WO2001087335A2 - Method for selectively inhibiting ghrelin action - Google Patents

Method for selectively inhibiting ghrelin action Download PDF

Info

Publication number
WO2001087335A2
WO2001087335A2 PCT/US2001/011752 US0111752W WO0187335A2 WO 2001087335 A2 WO2001087335 A2 WO 2001087335A2 US 0111752 W US0111752 W US 0111752W WO 0187335 A2 WO0187335 A2 WO 0187335A2
Authority
WO
WIPO (PCT)
Prior art keywords
ghrelin
ghs
compound
gna
mixture
Prior art date
Application number
PCT/US2001/011752
Other languages
French (fr)
Other versions
WO2001087335A3 (en
Inventor
Henry Uhlman Bryant
Mark Louis Heiman
Original Assignee
Eli Lilly And Company
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 Eli Lilly And Company filed Critical Eli Lilly And Company
Priority to AU2001259056A priority Critical patent/AU2001259056A1/en
Priority to US10/258,259 priority patent/US20030211967A1/en
Priority to EP01932539A priority patent/EP1286697A2/en
Publication of WO2001087335A2 publication Critical patent/WO2001087335A2/en
Publication of WO2001087335A3 publication Critical patent/WO2001087335A3/en
Priority to US11/106,899 priority patent/US20050201938A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5058Neurological cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • 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
    • A61K38/25Growth hormone-releasing factor [GH-RF] (Somatoliberin)
    • 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
    • A61K38/27Growth hormone [GH] (Somatotropin)
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/0004Screening or testing of compounds for diagnosis of disorders, assessment of conditions, e.g. renal clearance, gastric emptying, testing for diabetes, allergy, rheuma, pancreas functions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/04X-ray contrast preparations
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/502Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/035Animal model for multifactorial diseases
    • A01K2267/0362Animal model for lipid/glucose metabolism, e.g. obesity, type-2 diabetes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies

Definitions

  • the present invention is in the field of human medicine, particularly in the treatment of obesity and disorders associated with obesity such as diabetes mellitus. More specifically the invention relates to a method for treating obesity by administering a compound which blocks ghrelin action.
  • the present invention provides a method of selectively inhibiting ghrelin activity in a mammal comprising administering to a mammal in need thereof a therapeutically-effective amount of a compound selected from the group consisting of a growth hormone secretagogue receptor antagonist (GHS-RA) and a ghrelin neutralizing agent (GNA) .
  • the invention further provides a method for treating obesity and related disorders in a mammal comprising administering to a mammal in need thereof a therapeutically-effective amount of a compound selected from the group consisting of a growth hormone secretagogue receptor antagonist (GHS-RA) and a ghrelin neutralizing agent (GNA) .
  • Other embodiments include in vi tro and in vivo screening and assay methods .
  • GHRH growth hormone-releasing peptides
  • GHS-R GHS receptor
  • Obesity also called diverence or fatness
  • body fat usually caused by the consumption of more calories than the body uses. The excess calories are then stored as fat, or adipose tissue.
  • Overweight if moderate, is not necessarily obesity, particularly in muscular or large-boned individuals . In general, however, a body weight 20 percent or more over the optimum tends to be associated with obesity.
  • treating or treatment describes the management and care of a patient for the purpose of combating the disease, condition, or disorder.
  • Treating includes the administration of a compound of present invention to prevent the onset of the symptoms or complications, alleviating the symptoms or complications, or eliminating the disease, condition, or disorder.
  • Treating obesity therefore includes the inhibition of food intake, the inhibition of weight gain, and inducing weight loss in patients in need thereof.
  • the term 'related disorders' includes but is not limited to type II diabetes, cardiovascular disease, cancer, and other disease states whose etiology stems from obesity.
  • the term 'administering' or 'administration' as used herein includes any means for introducing a GHS-RA or GNA into the body such that the substance is able to interact with the GHS-R or secreted ghrelin.
  • Preferred routes of administration will introduce the substance into the systemic circulation. Examples include but are not limited to oral; transdermal; subcutaneous, intravenous, and intramuscular injection.
  • the active agents of the present invention are administered to a mammal, preferably a human, in accord with known methods, such as intravenous administration as a bolus or by continuous infusion over a period of time, by intramuscular, intraperitoneal, intracerebral, intracerobrospinal, subcutaneous, intra-articular, intrasynovial, intrathecal, intraocular, intralesional, oral, topical, inhalation or through sustained release.
  • a therapeutically-effective amount is at least the minimal dose, but less than a toxic dose, of an active agent which is necessary to impart therapeutic benefit to a mammal. Stated another way, a therapeutically-effective amount is an amount which induces, ameliorates or otherwise causes an improvement in the obese state of the mammal.
  • Carriers' as used herein include pharmaceutically- acceptable carriers, excipients, or stabilizers which are nontoxic to the cell or mammal being exposed thereto at the dosages and concentrations employed. Often the physiologically-acceptable carrier is an aqueous pH buffered solution.
  • physiologically acceptable carriers include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid; low molecule weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt- forming counterions such as sodium; and/or nonionic surfactants such as TWEEN®, polyethylene glycol (PEG) , and PLURONICS®.
  • buffers such as phosphate, citrate, and other organic acids
  • antioxidants including ascorbic acid
  • low molecule weight (less than about 10 residues) polypeptides proteins, such as serum
  • 'mammal' refers to any animal classified as a mammal, including humans, domestic, farm and zoo animals, and sports or companion animals, etc. In a preferred embodiment of the invention, the mammal is a human.
  • 'antibody' is used in the broadest sense and specifically includes monoclonal antibodies, chimeric antibodies, humanized antibodies, and fully human antibodies .
  • 'monoclonal antibody' refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally-occurring mutations that may be present in minor amounts .
  • Antibody fragments means a portion of an intact antibody, preferably the antigen binding or variable region of the intact antibody.
  • antibody fragments include Fab, Fab', F(ab')l and Fv fragments; diabodies; linear antibodies (Zapata et al . , Protein Engin . S(10): 1057-1 062 (1991)); single-chain antibody molecules; and multispecific antibodies formed from antibody fragments.
  • the term 'Fv' is the minimum antibody fragment, which contains a complete antigen-recognition and binding site. This region consists of a dimer of one heavy- and one light chain variable domain in tight, non-covalent association.
  • variable domain the three complementarity-determining regions (CDRs) of each variable domain interact to define an antigen-binding site on the surface of the VH-VL dimer.
  • CDRs complementarity-determining regions
  • the Fab fragment also contains the constant domain of the light chain and the 'first constant domain (CHI) of the heavy chain.
  • Fab fragments differ from Fv fragments by the addition of a few residues at the carboxy terminus of the heavy chain CHI domain including one or more cysteines from the antibody hinge region.
  • Fab'-SH is the designation herein for Fab' in which the cysteine residue (s) of the constant domains bear a free thiol group.
  • F(ab') z antibody fragments originally were produced as pairs of Fab' fragments which have hinge cysteines between them. Other chemical couplings of antibody fragments are also known.
  • Papain digestion of antibodies produces two identical antigen-binding fragments, called Fab fragments, each with a single antigen-binding site, and a residual Fc fragment, a designation reflecting the ability to crystallize readily.
  • Pepsin treatment yields an F(ab') 2 fragment that has two antigen-combining sites and is still capable of cross- linking antigen.
  • immunoglobulins The 'light chains' of antibodies (immunoglobulins) from any vertebrate species can be assigned to one of two clearly distinct types, called kappa and lambda, based on the amino acid sequences of their constant domains .
  • immunoglobulins can be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes) , e.g., IgGl, IgG2, IgG3, IgG4, IgA and IgA2.
  • Fv'Single-chain Fv' antibody fragments comprise the VH and VL domains of antibody, wherein these domains are present in a single polypeptide chain.
  • the Fv polypeptide further comprises a polypeptide linker between the VH and VL domain, which enables the sFv to form the desired structure for antigen binding.
  • the term ' immunoadhesion' designates antibodylike molecules that combine the binding specificity of a heterologous protein (an 'adhesion') with the effector functions of immunoglobulin constant domains.
  • the immunoadhesions comprise a fusion of an amino acid sequence with the desired binding specificity which is other than the antigen recognition and binding site of an antibody (i.e., is heterologous), and an immunoglobulin constant domain sequence.
  • the adhesion part of an immunoadhesion molecule typically is a contiguous amino acid sequence comprising at least the binding site of a receptor or a ligand.
  • the immunoglobulin constant domain sequence in the immunoadhesion may be obtained from any immunoglobulin, such as IgG-1, IgG-2, IgG-3 or IgG-4 subtypes, IgA (including IgG-1 and IgA-2) , IgE, IgD or IgM.
  • 'diabodies' refers to small antibody fragments with two antigen-binding sites, which fragments comprise a heavy-chain variable domain (VH) connected to a light chain variable domain (VL) in the same polypeptide chain (VH-VL) .
  • VH heavy-chain variable domain
  • VL light chain variable domain
  • Diabodies are described more fully in, for example, EP 404.097, WO 93/1 1161; and Hollinger et al . , Proc. Natl. Acad. Sci . USA 90: 6444-6448 (1993) .
  • a GHS-RA is any compound that partially or fully antagonizes, blocks, or otherwise inhibits the biological action of ghrelin by binding to the GHS-R without stimulating the release of growth hormone. Therefore GHS (compounds that bind the GHS-R and stimulate the release of GH) are not consistent with the claimed method.
  • GHS-RA are compounds useful in the presently claimed method and include but are not limited to natural products, synthetic organic compounds, peptides, proteins, antibodies, antibody fragments, single chain antibodies, and antibody based constructs. The current level of skill in the art of receptor binding and growth hormone assays places GHS-RAs well within the grasp of the ordinarily skilled artisan. There are several routine approaches for identifying a GHS-R.
  • GHS-RA test compound is first checked to determine if it binds GHS-R. This is accomplished using routine radiometric binding methods .
  • a second messenger reporter such as calcium can be used to determine binding.
  • Assay is described in Kojima et al . , Nature 402: 656-60 (1999). Compounds that bind GHS-R are then exposed to primary pituitary cells, for example, and release of growth hormone is determined using standard commercially available assays.
  • Compounds that bind but do not stimulate the release of GH should then be assayed for ghrelin antagonism by exposing pituitary cells to the GHS-RA in the presence of ghrelin and then assaying for GH release.
  • Antibody-based GHS-RAs are also consistent with the claimed method.
  • Anti-GHS-R antibodies may be generated by a variety of well-known methods that include traditional antisera production and monoclonal antibody techniques.
  • Ghrelin neutralizing agents represent another aspect of the invention.
  • ghrelin is neutralized or otherwise rendered biologically inactive apart from the receptor.
  • Agents suitable for this application are those which specifically bind ghrelin, preferably with a higher affinity constant than the GHS-R.
  • Antibody or antibody-based agents are preferred because they can be purposefully generated using well established techniques.
  • Kojima et al . Nature 402: 656-60 (1999) .
  • Immunoadhesions Fc fusion constructs, similar to Enbrel®, where the soluble ligand-binding domain of the GHS- R is fused to a human Fc
  • Dosages and desired drug concentration of pharmaceutical compositions of the present invention may vary depending on the particular use envisioned. The determination of the appropriate dosage or route of administration is well within the skill of an ordinary artisan. Animal experiments provide reliable guidance for the determination of effective doses for human therapy.
  • an article of manufacture containing materials useful in the presently claimed methods comprises a container and a label.
  • Suitable containers include, for example, bottles, vials, syringes, and test tubes .
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition which is effective for specifically inhibiting ghrelin action and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle) .
  • the active agent in the composition is a GHS-RA and/or a GNA.
  • the label on, or associated with, the container indicates that the composition is used for treating obesity and/or related disorders.
  • the article of manufacture may further comprise a second container comprising a pharmaceutically-acceptable buffer, such as phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial end user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
  • Five-minute fractions were collected while monitoring the U.V. at 214 nm (2.0A).
  • the appropriate fractions were combined, frozen and lyophilized.
  • MALDI- ass spectral analysis indicated a mass of 3313.85 g for the purified ghrelin, which was consistent with the theoretical molecular weight.
  • Example 2 Animals Wild-type mice (129SV strain) and NPY-knockout mice were obtained from Taconic Farms ® . Eight-week old dwarf rats were purchased at Harlan UK. Animals were housed individually in a temperature controlled environment (25 C°) with a 12-hour light and 12-hour dark (18.00 - 06.00) photoperiod. All mice had ad libi tum access to pelleted mouse food (5008 PMI ® Nutrition International) and tap water . Mice were between 9 and 13 weeks of age and were injected daily between 17.00 and 18.00 with 0.1 ml of phosphate buffered saline (PBS) containing 0 or 8 mg/kg/d ghrelin over 13 days . Food intake and body weights were measured daily at O ⁇ .OOh. All animal experiments were conducted in accordance with the principles and procedures outlined in the National Institute of Health (NIH) Guide for the Care and Use of Laboratory Animals.
  • NASH National Institute of Health
  • RQ is the ratio of VC0 2 to V0 2 .
  • CV calorific value of oxygen
  • Total calories expended were calculated to determine daily fuel utilization.
  • proportion of protein, fat and carbohydrate that is utilized during that 24-hour period we used Flatt's proposal and assumed that protein utilization was equivalent to protein intake for adult stable animals (Flatt, J.P., J. Nutr Biochem 2, 193-202 (1991)).
  • DXA dual-energy X-ray absorptiometry
  • Body composition was measured on day 14 of the treatment period by DXA using a Norland p-DEXA ® (Norland,
  • the system provides a non-invasive method for quantification of whole body composition and is based on the differential attenuation of high and low energy x-rays by the tissues in the scan area.
  • Soft tissues attenuate the energy beam less than bone; of the soft tissue mass, fat tissue attenuates the beam less than lean tissue.
  • Fat mass consists primarily of adipose tissue, but lean mass includes organs, tendons, cartilage, blood and body water in addition to skeletal muscle. In the present study, fat mass, lean mass and bone mineral content (bone mass) were measured and reported. Mice were anesthetized with inhalation of isoflorane and placed on the instrument platform in ventral position. Measurements were performed at a speed of 10 mm/ in and a resolution of 0.5 x 0.5 mm. Quality controls using phantom ID2232 and Calibration Standard 82315 (Norland) were performed before starting measurements .
  • Example 5 Example 5
  • mice were treated with GHRP-2 for 18 days.
  • Hypothalamic RNA levels (measured by RT-PCR) of neuropeptide Y (NPY) , agouti-related-protein (AGRP) , pro- opio-melanocortin (POMC) and melanocyte-concentrating hormone (MCH) were not changed.
  • GHRP-6 increases c- fos expression in NPY-neurons (Vernon, R. G.; J Endocrinol 150, 129-40 (1996)) and because these neurons also release AGRP, a natural melanocyte stimulating hormone antagonist
  • Recombinant CHO cells expressing the human growth hormone secretagogue receptor cDNA described by Howard et al . , Science 273: 974-977 (1996) are grown and harvested in nutrient medium.
  • Membrane preparations are then obtained by first washing the cells with PBS buffer, then twice washing with cold buffer (25 mM HEPES, 2 M MgCl 2 , 1 mM EDTA, 20 ⁇ g/ml Leupeptin, 1 mM PMSF, 2 ⁇ g/ml Aprotinin, 50 ⁇ g/ml Trypsin Inhibitor, pH 8.0) and resuspending in buffer.
  • the cell suspension is lysed in a glass Teflon® homogenizer, and the resulting sample is then centrifuged at 35,300 X g for 30 minutes at 4°C.
  • the supernatant is removed, and the pellet is resuspended in cold buffer and homogenized. Aliquots may then be prepared and stored at -80°C.
  • a sample of the membrane preparation is pre- incubated with a test compound or a control compound with and without added grhelin in buffer (25 mM HEPES, 0.2%
  • BSA 2.6 mM Mg, 0.8 mM ATP, 0.1 mM GTP, 5 mM creatine phosphate, creatine kinase 50 U/ml, 0.2 mM IBMX, pH 7.6) is added and incubated for an additional 30 minutes. Incubations are stopped by adding 10 mM EDTA.
  • cAMP-b phycoerythrin conjugate is added followed by the addition of affinity purified anti-cAMP rabbit antiserum.
  • anti-rabbit IgG coated assay beads are added and incubated for an additional 15 minutes. Plates are then evacuated and read on a Pandex® PFCIA reader.
  • ghrelin binding shows decreasing fluorescent intensity due to increased cAMP concentration. Fluorescent intensity values are correlated to rate of cAMP production (pmol/min/mg) . Conversely, inhibition of ghrelin binding by either receptor blockade or ghrelin neutralizations shows no decrease in fluorescent intensity.
  • ghrelin is mainly generated by the stomach and secreted into circulation
  • total energy 1795 ⁇ 105 kcal
  • plasma ghrelin decreased by 30% 2 hours after the meal (p ⁇ 0.01), and over the 24h sampling period (p ⁇ 0.05).
  • plasma ghrelin levels did not decrease after meals.
  • ghrelin release is a normal response to fasting.
  • Such elevated ghrelin stimulates appetite and the utilization of carbohydrate (determined in above examples using rodents) and thus corrects hypoglycemia resulting from fasting.
  • Ingestion of glucose rescues hypogylcemia and thus inhibits ghrelin secretion from the stomach to prevent hyperglycemia .
  • ghrelin plays an important role in the regulation of blood glucose. Agents that block ghrelin action may be useful for the treatment of diabetes .

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Urology & Nephrology (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Endocrinology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Pathology (AREA)
  • Toxicology (AREA)
  • Biochemistry (AREA)
  • Cell Biology (AREA)
  • Analytical Chemistry (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Biotechnology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • General Physics & Mathematics (AREA)
  • Organic Chemistry (AREA)
  • Diabetes (AREA)
  • Zoology (AREA)
  • Biophysics (AREA)
  • Child & Adolescent Psychology (AREA)
  • Obesity (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)

Abstract

The present invention provides a method selectively inhibiting ghrelin activity to treat a variety of diseases including obesity and related disorders, particularly in individuals who are genetically predisposed. One aspect of the invention comprises administering an agent that effectively neutralizes ghrelin. Another aspect comprises administering a ghrelin receptor (growth hormone secretagogue receptor) antagonist.

Description

METHOD FOR SELECTIVELY INHIBITING GHRELIN ACTION
The present invention is in the field of human medicine, particularly in the treatment of obesity and disorders associated with obesity such as diabetes mellitus. More specifically the invention relates to a method for treating obesity by administering a compound which blocks ghrelin action.
Obesity, and especially upper body obesity, is a common and very serious public health problem in the United States and throughout the world. According to recent statistics, more than 25% of the United States population and 27% of the Canadian population are overweight. uczmarski, Amer. J. of Clin . Nutr. 55: 495S - 502S, 1992;
Reeder et. al., Can . Med. Ass . J. , 23: 226-233, 1992. Upper body obesity is the strongest risk factor known for type II diabetes mellitus, and is a strong risk factor for cardiovascular disease and cancer as well. Recent estimates for the medical cost of obesity are $150,000,000,000 worldwide. The problem has become serious enough that the surgeon general has begun an initiative to combat the ever- increasing adiposity rampant in American society.
Much of this obesity-induced pathology can be attributed to the strong association with dysli'pidemia, hypertension, and insulin resistance. Many studies have demonstrated that reduction in obesity by diet and exercise reduces these risk factors dramatically. Unfortunately, these treatments are largely unsuccessful with a failure rate reaching 95%. This failure may be due to the fact that the condition is strongly associated with genetically inherited factors that contribute to increased appetite, preference for highly caloric foods, reduced physical activity, and increased lipogenic metabolism. This indicates that people inheriting these genetic traits are prone to becoming obese regardless of their efforts to combat the condition. Therefore, a means for effectively treating obese individuals, especially those who are genetically predisposed is needed.
The present invention provides a method of selectively inhibiting ghrelin activity in a mammal comprising administering to a mammal in need thereof a therapeutically-effective amount of a compound selected from the group consisting of a growth hormone secretagogue receptor antagonist (GHS-RA) and a ghrelin neutralizing agent (GNA) . The invention further provides a method for treating obesity and related disorders in a mammal comprising administering to a mammal in need thereof a therapeutically-effective amount of a compound selected from the group consisting of a growth hormone secretagogue receptor antagonist (GHS-RA) and a ghrelin neutralizing agent (GNA) . Other embodiments include in vi tro and in vivo screening and assay methods .
Physiologists have postulated for years that, when a mammal overeats, the resulting excess fat signals to the brain that the body is obese which, in turn, causes the body to eat less and bum more fuel. G. R. Hervey, Nature 227: 629-631 (1969) . This feedback model is supported by parabiotic experiments, which implicate circulating hormones that influence and regulate aspects of adiposity.
Growth hormone-releasing peptides (GHRPs) were first described in 1981 by Bowers and colleagues before the discovery of growth hormone- eleasing hormone (GHRH) . Momany FA, Bowers CY, Reynolds GA, Chang D, Hong A, and Newlander K . , Endocrinology 108: 31-39, 1981. Bowers CY, Momany FA, Reynolds GA, Hong A., Endocrinology 114: 1537- 1545 (1984) . While Bowers' group demonstrated that such peptides could stimulate growth hormone (GH) release from isolated pituitary glands, they almost always reported a greater GH response when the GHRPs were administered in vivo. These data, reported in the early 1980 's, suggested that such GHRPs have actions at both the hypothalamus and pituitary. After almost a decade, a non-peptidyl GH secretagogue (GHS) was reported and there have been many additional improvements in potency, bioavailability and Pharmacokinetics of GHS. Smith RG, Cheng K, Schoen WR, Pong S-S, Hickey GJ, Jacks TM, Butler BS, Chan W -S, Chaung -YP, Judith F, Taylor AM, Wyvratt Jr MJ, and Fisher MH., Science 260: 1640-1643 (1993) . A review of this general area was published recently. Smith RG, Van der Ploeg LHT, Howard AD, Feighner SD, Cheng K, Hickey GJ, Wyvratt Jr MJ, Fisher MH, Nargund RP, and Patchett AA. , Endocrine Rev. 18:621-645 (1997) .
After Smith and colleagues identified GHS, they isolated a GHS receptor (GHS-R) cDNA from both the pituitary and hypothalamus . Howard AD, Feighner SD, Cully DF, Arena JP, Liberator PA, Rosenblum CI, Hamelin M, Hreniuk DL, Palyha OC, Anderson J, Paress PS, Diaz C, Chou M, Liu KK, McKee KK, Pong SS, Chaung LY, Elbrecht A, Dashkevicz M, Heavens R, Rigby M, Sirinathsingh i DJS, Dean DC, Melillo DG, Van der Ploeg LHT, Science 273: 974-977 (1996).
In December 1999, the endogenous ligand for GHS-R was identified and named ghrelin. Koji a M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K. , Nature 402: 656-60 (1999) . They demonstrated that it is secreted by stomach tissue; and its mKNA is also expressed in the hypothalamus. Thus, the GHS-R now may be thought of as the ghrelin receptor. A review of this general area was recently published. Bowers CY., J" Clin . Endocrinol . Metab.86 : 1464- 1469 (2001) . Although most GHS and GHRP studies were designed to exploit stimulation of the somatotropic axis, it has been demonstrated that these synthetic molecules induce sleep. Copinschi G, Leproult R, Vanonderbergen A, Caufriez A, Cole KY, Schilling LM. , Neuroendocrinol . 66: 278-286 (1997). Others have demonstrated that the synthetic GHS and GHRP also induce food intake. Locke W, Kirgis HD, Bowers CY, and Abdo AA. , Life Sci . 56:1347-1352 (1995). Okada K, Ishii S, Minami S, Sugihara H, Shibasaki T, and Wakabayashi I., Endocrinology 137:5155-5158 (1996). Moreover, Bennett et al . demonstrated that GHS-R is highly expressed in the arcuate nucleus. Bennett PA, Thomas GB, Howard AD, Feighner SD, Van der Ploeg LHT, Smith RG, and Robinson ICAF., Endocrinology 138: 4552-4557 (1997). In 1993, Dickson and colleagues observed an activation of such hypothalamic neurons after peripheral administration of a GHRP. Dickson SL, Leng G, and Robinson ICAF., Neuroscience 53: 303-306 (1993) . Additionally, this group demonstrated that a majority of these activated neurons were those expressing neuropeptide-Y mRNA. Dickson SL and Luckman SM., Endocrinology 138: 771-777 (1997).
In view of this state of the art, the inventors of the presently claimed invention were most surprised when they demonstrated in an animal model that administration of ghrelin predominantly lead to fat deposition. Tschoep M. , Smiley DL., and Heiman ML., Nature 407: 908-913 (2000). This lead them to postulate that ghrelin signals the CNS when energy homeostasis requires increased metabolic efficiency to induce energy preservation and a partitioning of fuel utilization from fat to carbohydrate to prevent hypoglycemia. Consequently, blocking or antagonizing ghrelin action compromises metabolic efficiency and induces energy consumption, primarily from fat stores.
Obesity, also called corpulence or fatness, is the excessive accumulation of body fat, usually caused by the consumption of more calories than the body uses. The excess calories are then stored as fat, or adipose tissue. Overweight, if moderate, is not necessarily obesity, particularly in muscular or large-boned individuals . In general, however, a body weight 20 percent or more over the optimum tends to be associated with obesity.
For purposes of the present invention, treating or treatment describes the management and care of a patient for the purpose of combating the disease, condition, or disorder. Treating includes the administration of a compound of present invention to prevent the onset of the symptoms or complications, alleviating the symptoms or complications, or eliminating the disease, condition, or disorder. Treating obesity therefore includes the inhibition of food intake, the inhibition of weight gain, and inducing weight loss in patients in need thereof. For purposes of this invention, the term 'related disorders' includes but is not limited to type II diabetes, cardiovascular disease, cancer, and other disease states whose etiology stems from obesity. The term 'administering' or 'administration' as used herein includes any means for introducing a GHS-RA or GNA into the body such that the substance is able to interact with the GHS-R or secreted ghrelin. Preferred routes of administration will introduce the substance into the systemic circulation. Examples include but are not limited to oral; transdermal; subcutaneous, intravenous, and intramuscular injection.
The active agents of the present invention are administered to a mammal, preferably a human, in accord with known methods, such as intravenous administration as a bolus or by continuous infusion over a period of time, by intramuscular, intraperitoneal, intracerebral, intracerobrospinal, subcutaneous, intra-articular, intrasynovial, intrathecal, intraocular, intralesional, oral, topical, inhalation or through sustained release.
A therapeutically-effective amount is at least the minimal dose, but less than a toxic dose, of an active agent which is necessary to impart therapeutic benefit to a mammal. Stated another way, a therapeutically-effective amount is an amount which induces, ameliorates or otherwise causes an improvement in the obese state of the mammal. 'Carriers' as used herein include pharmaceutically- acceptable carriers, excipients, or stabilizers which are nontoxic to the cell or mammal being exposed thereto at the dosages and concentrations employed. Often the physiologically-acceptable carrier is an aqueous pH buffered solution. Examples of physiologically acceptable carriers include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid; low molecule weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt- forming counterions such as sodium; and/or nonionic surfactants such as TWEEN®, polyethylene glycol (PEG) , and PLURONICS®.
The term 'mammal' as used herein refers to any animal classified as a mammal, including humans, domestic, farm and zoo animals, and sports or companion animals, etc. In a preferred embodiment of the invention, the mammal is a human.
The term 'antibody' is used in the broadest sense and specifically includes monoclonal antibodies, chimeric antibodies, humanized antibodies, and fully human antibodies .
The term 'monoclonal antibody' as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally-occurring mutations that may be present in minor amounts .
Antibody, fragments means a portion of an intact antibody, preferably the antigen binding or variable region of the intact antibody. Examples of antibody fragments include Fab, Fab', F(ab')l and Fv fragments; diabodies; linear antibodies (Zapata et al . , Protein Engin . S(10): 1057-1 062 (1991)); single-chain antibody molecules; and multispecific antibodies formed from antibody fragments. The term 'Fv' is the minimum antibody fragment, which contains a complete antigen-recognition and binding site. This region consists of a dimer of one heavy- and one light chain variable domain in tight, non-covalent association. It is in this configuration that the three complementarity-determining regions (CDRs) of each variable domain interact to define an antigen-binding site on the surface of the VH-VL dimer. Collectively, the six CDRs confer antigen-binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDR specific for an antigen) has the ability to recognize and bind antigen, although at a lower avidity than a complete antibody .
The Fab fragment also contains the constant domain of the light chain and the 'first constant domain (CHI) of the heavy chain. Fab fragments differ from Fv fragments by the addition of a few residues at the carboxy terminus of the heavy chain CHI domain including one or more cysteines from the antibody hinge region. Fab'-SH is the designation herein for Fab' in which the cysteine residue (s) of the constant domains bear a free thiol group. F(ab')z antibody fragments originally were produced as pairs of Fab' fragments which have hinge cysteines between them. Other chemical couplings of antibody fragments are also known.
Papain digestion of antibodies produces two identical antigen-binding fragments, called Fab fragments, each with a single antigen-binding site, and a residual Fc fragment, a designation reflecting the ability to crystallize readily. Pepsin treatment yields an F(ab')2 fragment that has two antigen-combining sites and is still capable of cross- linking antigen.
The 'light chains' of antibodies (immunoglobulins) from any vertebrate species can be assigned to one of two clearly distinct types, called kappa and lambda, based on the amino acid sequences of their constant domains . Depending on the amino acid sequence of the constant domain of their heavy chains, immunoglobulins can be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes) , e.g., IgGl, IgG2, IgG3, IgG4, IgA and IgA2. 'Single-chain Fv' antibody fragments comprise the VH and VL domains of antibody, wherein these domains are present in a single polypeptide chain. Preferably, the Fv polypeptide further comprises a polypeptide linker between the VH and VL domain, which enables the sFv to form the desired structure for antigen binding. For a review of sFv, see Pluckthun in
The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds., Springer-Verlag, New York, pp. 269-3 15, 1994. As used herein, the term ' immunoadhesion' designates antibodylike molecules that combine the binding specificity of a heterologous protein (an 'adhesion') with the effector functions of immunoglobulin constant domains. Structurally, the immunoadhesions comprise a fusion of an amino acid sequence with the desired binding specificity which is other than the antigen recognition and binding site of an antibody (i.e., is heterologous), and an immunoglobulin constant domain sequence. The adhesion part of an immunoadhesion molecule typically is a contiguous amino acid sequence comprising at least the binding site of a receptor or a ligand. The immunoglobulin constant domain sequence in the immunoadhesion may be obtained from any immunoglobulin, such as IgG-1, IgG-2, IgG-3 or IgG-4 subtypes, IgA (including IgG-1 and IgA-2) , IgE, IgD or IgM.
The term 'diabodies' refers to small antibody fragments with two antigen-binding sites, which fragments comprise a heavy-chain variable domain (VH) connected to a light chain variable domain (VL) in the same polypeptide chain (VH-VL) . By using a linker that is too short to allow pairing between the two domains on the same chain, the domains are forced to pair with the complementary domains of another chain and create two antigen binding sites . Diabodies are described more fully in, for example, EP 404.097, WO 93/1 1161; and Hollinger et al . , Proc. Natl. Acad. Sci . USA 90: 6444-6448 (1993) .
A GHS-RA is any compound that partially or fully antagonizes, blocks, or otherwise inhibits the biological action of ghrelin by binding to the GHS-R without stimulating the release of growth hormone. Therefore GHS (compounds that bind the GHS-R and stimulate the release of GH) are not consistent with the claimed method. GHS-RA are compounds useful in the presently claimed method and include but are not limited to natural products, synthetic organic compounds, peptides, proteins, antibodies, antibody fragments, single chain antibodies, and antibody based constructs. The current level of skill in the art of receptor binding and growth hormone assays places GHS-RAs well within the grasp of the ordinarily skilled artisan. There are several routine approaches for identifying a GHS-R. One basic scheme involves a receptor binding assay followed by a GH release assay. In this scheme, the GHS-RA test compound is first checked to determine if it binds GHS-R. This is accomplished using routine radiometric binding methods . Alternatively, a second messenger reporter such as calcium can be used to determine binding. One such assay is described in Kojima et al . , Nature 402: 656-60 (1999). Compounds that bind GHS-R are then exposed to primary pituitary cells, for example, and release of growth hormone is determined using standard commercially available assays. Compounds that bind but do not stimulate the release of GH should then be assayed for ghrelin antagonism by exposing pituitary cells to the GHS-RA in the presence of ghrelin and then assaying for GH release.
Antibody-based GHS-RAs are also consistent with the claimed method. Anti-GHS-R antibodies may be generated by a variety of well-known methods that include traditional antisera production and monoclonal antibody techniques.
Modified antibody forms described above may then be produced using established techniques. Once generated, the antibodies are checked for GHS-RA activity in the manner described above. Ghrelin neutralizing agents (GNAs) represent another aspect of the invention. In this embodiment, ghrelin is neutralized or otherwise rendered biologically inactive apart from the receptor. Agents suitable for this application are those which specifically bind ghrelin, preferably with a higher affinity constant than the GHS-R. Antibody or antibody-based agents are preferred because they can be purposefully generated using well established techniques. Kojima et al . , Nature 402: 656-60 (1999) . Immunoadhesions (Fc fusion constructs, similar to Enbrel®, where the soluble ligand-binding domain of the GHS- R is fused to a human Fc) are also consistent with this aspect of the invention.
Dosages and desired drug concentration of pharmaceutical compositions of the present invention may vary depending on the particular use envisioned. The determination of the appropriate dosage or route of administration is well within the skill of an ordinary artisan. Animal experiments provide reliable guidance for the determination of effective doses for human therapy.
In another embodiment of the invention, an article of manufacture containing materials useful in the presently claimed methods is provided. The article of manufacture comprises a container and a label. Suitable containers include, for example, bottles, vials, syringes, and test tubes . The containers may be formed from a variety of materials such as glass or plastic. The container holds a composition which is effective for specifically inhibiting ghrelin action and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle) . The active agent in the composition is a GHS-RA and/or a GNA. The label on, or associated with, the container indicates that the composition is used for treating obesity and/or related disorders. The article of manufacture may further comprise a second container comprising a pharmaceutically-acceptable buffer, such as phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial end user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
The following examples are offered for illustrative purposes only, and are not intended to limit the scope of the present invention in any way.
Example 1 Ghrelin Synthesis Rat ghrelin was synthesized on an Advanced ChemTech® 396 synthesizer with FMOC amino acids and 50 minute diisopropylcarbodiimide (DIC) / 1- hydroxibenzotriazole (HOBT) activated double couplings. FMOC-SER(Trt) was used in the couplings for Ser3. Following trityl deprotection using l%TFA/5%tri-isopropylsilane in methylene chloride (DCM) , the Ser3-hydroxyl was acylated using excess octanoic acid and 1, 3 [ (Dimethylamino)propyl] -3- ethylcarbodii ide hydrochloride salt (EDAC) in the presence of 4-dimethylaminopyridine (DMAP) . After removal of the N- ter inal FMOC, a 2 hours cleavage was run using Reagent K. The precipitated peptide was washed with ethyl ether and dried in vacuo. The material was dissolved in aqueous acetic acid and purified over a 2.2x25 cm VydacClδ column using a gradient of 15%A to 55%B over 450 min (A=0.1%TFA, B=0.1%TFA/50%CH3CN) . Five-minute fractions were collected while monitoring the U.V. at 214 nm (2.0A). The appropriate fractions were combined, frozen and lyophilized. MALDI- ass spectral analysis indicated a mass of 3313.85 g for the purified ghrelin, which was consistent with the theoretical molecular weight.
We tested ono-octanoylated ghrelin and tested for ability to release GH in a primary rat pituitary cell assay.
Example 2' Animals Wild-type mice (129SV strain) and NPY-knockout mice were obtained from Taconic Farms®. Eight-week old dwarf rats were purchased at Harlan UK. Animals were housed individually in a temperature controlled environment (25 C°) with a 12-hour light and 12-hour dark (18.00 - 06.00) photoperiod. All mice had ad libi tum access to pelleted mouse food (5008 PMI® Nutrition International) and tap water . Mice were between 9 and 13 weeks of age and were injected daily between 17.00 and 18.00 with 0.1 ml of phosphate buffered saline (PBS) containing 0 or 8 mg/kg/d ghrelin over 13 days . Food intake and body weights were measured daily at Oδ.OOh. All animal experiments were conducted in accordance with the principles and procedures outlined in the National Institute of Health (NIH) Guide for the Care and Use of Laboratory Animals.
Example 3 Indirect Calorimetry
Twenty-four hour energy expenditure (EE) and respiratory quotient (RQ) were measured by indirect calorimetry using an open circuit calorimetry system (Oxymax, Columbus Instruments International Corporation; Columbus, OH) . The instrument was calibrated before each experiment using standard gas mixtures containing known concentrations of CO2, N2 and 02. After the first daily injection, animals were placed in calorimeter chambers containing food and water in a room maintained under identical conditions as those described above throughout the treatment period. Gas sampled from each of 10 chambers was first dried by a condenser. The volume of oxygen consumed (V02) and carbon dioxide produced (VC02) n an hour was measured using a paramagnetic oxygen sensor and a spectrophotometric C02 sensor. Such measurements were obtained hourly for 24 hours. RQ is the ratio of VC02 to V02. EE was calculated as the product of calorific value of oxygen (CV) and V02 per kilogram (kg) of body weight; where CV = 3.815 + 1.232*RQ (Elia, M. & Livesey, G, World Rev Nutr Diet 70, 68-131 (1992)). Total calories expended were calculated to determine daily fuel utilization. To calculate proportion of protein, fat and carbohydrate that is utilized during that 24-hour period, we used Flatt's proposal and assumed that protein utilization was equivalent to protein intake for adult stable animals (Flatt, J.P., J. Nutr Biochem 2, 193-202 (1991)). Using formulae and constants derived by Elia and Livesey (Elia, M. & Livesey, G., World Rev Nutr Diet, 70, 68-131 (1992)), we calculated the percent of daily fuel utilization derived from carbohydrate and fat. Daily caloric intake was calculated as (mass of daily food intake in g) * (physiological fuel value of the diet in kcal/g) . Locomotor activity was measured by counting the number of times an animal breaks a new light beam during each of 24 hours in the calorimeter.
Example 4
In-vivo analysis of body composition by dual-energy X-ray absorptiometry (DXA)
Body composition was measured on day 14 of the treatment period by DXA using a Norland p-DEXA® (Norland,
USA) . The system provides a non-invasive method for quantification of whole body composition and is based on the differential attenuation of high and low energy x-rays by the tissues in the scan area. Soft tissues attenuate the energy beam less than bone; of the soft tissue mass, fat tissue attenuates the beam less than lean tissue. Fat mass consists primarily of adipose tissue, but lean mass includes organs, tendons, cartilage, blood and body water in addition to skeletal muscle. In the present study, fat mass, lean mass and bone mineral content (bone mass) were measured and reported. Mice were anesthetized with inhalation of isoflorane and placed on the instrument platform in ventral position. Measurements were performed at a speed of 10 mm/ in and a resolution of 0.5 x 0.5 mm. Quality controls using phantom ID2232 and Calibration Standard 82315 (Norland) were performed before starting measurements . Example 5
In Vivo Administration Mice were treated with GHRP-2 for 18 days. A dose- dependent increase (n=42,p=0.001) in food intake and body weight was observed. A significant increase in fat mass (p=0.002) and bone mass (p=0.017) with no change in lean mass (p=0.63) was measured by dual-energy-X-ray- absorptiometry. This was partially a consequence of decreased (p=0.02) lipid utilization measured by indirect calorimetry. Hypothalamic RNA levels (measured by RT-PCR) of neuropeptide Y (NPY) , agouti-related-protein (AGRP) , pro- opio-melanocortin (POMC) and melanocyte-concentrating hormone (MCH) were not changed. Since GHRP-6 increases c- fos expression in NPY-neurons (Vernon, R. G.; J Endocrinol 150, 129-40 (1996)) and because these neurons also release AGRP, a natural melanocyte stimulating hormone antagonist, GHRP-2 treatment was repeated in NPY-knockout mice (NPY-/-) . Again, GHRP-2 induced a positive energy balance. However, an increase in AGRP mRNA levels (p=0.008,n=24 in GHRP-2 treated (NPY-/-)) was observed. Plasma levels of IGF-I, insulin, glucose and corticosterone were not „changed. Thus, peripheral administration of GHRP-2 induces a positive energy balance and fat gain by a hypothalamic mechanism. 200 μg of rat ghrelin was injected subcutaneously into wild-type mice, GHRP-2 or vehicle (phosphate buffered saline) . After 5 days of treatment, body weight increased (p=0.00) 12 % in both ghrelin- and GHRP-2- treated mice but not in controls. This weight gain was a consequence of decreased energy expenditure and decreased lipid utilization. Similar data have been observed in hypophysectomized rats indicating that GH and the other pituitary hormones do not mediate this anabolic activity. Such data indicate that the new stomach hormone, ghrelin, is a powerful stimulator of caloric accretion and that hypersecretion of ghrelin creates an obese state. Example 6 Pituitary Cell Culture Assay for Growth Hormone Secretion
Thirty-two 250 g male Sprague-Dawley rats are used for each assay. The animals are killed by decapitation and anterior pituitaries are removed and placed into ice cold culture medium. The pituitaries are sectioned into eighths and enzymatically digested using trypsin (Sigma Chemical) to weaken connective tissue. Pituitary cells are dispersed by mechanical agitation, collected, pooled and then seeded into 24-well plates (300,000 cells/well). After 4 days of culture, the cells form an even monolayer. Cells are then washed with medium and challenged to secrete GH by the addition of varying log concentrations of grhelin and the test compound to the medium. After 15 min at 37 °C, the medium is removed and stored frozen until standard radioimmunoassays for rat GH can be performed.
Example 7 In vitro Receptor Binding Assay
Recombinant CHO cells expressing the human growth hormone secretagogue receptor cDNA described by Howard et al . , Science 273: 974-977 (1996) are grown and harvested in nutrient medium. Membrane preparations are then obtained by first washing the cells with PBS buffer, then twice washing with cold buffer (25 mM HEPES, 2 M MgCl2, 1 mM EDTA, 20 μg/ml Leupeptin, 1 mM PMSF, 2μg/ml Aprotinin, 50 μg/ml Trypsin Inhibitor, pH 8.0) and resuspending in buffer. The cell suspension is lysed in a glass Teflon® homogenizer, and the resulting sample is then centrifuged at 35,300 X g for 30 minutes at 4°C.
The supernatant is removed, and the pellet is resuspended in cold buffer and homogenized. Aliquots may then be prepared and stored at -80°C.
A sample of the membrane preparation is pre- incubated with a test compound or a control compound with and without added grhelin in buffer (25 mM HEPES, 0.2%
(w/v) BSA, pH 7.6) at 32°C for 10 minutes. Reaction buffer (final concentration: 25 mM HEPES, 0.2% (w/v)
BSA, 2.6 mM Mg, 0.8 mM ATP, 0.1 mM GTP, 5 mM creatine phosphate, creatine kinase 50 U/ml, 0.2 mM IBMX, pH 7.6) is added and incubated for an additional 30 minutes. Incubations are stopped by adding 10 mM EDTA.
Production of cAMP is assayed using a fluorescent tracer-immuno assay method. In brief, after the incubation is stopped, fluorescent tracer (cAMP-b phycoerythrin conjugate) is added followed by the addition of affinity purified anti-cAMP rabbit antiserum. After incubation at room temperature for 45 minutes, anti-rabbit IgG coated assay beads are added and incubated for an additional 15 minutes. Plates are then evacuated and read on a Pandex® PFCIA reader. In this assay, ghrelin binding shows decreasing fluorescent intensity due to increased cAMP concentration. Fluorescent intensity values are correlated to rate of cAMP production (pmol/min/mg) . Conversely, inhibition of ghrelin binding by either receptor blockade or ghrelin neutralizations shows no decrease in fluorescent intensity.
Example 8 Rat ghrelin Response to fasting and refeeding
Because ghrelin is mainly generated by the stomach and secreted into circulation, we measured plasma ghrelin levels by radioimmunoassay. Elevated ghrelin levels after fasting in 250g male Sprague-Dawley rats (p=0.001) were decreased to normal levels (1.3±0.1 ng/ml) by re-feeding normal rat chow or by oral gavage of dextrose (p=0.001), but not after stomach expansion with water.
Example 9 Human ghrelin Response to glucose ingestion
Plasma ghrelin concentrations were measured in 5 women (BMI 23.5±3.3 kg/m2, body fat 23+35%) over 24 hours during which 3 meals (total energy = 1795±105 kcal) containing 55/30/15% of energy as carbohydrate, fat, and protein, respectively were consumed. When 30% of energy was in the form of a glucose-sweetened beverage, plasma ghrelin decreased by 30% 2 hours after the meal (p<0.01), and over the 24h sampling period (p<0.05). In contrast, when a fructose-containing beverage was consumed with each meal resulting in reduced postprandial glucose and insulin excursions, plasma ghrelin levels did not decrease after meals. We conclude that glucose ingestion and (or) the resulting insulin response appear to be candidates regulating ghrelin secretion. Further, we speculate that ghrelin release is a normal response to fasting. Such elevated ghrelin stimulates appetite and the utilization of carbohydrate (determined in above examples using rodents) and thus corrects hypoglycemia resulting from fasting. Ingestion of glucose rescues hypogylcemia and thus inhibits ghrelin secretion from the stomach to prevent hyperglycemia . Thus, ghrelin plays an important role in the regulation of blood glucose. Agents that block ghrelin action may be useful for the treatment of diabetes .

Claims

WE CLAIM:
1) A method of selectively inhibiting ghrelin activity in a mammal comprising administering to a mammal in need thereof a therapeutically-effective amount of a compound selected from the group consisting of a growth hormone secretagogue receptor antagonist (GHS-RA) and a ghrelin neutralizing agent (GNA) .
2) A method for treating obesity and related disorders in a mammal comprising administering to a mammal in need thereof a therapeutically-effective amount of a compound selected from the group consisting of a growth hormone secretagogue receptor antagonist (GHS-RA) and a ghrelin neutralizing agent (GNA) .
3) The method of any one of Claims 1 to 2 wherein the mammal is a human.
4) The method of any one of Claims 1 to 3 wherein the compound is a GHS-RA.
5) The method of Claim 4 wherein the GHS-RA is chosen from the group consisting of an isolated natural product, a synthetic organic compound, a protein, a peptide, an antibody, an antibody fragment, a single chain antibody, and an antibody-based construct.
6) The method of any on of Claims 1 to 3 wherein the compound is a GNA.
7) The method of Claim 6 wherein the GNA is selected from the group consisting of an antibody, an antibody fragment, a single chain antibody, and an antibody-based construct.
8) A method of assaying a compound for activity as a growth hormone secretagogue receptor antagonist (GHS-RA) comprising: a) preparing a mixture of the compound, and isolated pituitary cells; b) allowing said mixture to incubate for a period of time under conditions sufficient to permit binding; c) adding ghrelin to said mixture; and, d) measuring the release of growth hormone after a period of time.
9) A method of assaying a compound for activity as a ghrelin neutralizing agent (GNA) comprising: a) preparing a mixture of the compound, and ghrelin; b) allowing said mixture to incubate for a period of time under conditions sufficient to permit binding; c) adding isolated pituitary cells to said mixture; and, d) measuring the release of growth hormone after a period of time.
10) A method of assaying a compound for activity as a growth hormone secretagogue receptor antagonist (GHS-RA) comprising: a) preparing a mixture of the compound, and isolated pituitary cells; b) allowing said mixture to incubate for a period of time under conditions sufficient to permit binding; c) adding ghrelin to said mixture; d) assaying for levels of cAMP; and e) comparing the level of cAMP to control levels.
11) A method of assaying a compound for activity as a ghrelin neutralizing agent (GNA) comprising: a) preparing a mixture of the compound and ghrelin; b) allowing said mixture to incubate for a period of time under conditions sufficient to permit binding; c) adding isolated pituitary cells to said mixture; d) assaying for levels of cAMP; and e) comparing the level of cAMP to control levels.
12 ) An in vivo method of assaying a compound for activity as a GHS-RA or a GNA comprising: a) optionally fasting a rodent for at least 24 hours ; b) dosing the rodent with the compound; c) allowing the rodent to eat ad libi tum for at least 24 hours; and, d) comparing fat deposition, food intake, energy expenditure, and/or respiratory quotient to control rodents .
13) A pharmaceutical formulation comprising a GHS-RA and/or a GNA in combination with a pharmaceutically- acceptable carrier, diluent, or excipient for use in inhibiting ghrelin action.
14) A pharmaceutical formulation comprising a GHS-RA and/or a GNA in combination with a pharmaceutically acceptable carrier, diluent, or excipient for use in treating obesity and related disorders.
15) The use of a GHS-RA or a GNA for the manufacture of a medicament that selectively inhibits ghrelin action.
16) The use of a GHS-RA or a GNA for the manufacture of a medicament for treatment of obesity and related disorders.
17) An article of manufacture comprising a container, label, and therapeutically effective amount of GHS-RA and/or GNA in combination with a pharmaceutically-acceptable carrier.
PCT/US2001/011752 2000-05-17 2001-05-07 Method for selectively inhibiting ghrelin action WO2001087335A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
AU2001259056A AU2001259056A1 (en) 2000-05-17 2001-05-07 Method for selectively inhibiting ghrelin action
US10/258,259 US20030211967A1 (en) 2001-05-07 2001-05-07 Method for selectively inhibiting ghrelin action
EP01932539A EP1286697A2 (en) 2000-05-17 2001-05-07 Method for selectively inhibiting ghrelin action
US11/106,899 US20050201938A1 (en) 2000-05-17 2005-04-15 Method for selectively inhibiting ghrelin action

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US20489700P 2000-05-17 2000-05-17
US60/204,897 2000-05-17

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/106,899 Continuation US20050201938A1 (en) 2000-05-17 2005-04-15 Method for selectively inhibiting ghrelin action

Publications (2)

Publication Number Publication Date
WO2001087335A2 true WO2001087335A2 (en) 2001-11-22
WO2001087335A3 WO2001087335A3 (en) 2002-05-16

Family

ID=22759933

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2001/011752 WO2001087335A2 (en) 2000-05-17 2001-05-07 Method for selectively inhibiting ghrelin action

Country Status (3)

Country Link
EP (1) EP1286697A2 (en)
AU (1) AU2001259056A1 (en)
WO (1) WO2001087335A2 (en)

Cited By (77)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002060472A1 (en) * 2001-01-31 2002-08-08 Chugai Seiyaku Kabushiki Kaisha Remedies for hyponutrition status
WO2002090387A1 (en) * 2001-05-10 2002-11-14 Queensland University Of Technology Reproductive cancer diagnosis and therapy
WO2003051389A3 (en) * 2001-12-18 2003-09-12 Theratechnologies Inc Pharmaceutical compositions comprising unacylated ghrelin and therapeutical uses thereof
WO2004002986A2 (en) 2002-06-28 2004-01-08 Banyu Pharmaceutical Co., Ltd. Novel benzimidazole derivatives
WO2004004772A1 (en) * 2002-07-05 2004-01-15 Chugai Seiyaku Kabushiki Kaisha Remedy for diabetes
WO2004013274A2 (en) 2002-08-01 2004-02-12 Noxxon Pharma Ag Ghrelin binding nucleic acids
WO2004024183A1 (en) * 2002-09-12 2004-03-25 Pharmexa A/S Immunization against autologous ghrelin
US6716629B2 (en) 2000-10-10 2004-04-06 Biotrove, Inc. Apparatus for assay, synthesis and storage, and methods of manufacture, use, and manipulation thereof
WO2004056869A1 (en) * 2002-12-20 2004-07-08 7Tm Pharma A/S Ghrelin receptor inverse agonist for regulation of feeding behaviours
WO2004084943A1 (en) * 2003-03-19 2004-10-07 Beth Israel Deaconess Medical Center, Inc Use of antagonists of ghrelin or ghrelin receptor to treat intestinal inflammation
WO2005000343A1 (en) * 2003-06-11 2005-01-06 Allergan, Inc. Use of a clostridial toxin to reduce appetite
WO2005026211A2 (en) * 2003-09-05 2005-03-24 Eli Lilly And Company Anti-ghrelin antibodies
EP1546389A2 (en) * 2002-09-06 2005-06-29 Elixir Pharmaceuticals, Inc. Regulation of the growth hormone/igf-1 axis
WO2005097830A2 (en) * 2004-04-07 2005-10-20 Aditech Pharma Ab Uses of isolated binding members capable of reducing the biological activity of secretagogue compounds
WO2005097831A2 (en) * 2004-04-07 2005-10-20 Gastrotech Pharma A/S Uses of isolated binding members capable of binding specifically to secretagogues
WO2005113600A2 (en) * 2004-05-18 2005-12-01 Haptogen Ltd Methods for the control, treatment and management of obesity
WO2005114180A2 (en) * 2004-05-14 2005-12-01 Novo Nordisk A/S Functional ghs-r antagonists
WO2006019577A1 (en) * 2004-07-14 2006-02-23 Eli Lilly And Company Anti-ghrelin antibodies
EP1694355A1 (en) * 2003-10-24 2006-08-30 THE ACADEMIC HOSPITAL ROTTERDAM acting under the name Erasmus MC Use of ghrelin and unacylated ghrelin compostions in insulin-related disease conditions
WO2006091381A1 (en) * 2005-02-23 2006-08-31 Eli Lilly And Company Humanized anti-ghrelin antibodies
WO2007041052A2 (en) 2005-09-29 2007-04-12 Merck & Co., Inc. Acylated spiropiperidine derivatives as melanocortin-4 receptor modulators
JP2007513608A (en) * 2003-11-10 2007-05-31 ノクソン・フアルマ・アクチエンゲゼルシヤフト Nucleic acids that specifically bind to biologically active ghrelin
US7252665B2 (en) 2003-10-31 2007-08-07 Medtronic, Inc Ablation of stomach lining to reduce stomach acid secretion
WO2007095347A2 (en) * 2006-02-13 2007-08-23 The Gov. Of The Usa, As Represented By The Secretary, Dept. Of Health And Human Services Methods and compositions related to ghs-r antagonists
US7282050B2 (en) 2003-10-31 2007-10-16 Medtronic, Inc. Ablation of exterior of stomach to treat obesity
WO2008060476A2 (en) 2006-11-15 2008-05-22 Schering Corporation Nitrogen-containing heterocyclic compounds and methods of use thereof
EP2088154A1 (en) 2004-03-09 2009-08-12 Ironwood Pharmaceuticals, Inc. Methods and compositions for the treatment of gastrointestinal disorders
WO2009110510A1 (en) 2008-03-06 2009-09-11 萬有製薬株式会社 Alkylaminopyridine derivative
EP2127676A2 (en) 2004-11-01 2009-12-02 Amylin Pharmaceuticals, Inc. Treatment of obesity and related disorders
US7666833B2 (en) 2001-12-18 2010-02-23 Alizé Pharma SAS Pharmaceutical compositions comprising unacylated ghrelin and therapeutical uses thereof
WO2010047982A1 (en) 2008-10-22 2010-04-29 Merck Sharp & Dohme Corp. Novel cyclic benzimidazole derivatives useful anti-diabetic agents
WO2010051236A1 (en) 2008-10-30 2010-05-06 Merck Sharp & Dohme Corp. Isonicotinamide orexin receptor antagonists
WO2010051206A1 (en) 2008-10-31 2010-05-06 Merck Sharp & Dohme Corp. Novel cyclic benzimidazole derivatives useful anti-diabetic agents
US7736392B2 (en) 2005-04-28 2010-06-15 Medtronic, Inc. Bulking of upper esophageal sphincter for treatment of obesity
WO2010075068A1 (en) 2008-12-16 2010-07-01 Schering Corporation Pyridopyrimidine derivatives and methods of use thereof
WO2010075069A1 (en) 2008-12-16 2010-07-01 Schering Corporation Bicyclic pyranone derivatives as nicotinic acid receptor agonists
EP2305352A1 (en) 2004-04-02 2011-04-06 Merck Sharp & Dohme Corp. 5-alpha-reductase inhibitors for use in the treatment of men with metabolic and anthropometric disorders
WO2011053821A1 (en) 2009-10-30 2011-05-05 Tranzyme Pharma, Inc. Macrocyclic ghrelin receptor antagonists and inverse agonists and methods of using the same
EP2330124A2 (en) 2005-08-11 2011-06-08 Amylin Pharmaceuticals Inc. Hybrid polypeptides with selectable properties
EP2330125A2 (en) 2005-08-11 2011-06-08 Amylin Pharmaceuticals, Inc. Hybrid polypeptides with selectable properties
WO2011069038A2 (en) 2009-12-03 2011-06-09 Synergy Pharmaceuticals, Inc. Agonists of guanylate cyclase useful for the treatment of hypercholesterolemia, atherosclerosis, coronary heart disease, gallstone, obesity and other cardiovascular diseases
WO2011106273A1 (en) 2010-02-25 2011-09-01 Merck Sharp & Dohme Corp. Novel cyclic benzimidazole derivatives useful anti-diabetic agents
US8101734B2 (en) 2005-04-08 2012-01-24 Noxxon Pharma Ag Ghrelin binding nucleic acids
US8101576B2 (en) 2006-12-13 2012-01-24 Imperial Innovations Limited Compounds and their effects on feeding behaviour
US8217001B2 (en) 2001-09-24 2012-07-10 Imperial Innovations Limited Modification of feeding behavior
US8222217B2 (en) 2007-05-31 2012-07-17 Alize Pharma Sas Unacylated ghrelin as therapeutic agent in the treatment of metabolic disorders
WO2012116145A1 (en) 2011-02-25 2012-08-30 Merck Sharp & Dohme Corp. Novel cyclic azabenzimidazole derivatives useful as anti-diabetic agents
US8318664B2 (en) 2007-05-31 2012-11-27 Alize Pharma Sas Unacylated ghrelin fragments as therapeutic agent in the treatment of obesity
WO2013059222A1 (en) 2011-10-19 2013-04-25 Merck Sharp & Dohme Corp. 2-pyridyloxy-4-nitrile orexin receptor antagonists
US8440628B2 (en) * 2002-05-21 2013-05-14 Daiichi Sankyo Company, Limited Medical compositions containing ghrelin
US8476408B2 (en) 2008-06-13 2013-07-02 Alize Pharma Sas Unacylated ghrelin and analogs as therapeutic agents for vascular remodeling in diabetic patients and treatment of cardiovascular disease
WO2013138352A1 (en) 2012-03-15 2013-09-19 Synergy Pharmaceuticals Inc. Formulations of guanylate cyclase c agonists and methods of use
WO2014022528A1 (en) 2012-08-02 2014-02-06 Merck Sharp & Dohme Corp. Antidiabetic tricyclic compounds
EP2698157A1 (en) 2006-09-22 2014-02-19 Merck Sharp & Dohme Corp. Method of treatment using fatty acid synthesis inhibitors
WO2014130608A1 (en) 2013-02-22 2014-08-28 Merck Sharp & Dohme Corp. Antidiabetic bicyclic compounds
WO2014139388A1 (en) 2013-03-14 2014-09-18 Merck Sharp & Dohme Corp. Novel indole derivatives useful as anti-diabetic agents
WO2014151206A1 (en) 2013-03-15 2014-09-25 Synergy Pharmaceuticals Inc. Agonists of guanylate cyclase and their uses
EP2810951A2 (en) 2008-06-04 2014-12-10 Synergy Pharmaceuticals Inc. Agonists of guanylate cyclase useful for the treatment of gastrointestinal disorders, inflammation, cancer and other disorders
WO2014197720A2 (en) 2013-06-05 2014-12-11 Synergy Pharmaceuticals, Inc. Ultra-pure agonists of guanylate cyclase c, method of making and using same
US8992928B2 (en) 2006-02-11 2015-03-31 Victor Raso Isolated monoclonal antibody or antigen-binding fragment that cleaves octanoylated native ghrelin
WO2015054500A2 (en) 2013-10-09 2015-04-16 Synergy Pharmaceuticals, Inc. Agonists of guanylate cyclase useful for downregulation of pro-inflammatory cytokines
WO2015051725A1 (en) 2013-10-08 2015-04-16 Merck Sharp & Dohme Corp. Antidiabetic tricyclic compounds
WO2016030534A1 (en) 2014-08-29 2016-03-03 Tes Pharma S.R.L. INHIBITORS OF α-AMINO-β-CARBOXYMUCONIC ACID SEMIALDEHYDE DECARBOXYLASE
EP2998314A1 (en) 2007-06-04 2016-03-23 Synergy Pharmaceuticals Inc. Agonists of guanylate cyclase useful for the treatment of gastrointestinal disorders, inflammation, cancer and other disorders
US9314764B2 (en) 2000-10-10 2016-04-19 Life Technologies Corporation Apparatus for assay, synthesis and storage, and methods of manufacture, use, and manipulation thereof
US9550821B2 (en) 2011-12-15 2017-01-24 Alize Pharma Sas Modulation of ghrelin levels and ghrelin/unacylated ghrelin ratio using unacylated ghrelin
EP3241839A1 (en) 2008-07-16 2017-11-08 Synergy Pharmaceuticals Inc. Agonists of guanylate cyclase useful for the treatment of gastrointestinal, inflammation, cancer and other disorders
WO2018069532A1 (en) 2016-10-14 2018-04-19 Tes Pharma S.R.L. Inhibitors of alpha-amino-beta-carboxymuconic acid semialdehyde decarboxylase
WO2018106518A1 (en) 2016-12-06 2018-06-14 Merck Sharp & Dohme Corp. Antidiabetic heterocyclic compounds
WO2018118670A1 (en) 2016-12-20 2018-06-28 Merck Sharp & Dohme Corp. Antidiabetic spirochroman compounds
US10065189B2 (en) 2004-03-12 2018-09-04 Life Technologies Corporation Nanoliter array loading
US10213761B2 (en) 2004-08-04 2019-02-26 Life Technologies Corporation Coating process for microfluidic sample arrays
US10227644B2 (en) 2000-02-18 2019-03-12 The Board Of Trustees Of The Leland Stanford Junior University Apparatus and methods for parallel processing of microvolume liquid reactions
WO2020104456A1 (en) 2018-11-20 2020-05-28 Tes Pharma S.R.L INHIBITORS OF α-AMINO-β-CARBOXYMUCONIC ACID SEMIALDEHYDE DECARBOXYLASE
WO2020167706A1 (en) 2019-02-13 2020-08-20 Merck Sharp & Dohme Corp. 5-alkyl pyrrolidine orexin receptor agonists
WO2021026047A1 (en) 2019-08-08 2021-02-11 Merck Sharp & Dohme Corp. Heteroaryl pyrrolidine and piperidine orexin receptor agonists
WO2022040070A1 (en) 2020-08-18 2022-02-24 Merck Sharp & Dohme Corp. Bicycloheptane pyrrolidine orexin receptor agonists

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7682565B2 (en) 2002-12-20 2010-03-23 Biotrove, Inc. Assay apparatus and method using microfluidic arrays

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997021730A1 (en) * 1995-12-13 1997-06-19 Merck & Co., Inc. Growth hormone secretagogue receptor family
WO2000002919A1 (en) * 1998-07-13 2000-01-20 Merck & Co., Inc. Growth hormone secretagogue related receptors and nucleic acids
WO2000009538A2 (en) * 1998-08-10 2000-02-24 Merck & Co., Inc. Canine growth hormone secretagogue receptor

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997021730A1 (en) * 1995-12-13 1997-06-19 Merck & Co., Inc. Growth hormone secretagogue receptor family
WO2000002919A1 (en) * 1998-07-13 2000-01-20 Merck & Co., Inc. Growth hormone secretagogue related receptors and nucleic acids
WO2000009538A2 (en) * 1998-08-10 2000-02-24 Merck & Co., Inc. Canine growth hormone secretagogue receptor

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
KOJIMA MASAYASU ET AL: "Ghrelin is a growth-hormone-releasing acylated peptide from stomach." NATURE (LONDON), vol. 402, no. 6762, 9 December 1999 (1999-12-09), pages 656-660, XP002187791 ISSN: 0028-0836 cited in the application *
SMITH R G ET AL: "A nonpeptidyl growth hormone secretagogue." SCIENCE, (1993 JUN 11) 260 (5114) 1640-3. , XP002187790 cited in the application *

Cited By (111)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10378049B2 (en) 2000-02-18 2019-08-13 The Board Of Trustees Of The Leland Stanford Junior University Apparatus and methods for parallel processing of microvolume liquid reactions
US10227644B2 (en) 2000-02-18 2019-03-12 The Board Of Trustees Of The Leland Stanford Junior University Apparatus and methods for parallel processing of microvolume liquid reactions
US9314764B2 (en) 2000-10-10 2016-04-19 Life Technologies Corporation Apparatus for assay, synthesis and storage, and methods of manufacture, use, and manipulation thereof
US6716629B2 (en) 2000-10-10 2004-04-06 Biotrove, Inc. Apparatus for assay, synthesis and storage, and methods of manufacture, use, and manipulation thereof
US9968903B2 (en) 2000-10-10 2018-05-15 Life Technologies Corporation Apparatus for assay, synthesis and storage, and methods of manufacture, use, and manipulation thereof
WO2002060472A1 (en) * 2001-01-31 2002-08-08 Chugai Seiyaku Kabushiki Kaisha Remedies for hyponutrition status
WO2002090387A1 (en) * 2001-05-10 2002-11-14 Queensland University Of Technology Reproductive cancer diagnosis and therapy
US8217001B2 (en) 2001-09-24 2012-07-10 Imperial Innovations Limited Modification of feeding behavior
US8071368B2 (en) 2001-12-18 2011-12-06 Alizé Pharma SAS Methods for promoting growth and survival of insulin-secreting cells
US7485620B2 (en) 2001-12-18 2009-02-03 Alizé Pharma SAS Pharmaceutical compositions comprising unacylated ghrelin and therapeutical uses thereof
WO2003051389A3 (en) * 2001-12-18 2003-09-12 Theratechnologies Inc Pharmaceutical compositions comprising unacylated ghrelin and therapeutical uses thereof
US7666833B2 (en) 2001-12-18 2010-02-23 Alizé Pharma SAS Pharmaceutical compositions comprising unacylated ghrelin and therapeutical uses thereof
US8518893B2 (en) 2002-05-21 2013-08-27 Asubio Pharma Co. Ltd. Medical compositions containing ghrelin
US8440628B2 (en) * 2002-05-21 2013-05-14 Daiichi Sankyo Company, Limited Medical compositions containing ghrelin
WO2004002986A2 (en) 2002-06-28 2004-01-08 Banyu Pharmaceutical Co., Ltd. Novel benzimidazole derivatives
WO2004004772A1 (en) * 2002-07-05 2004-01-15 Chugai Seiyaku Kabushiki Kaisha Remedy for diabetes
WO2004013274A2 (en) 2002-08-01 2004-02-12 Noxxon Pharma Ag Ghrelin binding nucleic acids
JP2006501864A (en) * 2002-08-01 2006-01-19 ノクソン・フアルマ・アクチエンゲゼルシヤフト Ghrelin binding nucleic acid
WO2004013274A3 (en) * 2002-08-01 2004-05-13 Noxxon Pharma Ag Ghrelin binding nucleic acids
US7750140B2 (en) 2002-08-01 2010-07-06 Noxxon Pharma Ag Ghrelin binding nucleic acids
EP1546389A4 (en) * 2002-09-06 2006-07-26 Elixir Pharmaceuticals Inc Regulation of the growth hormone/igf-1 axis
EP1546389A2 (en) * 2002-09-06 2005-06-29 Elixir Pharmaceuticals, Inc. Regulation of the growth hormone/igf-1 axis
WO2004024183A1 (en) * 2002-09-12 2004-03-25 Pharmexa A/S Immunization against autologous ghrelin
WO2004056869A1 (en) * 2002-12-20 2004-07-08 7Tm Pharma A/S Ghrelin receptor inverse agonist for regulation of feeding behaviours
WO2004084943A1 (en) * 2003-03-19 2004-10-07 Beth Israel Deaconess Medical Center, Inc Use of antagonists of ghrelin or ghrelin receptor to treat intestinal inflammation
WO2005000343A1 (en) * 2003-06-11 2005-01-06 Allergan, Inc. Use of a clostridial toxin to reduce appetite
WO2005026211A3 (en) * 2003-09-05 2005-06-16 Lilly Co Eli Anti-ghrelin antibodies
WO2005026211A2 (en) * 2003-09-05 2005-03-24 Eli Lilly And Company Anti-ghrelin antibodies
EP1694355A4 (en) * 2003-10-24 2009-07-29 Alize Pharma Sas Use of ghrelin and unacylated ghrelin compostions in insulin-related disease conditions
EP1694355A1 (en) * 2003-10-24 2006-08-30 THE ACADEMIC HOSPITAL ROTTERDAM acting under the name Erasmus MC Use of ghrelin and unacylated ghrelin compostions in insulin-related disease conditions
US7825090B2 (en) 2003-10-24 2010-11-02 Alizé Pharma SAS Use of ghrelin and unacylated ghrelin compositions for treating insulin resistance
US7252665B2 (en) 2003-10-31 2007-08-07 Medtronic, Inc Ablation of stomach lining to reduce stomach acid secretion
US7282050B2 (en) 2003-10-31 2007-10-16 Medtronic, Inc. Ablation of exterior of stomach to treat obesity
JP2007513608A (en) * 2003-11-10 2007-05-31 ノクソン・フアルマ・アクチエンゲゼルシヤフト Nucleic acids that specifically bind to biologically active ghrelin
JP4823067B2 (en) * 2003-11-10 2011-11-24 ノクソン・フアルマ・アクチエンゲゼルシヤフト Nucleic acids that specifically bind to biologically active ghrelin
EP2088154A1 (en) 2004-03-09 2009-08-12 Ironwood Pharmaceuticals, Inc. Methods and compositions for the treatment of gastrointestinal disorders
US10974247B2 (en) 2004-03-12 2021-04-13 Life Technologies Corporation Nanoliter array loading
US10065189B2 (en) 2004-03-12 2018-09-04 Life Technologies Corporation Nanoliter array loading
EP2305352A1 (en) 2004-04-02 2011-04-06 Merck Sharp & Dohme Corp. 5-alpha-reductase inhibitors for use in the treatment of men with metabolic and anthropometric disorders
WO2005097830A3 (en) * 2004-04-07 2005-12-22 Aditech Pharma Ab Uses of isolated binding members capable of reducing the biological activity of secretagogue compounds
WO2005097830A2 (en) * 2004-04-07 2005-10-20 Aditech Pharma Ab Uses of isolated binding members capable of reducing the biological activity of secretagogue compounds
WO2005097831A2 (en) * 2004-04-07 2005-10-20 Gastrotech Pharma A/S Uses of isolated binding members capable of binding specifically to secretagogues
WO2005097831A3 (en) * 2004-04-07 2005-12-22 Gastrotech Pharma As Uses of isolated binding members capable of binding specifically to secretagogues
WO2005112903A2 (en) * 2004-05-14 2005-12-01 Novo Nordisk A/S Use of ghrelin antagonists for improving cognition and memory
WO2005114180A2 (en) * 2004-05-14 2005-12-01 Novo Nordisk A/S Functional ghs-r antagonists
WO2005112903A3 (en) * 2004-05-14 2006-02-02 Novo Nordisk As Use of ghrelin antagonists for improving cognition and memory
WO2005114180A3 (en) * 2004-05-14 2006-04-13 Novo Nordisk As Functional ghs-r antagonists
WO2005113600A2 (en) * 2004-05-18 2005-12-01 Haptogen Ltd Methods for the control, treatment and management of obesity
WO2005113600A3 (en) * 2004-05-18 2006-06-08 Haptogen Ltd Methods for the control, treatment and management of obesity
WO2006019577A1 (en) * 2004-07-14 2006-02-23 Eli Lilly And Company Anti-ghrelin antibodies
US11154834B2 (en) 2004-08-04 2021-10-26 Life Technologies Corporation Coating process for microfluidic sample arrays
US10213761B2 (en) 2004-08-04 2019-02-26 Life Technologies Corporation Coating process for microfluidic sample arrays
EP2286837A2 (en) 2004-11-01 2011-02-23 Amylin Pharmaceuticals, Inc. Treatment of obesity and obesity related diseases
EP2286839A2 (en) 2004-11-01 2011-02-23 Amylin Pharmaceuticals, Inc. Treatment of obesity and related diseases
EP2286840A2 (en) 2004-11-01 2011-02-23 Amylin Pharmaceuticals, Inc. Treatment of obesity and related diseases
EP2286838A2 (en) 2004-11-01 2011-02-23 Amylin Pharmaceuticals, Inc. Treatment of obesity and related disorders
EP2127676A2 (en) 2004-11-01 2009-12-02 Amylin Pharmaceuticals, Inc. Treatment of obesity and related disorders
WO2006091381A1 (en) * 2005-02-23 2006-08-31 Eli Lilly And Company Humanized anti-ghrelin antibodies
US7479271B2 (en) 2005-02-23 2009-01-20 Eli Lilly And Company Humanized anti-ghrelin antibodies
US8101734B2 (en) 2005-04-08 2012-01-24 Noxxon Pharma Ag Ghrelin binding nucleic acids
US7736392B2 (en) 2005-04-28 2010-06-15 Medtronic, Inc. Bulking of upper esophageal sphincter for treatment of obesity
EP2330124A2 (en) 2005-08-11 2011-06-08 Amylin Pharmaceuticals Inc. Hybrid polypeptides with selectable properties
EP2330125A2 (en) 2005-08-11 2011-06-08 Amylin Pharmaceuticals, Inc. Hybrid polypeptides with selectable properties
WO2007041052A2 (en) 2005-09-29 2007-04-12 Merck & Co., Inc. Acylated spiropiperidine derivatives as melanocortin-4 receptor modulators
US8992928B2 (en) 2006-02-11 2015-03-31 Victor Raso Isolated monoclonal antibody or antigen-binding fragment that cleaves octanoylated native ghrelin
WO2007095347A3 (en) * 2006-02-13 2007-12-21 Us Gov Health & Human Serv Methods and compositions related to ghs-r antagonists
WO2007095347A2 (en) * 2006-02-13 2007-08-23 The Gov. Of The Usa, As Represented By The Secretary, Dept. Of Health And Human Services Methods and compositions related to ghs-r antagonists
EP2946778A1 (en) 2006-09-22 2015-11-25 Merck Sharp & Dohme Corp. Method of treatment using fatty acid synthesis inhibitors
EP2698157A1 (en) 2006-09-22 2014-02-19 Merck Sharp & Dohme Corp. Method of treatment using fatty acid synthesis inhibitors
WO2008060476A2 (en) 2006-11-15 2008-05-22 Schering Corporation Nitrogen-containing heterocyclic compounds and methods of use thereof
US8101576B2 (en) 2006-12-13 2012-01-24 Imperial Innovations Limited Compounds and their effects on feeding behaviour
US8318664B2 (en) 2007-05-31 2012-11-27 Alize Pharma Sas Unacylated ghrelin fragments as therapeutic agent in the treatment of obesity
US8222217B2 (en) 2007-05-31 2012-07-17 Alize Pharma Sas Unacylated ghrelin as therapeutic agent in the treatment of metabolic disorders
EP2998314A1 (en) 2007-06-04 2016-03-23 Synergy Pharmaceuticals Inc. Agonists of guanylate cyclase useful for the treatment of gastrointestinal disorders, inflammation, cancer and other disorders
WO2009110510A1 (en) 2008-03-06 2009-09-11 萬有製薬株式会社 Alkylaminopyridine derivative
EP2810951A2 (en) 2008-06-04 2014-12-10 Synergy Pharmaceuticals Inc. Agonists of guanylate cyclase useful for the treatment of gastrointestinal disorders, inflammation, cancer and other disorders
US8476408B2 (en) 2008-06-13 2013-07-02 Alize Pharma Sas Unacylated ghrelin and analogs as therapeutic agents for vascular remodeling in diabetic patients and treatment of cardiovascular disease
EP3241839A1 (en) 2008-07-16 2017-11-08 Synergy Pharmaceuticals Inc. Agonists of guanylate cyclase useful for the treatment of gastrointestinal, inflammation, cancer and other disorders
WO2010047982A1 (en) 2008-10-22 2010-04-29 Merck Sharp & Dohme Corp. Novel cyclic benzimidazole derivatives useful anti-diabetic agents
WO2010051236A1 (en) 2008-10-30 2010-05-06 Merck Sharp & Dohme Corp. Isonicotinamide orexin receptor antagonists
WO2010051206A1 (en) 2008-10-31 2010-05-06 Merck Sharp & Dohme Corp. Novel cyclic benzimidazole derivatives useful anti-diabetic agents
WO2010075068A1 (en) 2008-12-16 2010-07-01 Schering Corporation Pyridopyrimidine derivatives and methods of use thereof
WO2010075069A1 (en) 2008-12-16 2010-07-01 Schering Corporation Bicyclic pyranone derivatives as nicotinic acid receptor agonists
WO2011053821A1 (en) 2009-10-30 2011-05-05 Tranzyme Pharma, Inc. Macrocyclic ghrelin receptor antagonists and inverse agonists and methods of using the same
WO2011069038A2 (en) 2009-12-03 2011-06-09 Synergy Pharmaceuticals, Inc. Agonists of guanylate cyclase useful for the treatment of hypercholesterolemia, atherosclerosis, coronary heart disease, gallstone, obesity and other cardiovascular diseases
WO2011106273A1 (en) 2010-02-25 2011-09-01 Merck Sharp & Dohme Corp. Novel cyclic benzimidazole derivatives useful anti-diabetic agents
WO2012116145A1 (en) 2011-02-25 2012-08-30 Merck Sharp & Dohme Corp. Novel cyclic azabenzimidazole derivatives useful as anti-diabetic agents
EP3243385A1 (en) 2011-02-25 2017-11-15 Merck Sharp & Dohme Corp. Novel cyclic azabenzimidazole derivatives useful as anti-diabetic agents
WO2013059222A1 (en) 2011-10-19 2013-04-25 Merck Sharp & Dohme Corp. 2-pyridyloxy-4-nitrile orexin receptor antagonists
US9550821B2 (en) 2011-12-15 2017-01-24 Alize Pharma Sas Modulation of ghrelin levels and ghrelin/unacylated ghrelin ratio using unacylated ghrelin
EP3708179A1 (en) 2012-03-15 2020-09-16 Bausch Health Ireland Limited Formulations of guanylate cyclase c agonists and methods of use
WO2013138352A1 (en) 2012-03-15 2013-09-19 Synergy Pharmaceuticals Inc. Formulations of guanylate cyclase c agonists and methods of use
EP4309673A2 (en) 2012-03-15 2024-01-24 Bausch Health Ireland Limited Formulations of guanylate cyclase c agonists and methods of use
WO2014022528A1 (en) 2012-08-02 2014-02-06 Merck Sharp & Dohme Corp. Antidiabetic tricyclic compounds
WO2014130608A1 (en) 2013-02-22 2014-08-28 Merck Sharp & Dohme Corp. Antidiabetic bicyclic compounds
WO2014139388A1 (en) 2013-03-14 2014-09-18 Merck Sharp & Dohme Corp. Novel indole derivatives useful as anti-diabetic agents
WO2014151206A1 (en) 2013-03-15 2014-09-25 Synergy Pharmaceuticals Inc. Agonists of guanylate cyclase and their uses
WO2014197720A2 (en) 2013-06-05 2014-12-11 Synergy Pharmaceuticals, Inc. Ultra-pure agonists of guanylate cyclase c, method of making and using same
WO2015051725A1 (en) 2013-10-08 2015-04-16 Merck Sharp & Dohme Corp. Antidiabetic tricyclic compounds
WO2015054500A2 (en) 2013-10-09 2015-04-16 Synergy Pharmaceuticals, Inc. Agonists of guanylate cyclase useful for downregulation of pro-inflammatory cytokines
US9708272B2 (en) 2014-08-29 2017-07-18 Tes Pharma S.R.L. Inhibitors of α-amino-β-carboxymuconic acid semialdehyde decarboxylase
WO2016030534A1 (en) 2014-08-29 2016-03-03 Tes Pharma S.R.L. INHIBITORS OF α-AMINO-β-CARBOXYMUCONIC ACID SEMIALDEHYDE DECARBOXYLASE
US10513499B2 (en) 2014-08-29 2019-12-24 Tes Pharma S.R.L. Inhibitors of alpha-amino-beta-carboxymuconic acid semialdehyde decarboxylase
US11254644B2 (en) 2014-08-29 2022-02-22 Tes Pharma S.R.L. Inhibitors of alpha-amino-beta-carboxymuconic acid semialdehyde decarboxylase
WO2018069532A1 (en) 2016-10-14 2018-04-19 Tes Pharma S.R.L. Inhibitors of alpha-amino-beta-carboxymuconic acid semialdehyde decarboxylase
WO2018106518A1 (en) 2016-12-06 2018-06-14 Merck Sharp & Dohme Corp. Antidiabetic heterocyclic compounds
WO2018118670A1 (en) 2016-12-20 2018-06-28 Merck Sharp & Dohme Corp. Antidiabetic spirochroman compounds
WO2020104456A1 (en) 2018-11-20 2020-05-28 Tes Pharma S.R.L INHIBITORS OF α-AMINO-β-CARBOXYMUCONIC ACID SEMIALDEHYDE DECARBOXYLASE
WO2020167706A1 (en) 2019-02-13 2020-08-20 Merck Sharp & Dohme Corp. 5-alkyl pyrrolidine orexin receptor agonists
WO2021026047A1 (en) 2019-08-08 2021-02-11 Merck Sharp & Dohme Corp. Heteroaryl pyrrolidine and piperidine orexin receptor agonists
WO2022040070A1 (en) 2020-08-18 2022-02-24 Merck Sharp & Dohme Corp. Bicycloheptane pyrrolidine orexin receptor agonists

Also Published As

Publication number Publication date
EP1286697A2 (en) 2003-03-05
WO2001087335A3 (en) 2002-05-16
AU2001259056A1 (en) 2001-11-26

Similar Documents

Publication Publication Date Title
US20050201938A1 (en) Method for selectively inhibiting ghrelin action
WO2001087335A2 (en) Method for selectively inhibiting ghrelin action
Castaneda et al. Ghrelin in the regulation of body weight and metabolism
RU2351359C2 (en) Application of oxyntomodulin, method and pharmaceutical composition for prevention or treatment of excessive body weight
JP6312748B2 (en) Glycoproteins with lipid mobilization properties and therapeutic uses thereof
JP2021185178A (en) Methods for treatment of bile acid-related disorders
BRPI0618160A2 (en) Alternative complement pathway inhibition for treatment of traumatic brain damage, spinal cord damage and related conditions
HU228621B1 (en) Modification of feeding behavior
KR20150104579A (en) Methods for modulating bile acid homeostasis and treatment of bile acid disorders and diseases
CN103732618A (en) Novel oxyntomodulin derivatives and pharmaceutical composition for treating obesity comprising same
BR112013033175A2 (en) peptide sequence, subsequence, composition, pharmaceutical composition, nucleic acid molecule, vector, transformed or host cell, method of treatment for an individual, method for improving glucose metabolism in an individual, and method for identifying a peptide sequence
JP2008518941A (en) Treatment of patients with short bowel syndrome with colorectal continuity
AU2016242935A1 (en) Methods for treating obesity and nonalcoholic fatty liver disease or nonalcoholic steatohepatitis using glucagon receptor antagonistic antibodies
US20220340668A1 (en) Treatment of liver disease or disorder comprising actrii receptor antagonists
JP7165495B2 (en) Motor control function improver
US20180340027A1 (en) Milk fat globule epidermal growth factor 8 regulates fatty acid uptake
US20060269550A1 (en) Anti-ghrelin fab antibodies
JP2011506371A (en) Treatment for pemphigus containing anti-Fas ligand antibody
JP5875190B2 (en) Methylglyoxal-scavenging compounds and their use for the prevention and treatment of pain and / or hyperalgesia
CN111905096A (en) Identification of novel polypeptide hormones for maintaining optimal body weight and blood glucose
EP3892289A1 (en) Pharmaceutical composition, comprising inhibitory peptide against fas signaling, for prevention or treatment of obesity, fatty liver, or steatohepatitis
JP2007523196A (en) Method for treating obesity or diabetes using NT-4 / 5
WO2006114284A2 (en) AGONISTIC ANTIBODIES THAT BIND TO THE LT-β-RECEPTOR AND THEREBY MODULATE ADIPOSITY-ASSOCIATED PHENOTYPES AS WELL AS THEIR USE IN THERAPY
US20240024426A1 (en) Hypotensive pharmaceutical composition comprising triple activator having activity for all of glucagon, glp-1, and gip receptors
CN110177567A (en) The pharmaceutical composition for the treatment of growth hormone deficiency containing hGH fusion protein

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
AK Designated states

Kind code of ref document: A3

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: 10258259

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2001932539

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2001932539

Country of ref document: EP

NENP Non-entry into the national phase in:

Ref country code: JP