/search/docservicepdf_pct/id00000008776887 (downloaded on November 14, 2012) *

Cited By (80)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9724293B2 (en) 2003-11-17 2017-08-08 Intarcia Therapeutics, Inc. Methods of manufacturing viscous liquid pharmaceutical formulations
US9682127B2 (en) 2005-02-03 2017-06-20 Intarcia Therapeutics, Inc. Osmotic delivery device comprising an insulinotropic peptide and uses thereof
US10363287B2 (en) 2005-02-03 2019-07-30 Intarcia Therapeutics, Inc. Method of manufacturing an osmotic delivery device
US11246913B2 (en) 2005-02-03 2022-02-15 Intarcia Therapeutics, Inc. Suspension formulation comprising an insulinotropic peptide
US9526763B2 (en) 2005-02-03 2016-12-27 Intarcia Therapeutics Inc. Solvent/polymer solutions as suspension vehicles
US9539200B2 (en) 2005-02-03 2017-01-10 Intarcia Therapeutics Inc. Two-piece, internal-channel osmotic delivery system flow modulator
US10527170B2 (en) 2006-08-09 2020-01-07 Intarcia Therapeutics, Inc. Osmotic delivery systems and piston assemblies for use therein
US10441528B2 (en) 2008-02-13 2019-10-15 Intarcia Therapeutics, Inc. Devices, formulations, and methods for delivery of multiple beneficial agents
US9572889B2 (en) 2008-02-13 2017-02-21 Intarcia Therapeutics, Inc. Devices, formulations, and methods for delivery of multiple beneficial agents
US10869830B2 (en) 2009-09-28 2020-12-22 Intarcia Therapeutics, Inc. Rapid establishment and/or termination of substantial steady-state drug delivery
US10231923B2 (en) 2009-09-28 2019-03-19 Intarcia Therapeutics, Inc. Rapid establishment and/or termination of substantial steady-state drug delivery
US10159714B2 (en) 2011-02-16 2018-12-25 Intarcia Therapeutics, Inc. Compositions, devices and methods of use thereof for the treatment of cancers
US9844584B2 (en) 2012-05-11 2017-12-19 Gemvax & Kael Co., Ltd. Composition for preventing or treating sepsis
US9540419B2 (en) 2012-05-11 2017-01-10 Gemvax & Kael Co., Ltd. Anti-inflammatory peptides and composition comprising the same
US10960056B2 (en) 2012-05-11 2021-03-30 Gemvax & Kael Co., Ltd. Anti-inflammatory peptides and composition comprising the same
US9730984B2 (en) 2012-05-11 2017-08-15 Gemvax & Kael Co., Ltd. Composition for preventing or treating rheumatoid arthritis
US9907837B2 (en) 2012-05-11 2018-03-06 Gemvax & Kael Co., Ltd. Composition for preventing or treating cachexia
US11369665B2 (en) 2012-05-11 2022-06-28 Gemvax & Kael Co., Ltd. Anti-inflammatory peptides and composition comprising the same
US10039811B2 (en) 2012-05-11 2018-08-07 Gemvax & Kael Co., Ltd. Anti-inflammatory peptides and composition comprising the same
US11857607B2 (en) 2012-05-11 2024-01-02 Gemvax & Kael Co., Ltd. Anti-inflammatory peptides and composition comprising the same
US10967000B2 (en) 2012-07-11 2021-04-06 Gemvax & Kael Co., Ltd. Cell-penetrating peptide, conjugate comprising same and composition comprising same
US10758592B2 (en) 2012-10-09 2020-09-01 Sanofi Exendin-4 derivatives as dual GLP1/glucagon agonists
US9670261B2 (en) 2012-12-21 2017-06-06 Sanofi Functionalized exendin-4 derivatives
US10253079B2 (en) 2012-12-21 2019-04-09 Sanofi Functionalized Exendin-4 derivatives
US9745360B2 (en) 2012-12-21 2017-08-29 Sanofi Dual GLP1/GIP or trigonal GLP1/GIP/glucagon agonists
US9907838B2 (en) 2013-04-19 2018-03-06 Gemvax & Kael Co., Ltd. Composition and methods for treating ischemic damage
US10383926B2 (en) 2013-06-07 2019-08-20 Gemvax & Kael Co., Ltd. Biological markers useful in cancer immunotherapy
US10561703B2 (en) 2013-06-21 2020-02-18 Gemvax & Kael Co., Ltd. Method of modulating sex hormone levels using a sex hormone secretion modulator
US10034922B2 (en) 2013-11-22 2018-07-31 Gemvax & Kael Co., Ltd. Peptide having angiogenesis inhibitory activity and composition containing same
US9694053B2 (en) 2013-12-13 2017-07-04 Sanofi Dual GLP-1/glucagon receptor agonists
US9751926B2 (en) 2013-12-13 2017-09-05 Sanofi Dual GLP-1/GIP receptor agonists
US9750788B2 (en) 2013-12-13 2017-09-05 Sanofi Non-acylated exendin-4 peptide analogues
US9789165B2 (en) 2013-12-13 2017-10-17 Sanofi Exendin-4 peptide analogues as dual GLP-1/GIP receptor agonists
CN105939723A (en) * 2013-12-17 2016-09-14 杰姆维克斯&凯尔有限公司 Composition for treating prostate cancer
US11058744B2 (en) 2013-12-17 2021-07-13 Gemvax & Kael Co., Ltd. Composition for treating prostate cancer
KR102314231B1 (en) * 2013-12-17 2021-10-19 주식회사 젬백스앤카엘 Composition for treating prostate cancer
KR20160089523A (en) * 2013-12-17 2016-07-27 주식회사 젬백스앤카엘 Composition for treating prostate cancer
WO2015093854A1 (en) * 2013-12-17 2015-06-25 주식회사 카엘젬백스 Composition for treating prostate cancer
US9758561B2 (en) 2014-04-07 2017-09-12 Sanofi Dual GLP-1/glucagon receptor agonists derived from exendin-4
US9771406B2 (en) 2014-04-07 2017-09-26 Sanofi Peptidic dual GLP-1/glucagon receptor agonists derived from exendin-4
US9775904B2 (en) 2014-04-07 2017-10-03 Sanofi Exendin-4 derivatives as peptidic dual GLP-1/glucagon receptor agonists
US9937240B2 (en) 2014-04-11 2018-04-10 Gemvax & Kael Co., Ltd. Peptide having fibrosis inhibitory activity and composition containing same
US10662223B2 (en) 2014-04-30 2020-05-26 Gemvax & Kael Co., Ltd. Composition for organ, tissue, or cell transplantation, kit, and transplantation method
US9932381B2 (en) 2014-06-18 2018-04-03 Sanofi Exendin-4 derivatives as selective glucagon receptor agonists
EP3160491A4 (en) * 2014-06-25 2018-01-17 GlaxoSmithKline LLC Pharmaceutical compositions
US10583080B2 (en) 2014-09-30 2020-03-10 Intarcia Therapeutics, Inc. Therapeutic methods for the treatment of diabetes and related conditions for patients with high baseline HbA1c
US9889085B1 (en) 2014-09-30 2018-02-13 Intarcia Therapeutics, Inc. Therapeutic methods for the treatment of diabetes and related conditions for patients with high baseline HbA1c
WO2016061583A1 (en) * 2014-10-17 2016-04-21 University Of Virginia Patent Foundation Compositions and methods for treating pituitary tumors
US11077163B2 (en) 2014-12-23 2021-08-03 Gemvax & Kael Co., Ltd. Peptide for treating ocular diseases and composition for treating ocular diseases comprising same
US10463708B2 (en) 2014-12-23 2019-11-05 Gemvax & Kael Co., Ltd. Peptide for treating ocular diseases and composition for treating ocular diseases comprising same
US10835582B2 (en) 2015-02-27 2020-11-17 Gemvax & Kael Co. Ltd. Peptide for preventing hearing loss, and composition comprising same
US10925639B2 (en) 2015-06-03 2021-02-23 Intarcia Therapeutics, Inc. Implant placement and removal systems
US10806797B2 (en) 2015-06-05 2020-10-20 Sanofi Prodrugs comprising an GLP-1/glucagon dual agonist linker hyaluronic acid conjugate
US11015179B2 (en) 2015-07-02 2021-05-25 Gemvax & Kael Co., Ltd. Peptide having anti-viral effect and composition containing same
US9982029B2 (en) 2015-07-10 2018-05-29 Sanofi Exendin-4 derivatives as selective peptidic dual GLP-1/glucagon receptor agonists
US10898540B2 (en) 2016-04-07 2021-01-26 Gem Vax & KAEL Co., Ltd. Peptide having effects of increasing telomerase activity and extending telomere, and composition containing same
US11214607B2 (en) 2016-05-16 2022-01-04 Intarcia Therapeutics Inc. Glucagon-receptor selective polypeptides and methods of use thereof
US10501517B2 (en) 2016-05-16 2019-12-10 Intarcia Therapeutics, Inc. Glucagon-receptor selective polypeptides and methods of use thereof
US11840559B2 (en) 2016-05-16 2023-12-12 I2O Therapeutics, Inc. Glucagon-receptor selective polypeptides and methods of use thereof
USD835783S1 (en) 2016-06-02 2018-12-11 Intarcia Therapeutics, Inc. Implant placement guide
USD912249S1 (en) 2016-06-02 2021-03-02 Intarcia Therapeutics, Inc. Implant removal tool
USD962433S1 (en) 2016-06-02 2022-08-30 Intarcia Therapeutics, Inc. Implant placement guide
USD860451S1 (en) 2016-06-02 2019-09-17 Intarcia Therapeutics, Inc. Implant removal tool
USD840030S1 (en) 2016-06-02 2019-02-05 Intarcia Therapeutics, Inc. Implant placement guide
CN108030997B (en) * 2016-08-03 2021-01-26 尼尔·希夫拉杰·戴维 Adjustable rate drug delivery implantable devices
CN108030997A (en) * 2016-08-03 2018-05-15 尼尔·希夫拉杰·戴维 Adj sp medicine delivery implantable device
CN108066762A (en) * 2016-11-07 2018-05-25 赫兰 The purposes of GLP-1 receptor stimulating agents and biguanides
CN108066762B (en) * 2016-11-07 2022-04-01 赫兰 Application of GLP-1 receptor agonist drugs
US10835580B2 (en) 2017-01-03 2020-11-17 Intarcia Therapeutics, Inc. Methods comprising continuous administration of a GLP-1 receptor agonist and co-administration of a drug
US11654183B2 (en) 2017-01-03 2023-05-23 Intarcia Therapeutics, Inc. Methods comprising continuous administration of exenatide and co-administration of a drug
CN110545838A (en) * 2017-03-08 2019-12-06 因塔西亚制药公司 apparatus and method for administering nausea-causing compounds from a drug delivery device
WO2018165462A1 (en) * 2017-03-08 2018-09-13 Intarcia Therapeutics, Inc Apparatus and methods for administration of a nauseogenic compound from a drug delivery device
US11752198B2 (en) 2017-08-24 2023-09-12 Novo Nordisk A/S GLP-1 compositions and uses thereof
US10888605B2 (en) 2017-08-24 2021-01-12 Novo Nordisk A/S GLP-1 compositions and uses thereof
USD933219S1 (en) 2018-07-13 2021-10-12 Intarcia Therapeutics, Inc. Implant removal tool and assembly
US11439802B2 (en) * 2018-11-19 2022-09-13 Biora Therapeutics, Inc. Ingestible device for delivery of therapeutic agent to the gastrointestinal tract
US11318191B2 (en) 2020-02-18 2022-05-03 Novo Nordisk A/S GLP-1 compositions and uses thereof
US11338119B2 (en) * 2020-03-20 2022-05-24 The Regents Of The University Of California Implantable drug delivery devices for localized drug delivery
US11344526B2 (en) 2020-03-20 2022-05-31 The Regents Of The University Of California Implantable drug delivery devices for localized drug delivery
US11173291B2 (en) 2020-03-20 2021-11-16 The Regents Of The University Of California Implantable drug delivery devices for localized drug delivery

Also Published As

Publication number Publication date
EP2675469A4 (en) 2015-04-08
WO2012112626A2 (en) 2012-08-23
US20170252409A1 (en) 2017-09-07
US10159714B2 (en) 2018-12-25
CA2823721C (en) 2020-07-07
CA2823721A1 (en) 2012-08-23
US20150057227A1 (en) 2015-02-26
EP2675469A2 (en) 2013-12-25
WO2012112626A3 (en) 2014-04-17

Similar Documents

Publication Publication Date Title
US10159714B2 (en) Compositions, devices and methods of use thereof for the treatment of cancers
JP6835933B2 (en) Rapid establishment and / or termination of substantial steady-state drug delivery
US20120289944A1 (en) Highly concentrated drug particles, formulations, suspensions and uses thereof
AU2014280920B2 (en) Rapid establishment and/or termination of substantial steady-state drug delivery

Legal Events

Date Code Title Description
AS Assignment

Owner name: INTARCIA THERAPEUTICS, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:LEUNG, KARLING ALICE;REEL/FRAME:028009/0244

Effective date: 20120308

AS Assignment

Owner name: U.S. BANK NATIONAL ASSOCIATION, AS COLLATERAL AGEN

Free format text: SECURITY AGREEMENT;ASSIGNOR:INTARCIA THERAPEUTICS, INC.;REEL/FRAME:029299/0678

Effective date: 20121114

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: INTARCIA THERAPEUTICS, INC., CALIFORNIA

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:U.S. BANK NATIONAL ASSOCIATION, AS COLLATERAL AGENT;REEL/FRAME:036942/0544

Effective date: 20150813