MXPA02001882A - Modulation of the blood brain barrier transporter for leptin. - Google Patents

Modulation of the blood brain barrier transporter for leptin.

Info

Publication number
MXPA02001882A
MXPA02001882A MXPA02001882A MXPA02001882A MXPA02001882A MX PA02001882 A MXPA02001882 A MX PA02001882A MX PA02001882 A MXPA02001882 A MX PA02001882A MX PA02001882 A MXPA02001882 A MX PA02001882A MX PA02001882 A MXPA02001882 A MX PA02001882A
Authority
MX
Mexico
Prior art keywords
leptin
seq
adrenergic
group
mammal
Prior art date
Application number
MXPA02001882A
Other languages
Spanish (es)
Inventor
William A Banks
Original Assignee
Univ Tulane
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 Univ Tulane filed Critical Univ Tulane
Publication of MXPA02001882A publication Critical patent/MXPA02001882A/en

Links

Classifications

    • 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
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/137Arylalkylamines, e.g. amphetamine, epinephrine, salbutamol, ephedrine or methadone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid or pantothenic acid
    • A61K31/198Alpha-amino acids, e.g. alanine or edetic acid [EDTA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/38Heterocyclic compounds having sulfur as a ring hetero atom
    • A61K31/39Heterocyclic compounds having sulfur as a ring hetero atom having oxygen in the same ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/417Imidazole-alkylamines, e.g. histamine, phentolamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/475Quinolines; Isoquinolines having an indole ring, e.g. yohimbine, reserpine, strychnine, vinblastine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7076Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid
    • 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
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/191Tumor necrosis factors [TNF], e.g. lymphotoxin [LT], i.e. TNF-beta
    • 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
    • A61K38/2264Obesity-gene products, e.g. leptin
    • 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

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Obesity (AREA)
  • Emergency Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Child & Adolescent Psychology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Hematology (AREA)
  • Diabetes (AREA)
  • Genetics & Genomics (AREA)
  • Endocrinology (AREA)
  • Molecular Biology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

Materials and methods for modulating the transport of leptin across the blood brain barrier.

Description

MODULATION OF LEPTIN TRANSPORTATION THROUGH THE HEMATO-ENCEPHAL BARRIER RELATED REQUESTS This application claims priority of the US Provisional Patent Application Serial No. 60 / 150,300, filed on August 23, 1999. FIELD OF THE INVENTION The present invention it generally refers to the modulation of body weight and / or appetite of mammals, including humans. More particularly, this invention relates to compositions and methods that modulate the transport of weight-controlling molecules, such as for example leptin, through the blood-brain barrier. RIGHTS OF THE GOVERNMENT This invention was made with the support of the Government of the United States of America. The Government of the United States of America retains certain rights in this invention. BACKGROUND OF THE INVENTION Obesity is defined as an excess of body fat in relation to lean body mass and is related to important psychological and medical morbidity, including hypertension, elevated levels of blood lipids and diabetes. It is believed that body weight and energy balance are regulated by a feedback mechanism where regions of the brain, such as the hypothalamus, detect the amount of energy stored in the body and then adjust the food intake and the level of activity correspondingly [Brobeck, JR, Yale J. Biol. Med, 20: 545-552 (1948)]. Initial experiments have shown that the arterial transfer of blood from an animal having a hypothalamic lesion to a normal healthy animal results in the reduction of food intake by the normal animal [Hervey, GH, J. Physiol., 45: 336- 352 (1959)]. From these results, it was hypothesized that at least one component of the feedback mechanism circulated through the bloodstream and that the component had an action on the brain. It has been suggested that the OB gene may be responsible for the generation of this factor carried by the blood [see also Coleman, D.L., Diabetológica, 14: 141-148 (1978)]. Recent studies of the OB gene have confirmed that the OB gene product that is known as leptin is the factor carried by blood that works to maintain body weight and energy balance [Zhang et al., Nature., 372: 425-432 (1994); and Freidman et al., PCT Application No. PCT / US95 / 10479].
It has also been determined that the administration of leptin results in a lower amount of body fat [Pelley ounter, M.A. et al. Science, 269: 540-543 (1995); Halaas, et al. Science, 269: 546-549 (1995) .It is believed that leptin acts on the brain to inhibit food intake, regulate energy expenditure, and control body weight.In order for leptin to perform this type of function, function must cross the blood-brain barrier to penetrate the brain.The amount of leptin detected by the brain results from a combination of the permeability of the blood-brain barrier and the amount of leptin in the bloodstream which depends on its the level of stored energy or body fat of an individual [Considine, RV et al., N. Eng. J. Med. 334: 292-295 (1986)] Obesity can occur when the brain detects incorrectly a low level of leptin and therefore initiates mechanisms to raise this level by increasing the amount of body fat.This cycle usually continues until the brain detects an appropriate amount of leptin and at that time the body weight stop increasing. In accordance with what is described here, it is believed that increasing the efficiency of leptin transport through the blood-brain barrier could be an effective treatment for obesity, in most cases. Leptin carried by blood can penetrate the brain due to the presence of a specific saturable transporter that is located in the blood-brain barrier [Banks et al., Peptides 17 (2): 305-311 (1996)]. Since leptin is a large protein, leptin in the blood would be largely excluded from the brain in the absence of a transporter of this type. It is believed that the transporter is close to or contains within its structure some sites that, when activated, modify the transport regime of leptin. Such sites, conceptually analogous to binding sites with cofactors for enzymes or halosteric regulatory sites for receptors, provide therapeutic targets that can be manipulated to alter the transport regime of leptin from the blood to the brain in order to control the body weight. The mechanism of transport of proteins and peptides such as leptin through the blood-brain barrier is poorly understood. Certain proteins / certain peptides cross this barrier by diffusion directly through the endothelial / ependiral membranes according to their lipophilicity and / or molecular weight with smaller lipophilic molecules passing more freely [Banks & amp; amp;; Kastin, Psychoneuroendocrinology, 10: 385-399 (1985), Others are transported by saturable systems mediated by vehicles [Banks &Kastin, Pharmacol. Biochem. Behav, 21: 943-946 (1984)], as for example the system that Tyr-MIF transport (Tyr-Pro-Leu-Gly-amide) [Banks &Kastin, J. Pharmacol, Exp. Ther 239: 668-672 (1986)] Another mechanism through which proteins or «** -. polypeptides traverse the blood-brain barrier is through receptor-mediated permeabilization that is enabled by the administration of molecules such as bradykinin, leukotrienes, histamine, and 5-hydroxytryptamine [Unterberg, A., J. Cereb, Blood Flow Metab., 4: 574-585 (1984)]. Similarly, molecules such as leucine enkephalin, α-adrenergic, arachidonic acid, aluminum, forbomyristate esters, and α-thrombin increase the permeability of the blood-brain barrier while angiotensin II and β-adrenergics reduce permeability [Grieg, N., Physiology and Pharmacology of the Blood-Brain Barrier Handbook of Experimental Pharmacology, 103: 487-523 Springer-Verlag, Berlin (1992)]. There is a considerable need for molecules or compositions and methods for using these molecules and compositions that modulate (ie, increase or inhibit) the transport of weight-controlling molecules, such as leptin, through the blood-brain barrier. COMPENDIUM OF THE INVENTION The present invention focuses on methods and compositions for modulating the feeding and / or appetite behavior in mammals as well as for modulating body weight in mammals. More particularly, the present invention focuses on methods and compositions for modulating (increasing or inhibiting) the transport of leptin through the blood-brain barrier or through other blood / tissue barriers. The invention also focuses on methods and compositions for modulating (increasing or decreasing) body weight and / or metabolism by altering the transport of leptin through the blood-brain barrier. In accordance with the present invention, the transport of leptin through the blood-brain barrier can be increased, resulting in a reduction in body weight, and / or a decrease in appetite. Conversely, the transport of leptin through the blood-brain barrier can be reduced (or inhibited), which results in an increase in appetite and / or body weight in patients who require it (for example, anorexia, cachexia caused by advanced age, tumor-induced cachexia). The compositions can act on both sides of the blood-brain barrier (or other blood-tissue barriers) to result in an altered transport of leptin, even though preferred compositions and methods act on the blood side of the barrier. A preferred method of the invention comprises administration to a subject requiring such administration of a composition comprising an adrenergic agonist in an amount effective to increase the transport of leptin or variant of leptin, analogs, fragments, consensus leptin, or derivatives (including without limitation to this example a fusion protein, or chemically modified derivatives of leptin through the blood-brain barrier.) A fusion protein refers to a protein comprising a leptin polypeptide and a different protein. of the present invention allow the increase of transport of either endogenous leptin or exogenous leptin (including analogs, fragments, consensus leptin, chemical derivatives thereof or fusion proteins) through the blood-brain barrier. In reverse, in another embodiment of the invention, an adrenergic antagonist can be used to inhibit Go to transport leptin through the blood-brain barrier. An exemplary adrenergic antagonist which acts to inhibit the transport of leptin in the brain thereby resulting in an increase in body weight includes, without being limited to this example, benoxat, and may be administered to an individual to increase body weight. In accordance with the present invention, both purinergic and glutaminergic agonists can be employed to modulate the transport of leptin in the brain. An exemplary purinergic agonist comprises adenosine while an exemplary glutaminergic agonist comprises glutamate. Ways of administration of the compositions useful for the practice of the present invention include, not limited to, intravenous, intraarterial, intraperitoneal, intramuscular, intradermal, topical, intraocular, subcutaneous, intranasal, oral, intracisternal, intracerebroventricular, intrathecal, topical, intradermal or pulmonary. Conversely, in another preferred embodiment, the adrenergic antagonist can be used to inhibit the transport of leptin through the blood-brain barrier. In one embodiment of the present invention, the composition comprises one or more compounds selected from the group consisting of adrenergic agonists such as, but not limited to, epinephrine, isoproterenol, arterenol, cirazoline, phenylethylamine, epinephrine, norepinephrine, dopamine, nordefrin. , protoquilol, metaproterenol, metaraminol, phenylephrine, tyramine, hydroxyamfetamine, nilidrine, isoxsuprine, methoxyphenamine, methoxamine, amphetamine, methamphetamine, ephedrine, phenylpropanolamine, mefenter ina, chlorphentermine, tuaminoheptane, cyclopentane, propylhexedrine, and analogs and derivatives or metabolites thereof and optionally, a pharmaceutically acceptable carrier, excipient or diluent. Exemplary adrenergic antagonists include, but are not limited to, phentolamine, prazosin, benoxatin, phenothibenzamine, and related lal oalkylamines. In another embodiment, the compositions of the present invention comprise purinergic and glutaminergic agonists or ||| 8jÍMfe .._- g ^ _¿_ toafca-á B .. ______ * ._. * - .. , ... .. .. ... i. . T ^ -? . ? .__ .. ^ ____ < _ ^ "...-, • __ .., • -.__....., - _.__.._,. * ___. combinations thereof and their use in the methods of the invention. Active adenosine, the adenosine or purinergic receptor 1 (Pl). Pl receptors are widespread in the body including the cardiovascular system, the respiratory system, the immune system and the nervous system. Adenosine blocks opioid-induced feeding and caffeine is a PI antagonist. Glutamate is the endogenous ligand for glutamate receptors (glutaminergic). Glutamate receptors include ionotropic receptors (AMPA receptors, kainate, N-methyl-D-aspartate), which directly control ion channels, and metabotropic receptors that act through second-messenger systems. Glutamate receptors are the most common mediators of rapid excitatory synaptic transmission in the central nervous system. They participate in the mechanisms of memory and feeding. Other compounds that affect feeding, suppress appetite, induce anorexia, stimulate appetite, affect weight, or alter metabolism and may ultimately affect the transport of leptin through the blood-brain barrier and are useful in the practice of the present invention includes free fatty acids, sugars such as glucose, cytokines, drugs such as amphetamines, calcium channel blockers, monoamines, amino acids, hormones including steroid hormones, dietary supplements, ketones, starches, micronutrients, lipoproteins, prostaglandins, prostacyclines, peptides, proteins, regulators of nitric oxide production, agonists and antagonists of GABA and NMDA, vitamins, minerals and melatonin, and its precursors and metabolites. Preferred cytokines useful in the practice of the present invention include, without being limited to interleukin, interleukin-1, interleukin-1 receptor antagonist, interleukin-2, interleukin-6, interleukin-12, macrophage colony stimulation factor, inflammatory peptides of macrophages such as IP-la, MlP-lß and tumor necrosis factor a (TNFa). Other compounds used in the practice of the present invention include fenflurimine and related compounds. Other peptides and proteins useful for practicing the present invention either alone or in combination with other compounds described herein include, without being limited to, adrenocorticotropin hormone (ACTH), amylin, atrial natriuretic peptide (ANP), bombesin, calcitonin, peptide related to the calcitonin gene (CGRP), caerulein, transcript peptide regulated by cocaine and amphetamine (CART), colecistquinines (CCK), hormone release i < * Aii.jJ kJlálÍ & »A .. *. - * Mua. . .. ..._ t. ^ ,, ... ... ....._. ..., _............ i ^, ........ .... . i. ^ - ^. & thyroid corticotropin (CRH), Cyclo-His-Pro, enterostatin, FMRF-amide, galanin, glucagon, glucagon-like peptide (GLP), growth hormone, growth hormone releasing hormone (GHRH) ), gonadotropin hormone releasing hormone (GnRH or LHRH), insulin, insulin-like growth factors, macrophage migration inhibiting factor, melanocyte stimulating hormone (MSH), motilin, MSH inhibitor peptide (MIF-1) , nerve growth factor (NGF), neuromedins, neuropeptide Y (NPY), neurotensin, neurotrophins (NT-3, NT-4), opioid peptides (endorphins, enkephalins, endophors, dinoorphins, chiotorfins), orexin, oxytocin, pancreatic polypeptide, parathyroid hormone (PTH), polypeptides that activate pituitary adenylate cyclase (PACAP), sauvagine, somatostatin, substance P, thyroid stimulating hormone (TSH), thyrotropin releasing hormone (TRH), MIF-1 tyrosine, polypeptide intestinal glass active, and vasopressins. Other compositions used in the practice of the present invention comprise any of the above compositions in combination with another and / or in combination with one or more of the leptins described herein. This invention also focuses on a method for the treatment of obesity comprising the increase of leptin transport, leptin variants, analogues, . ..., - J, A .. To consensus leptins, fragments, or leptin derivatives through the blood-brain barrier according to any of the above aspects or modalities. In another embodiment, this invention provides methods and compositions for the treatment of metabolic disorders including obesity, diabetes mellitus, including type I and type II diabetes as well as insulin-resistant pathologies comprising increased leptin transport, variants of leptin, analogues, consensus leptins, fragments, or derivatives thereof through the blood-brain barrier according to any of the above aspects or modalities. Also within the scope of the present invention are pharmaceutical compositions useful for modulating body weight, the composition comprising leptin comprising the amino acid sequence presented in SEQ ID NO: 2 or 4, SEQ ID NO: 5 and SEQ ID NO: 6 , consensus leptins, variants, analogs, leptin fusion proteins, chemically modified leptin derivatives, and fragments thereof, and one or more agents selected from the group consisting of adrenergic agonists, adrenergic antagonists, neurotransmitters, peptide hormones , cytokines, amino acids, peptides or opiates, purinergic agonists, purinergic antagonists, glutaminergic agonists and glutaminergic antagonists, and metabolites thereof. The invention also includes compositions and methods for modulating body weight and / or treating metabolic disorders by modulating the regulatory pathways that control appetite and / or metabolism. Since leptin appears to play a controlling role in the regulation of appetite, the methods and compositions of the present invention are useful for modulating regulatory pathways where leptin plays a role, perhaps ultimately by regulating the transport of leptin through the blood-brain barrier. The invention also comprises the use of adrenergic agonists, adrenergic antagonists, neurotransmitters, peptide hormones, cytokines, amino acids, opioid peptides, purinergic agonists or purinergic antagonists, glutaminergic agonists or antagonists or metabolites thereof for the manufacture of a drug for modulating the transport of leptin in the brain and / or to modulate the body weight and / or to modulate the appetite in a mammal. The use may further comprise the use of any of the leptins within the scope of the present invention for the manufacture of the drug to modulate the transport of leptin through the blood-brain barrier and / or to modulate the body weight of a mammal. Preferred mammals for the practice of the present invention are humans.
DETAILED DESCRIPTION OF THE INVENTION The mammalian brain plays a central role in regulating the amount of fat in a mammal in part by regulating food intake, food selection, and thermogenesis. The brain detects the level of fat (adiposity) of the organism by detecting the amount of leptin in the blood of the organism that is transported in the brain through a specific saturable leptin transporter located in the blood-brain barrier. Obesity can occur when the brain incorrectly detects an amount less than the appropriate amount of leptin in the body thus activating mechanisms to increase adiposity (for example by increasing the diet, decreasing metabolism). The adiposity then rises until the brain detects an appropriate level of leptin. Therefore, increasing the efficiency of leptin transport through the blood-brain barrier would be an effective way to reduce adiposity by increasing the amount of leptin effectively. detected by the brain. The evidence suggests that the transporter responsible for the transport of leptin through the blood-brain barrier is related to sites that modify the transport regime of leptin when they are activated or contains those sites within their structure. These transporter regime modification sites are conceptually analogous to alesteric regulatory sites and / or co-factors for enzymes or co-factors. The presence of these 5 sites therefore provides attractive therapeutic targets that can be used to regulate the transport of leptin through the blood-brain barrier thereby regulating the adiposity in the mammal. The present invention offers compositions and methods for modulating body weight by modulating the signaling pathways involved in weight regulation and / or appetite regulation. The invention also offers compositions and methods for modulating leptin transport 15 through the blood-brain barrier and materials and methods to modulate the appetite. More particularly, the present invention focuses on compositions that include pharmaceutical compositions and methods for increasing or inhibiting the transport of • 20 leptin (OB) polypeptides through the blood-brain barrier. Such methods and compositions are useful for controlling the body weight of mammals, including humans. The methods and compositions are also useful in the treatment of metabolic disorders including diabetes 25 mellitus (type I and type II). The compositions and methods of bitAl »-.j» ¡m, "t .. t¡fcfc > The present invention exploits the central function of leptin in the regulation of appetite and metabolism by modulating the transport of leptin through the blood-brain barrier to a site of action in the brain. For the purposes of this invention, leptin and OB are used interchangeably and refer to a polypeptide having about 146 amino acids as a mature form. Any leptin molecule, including leptin variants, analogs, fragments, consensus leptins, or derivatives, which has the ability to modulate weight, or alter the metabolism in a host mammal, is useful in the practice of the present invention . Preferred leptin proteins useful in the practice of the present invention may be native murine leptin established as SEQ ID NO: 2 which includes its signal sequence, or its mature form starting at amino acid 21 (in accordance with what is numbered in SEQ ID NO: 2) of native leptin and presented as SEQ ID NO: 5 or protein according to that presented in Zhang et al. (Na tura, supra, which is incorporated herein by reference) either in the OB protein of a native human (SEQ ID NO: 4) or its mature sequence starting at amino acids 21 to 166 presented by SEQ ID NO: 6 (See Zhang et al., Nature supra, on page 428). Variants or analogs of leptin proteins useful in the practice of the present .. * ..__ *, ..._._ *. These include those that have a substitution of one or more of their amino acids with another while they are mowing while maintaining a biological activity of leptin. Natural variants of any leptin that does not have a glutamine residue at position 28 of the mature sequence or other natural variants are also useful in the practice of the present invention. Another example of a human leptin useful in the practice of the invention is an analog of SEQ ID NO: 6, comprising 1) an arginine in place of lysine at position 35; and 2) a leucine in place of isoleucine at position 74. (An abbreviation for this analogue is the R-> K35, L-> I74) recombinant human. Leptin molecules useful in the practice of the present invention may also optionally contain a methionine at the N-terminal (position-1). The murine leptin protein has significant homology to the human protein, particularly as a mature protein and, in addition, particularly at the N-terminus. A recombinant human protein analogue can be prepared for use in the practice of the present invention by the alteration (for example substitution of amino acid residues) in the recombinant human sequence, the amino acids that diverge from the murine sequence. For example, using a human protein having a lysine at residue 35 and an isoleucine at residue 74 according to the numbering of SEQ ID NO: 6, wherein the first amino acid is valine and the amino acid at position 146 is cysteine, it can be substituted with another amino acid or one or more of the amino acids at positions 32, 35, 50, 64, 68, 71, 74, 77, 89, 97, 100, 101, 105, 106, 107, 108, 111, 118, 136, 138, 142 and 145. The amino acid can be selected in the corresponding position of the murine protein, (SEQ ID NO: 6), or in another amino acid. It is also possible to prepare "consensus" molecules (consensus leptin or consensus OB) based on the rat OB protein sequence [Muraka i et al., Biochem. Biophys. Res. Com. 209: 944-952 (1995) which are incorporated herein by reference]. The rat OB protein differs from the human OB protein in the following positions (using the numbering of SEQ ID NO: 6): 4, 32, 33, 35, 50, 68, 71, 74, 77, 78, 89 , 97, 100, 101, 102, 105, 106, 107, 108, 111, 118, 136, 138, and 145. One or more of the amino acids in these divergent positions can be substituted with another amino acid. The underlined and bold positions are the positions in which the murine leptin protein as well as the rat OB protein diverge from the human OB protein, and therefore, are especially suitable for alteration. In one or more of these positions, one amino acid of the corresponding rat OB protein or another amino acid can be substituted.
The positions of the mature rat OB protein and mature murine protein that diverge from the mature human leptin protein are: 4, 32, 33, 35, 50, 64, 68, 71, 74, 77, 78, 89, 97, 100, 101, 102, 105, 106., 107, 108, 111, 118, 136, 138, 142, and 5 145. The human OB protein according to SEQ ID NO: 6 having one or more of the amino acids above removed or replaced with another amino acid, for example, the • amino acid found in the corresponding rat or murine sequence can also be effective. In addition, the amino acids found in rhesus monkey leptin protein that diverge from the mature human OB protein are (with identities indicated in parentheses in one letter amino acid abbreviation): 8 (S), 35 (R), 48 (V), 53 (Q), 60 (1), 66 (1), 67 (N), 68 (L), 89 (L), 100 (L), 108 (E), 15 112 (D), and 118 (L). Since the OB protein of recombinant human is active in cynomolgus monkeys, a human OB protein in accordance with SEQ ID NO: 4 or 6 having one or more of the rhesus monkey divergent amino acids replaced with another amino acid, by example the amino acids 20 in parentheses, it can be effective. It will be noted that certain divergent rhesus amino acids are also found in the aforementioned murine species (positions 35, 68, 108 and 118). Thus, a murine / rat / rhesus / human consensus molecule can be prepared (using the numbering of SEQ ID NO: 6 having one or more of the amino acids in the positions replaced by another amino acid: 4, 8, 32, 33, 35, 48, 50, 53, 60, 64, 66, 67, 68, 71, 74, 77, 78, 89, 97, 100, 101, 102, 105, 106, 107, 108, 111, 112, 118, 136, 138, 142, and 145. The positions underlined and in bold are the positions in the which the three species are divergent from the OB protein of a human being, and therefore are especially suitable for alteration. Other analogs can be prepared by removing a portion of the protein amino acid sequence that results in a fragment of a leptin polypeptide. For example, the mature protein does not have a leader sequence corresponding to amino acids 1-21 of SEQ ID NO: 4. The following truncated forms of the native human leptin protein molecules can be prepared (using the SEQ numbering). ID NO: 6): (a) amino acids 98-146 (b) amino acids 1-32 (c) amino acids 40-116 (d) amino acids 1-99 and (connected to) 112-146 (e) amino acids 1-99 and (connected to) 112-146 having one or more amino acids 100-111 placed between amino acids 99 and 112. Likewise, truncated forms (fragments) may also have altered one or more of the amino acids that are divergent (in the protein OB of murine, rat or rhesus) of the OB protein of being human. In addition, many alterations can take the form of altered amino acids, for example peptidomimetic or D-amino acids. In addition, leptin molecules having 83% or more of amino acid identity with leptins having the amino acid sequence presented in SEQ ID NOs: 2, 4, 5 or 6 can also be used in the practice of the present invention. Any of the aforementioned leptin molecules may optionally have an N-terminal methionine. Also included within the scope of the present invention are leptins encoded by any of the polynucleotides presented in US Patent No. 5,935,810 or any of the polypeptides presented herein. US Patent No. 6,001,968 which is incorporated by reference in its entirety. The present protein (herein the term "protein" is used to include "peptides" and OB analogues, such as those mentioned above, unless otherwise indicated) can also be derived by the attachment of one or more chemical portions on the portion of protein. The chemically modified derivatives can be further formulated for intraarterial, intraperitoneal, intramuscular, subcutaneous, intravenous, oral, nasal, pulmonary, topical, ocular, intracisternal, intrathecal, transdermal, intracerebral ventricular administration, or other routes of administration. The chemical modification of biologically active proteins provides additional advantages in certain circumstances, for example the increase in stability and circulation time of the therapeutic protein and the decrease in immunogenicity. See U.S. Patent No. 4,179,337, Davis et al., Issued December 18, 1979. For a review, see Abuchowski et al. Enzimes as Drugs (J.S. Holcenberg and J. Roberts, eds.pp. 367-383 (1981)). A review article describing modification of protein and fusion proteins is Francis, Focus on Growth Factors 3: 4-10 (May 1992) (published by Mediscript, Mountview Court, Friern Barnet Lane, London N20, OLD, UNITED KINGDOM) . The chemical portions suitable for derivatization can be selected from various water-soluble polymers. The selected polymer must be soluble in water in such a way that the protein to which it is bound does not precipitate in an aqueous environment, for example a physiological environment. Preferably, for therapeutic use of the final product preparation, the polymer will be pharmaceutically acceptable. One skilled in the art will be able to select the desired polymer based on considerations such as whether the polymer / protein conjugate will be used therapeutically and, if appropriate, the desired dosage, circulation time, resistance to proteolysis, and other considerations. For the proteins and peptides present, the effectiveness of the derivation can be determined by administration of the derivative, in the desired form (ie, by osmotic pump, or, more preferably, by injection or infusion or, can also be formulated for oral, pulmonary or nasal administration, for example), and by observing biological effects in accordance with what is described herein. The water-soluble polymer can be selected from the group consisting of, for example, polyethylene glycol, ethylene glycol / propylene glycol / carboxymethyl cellulose copolymers, dextran, polyvinyl alcohol, polyvinyl pyrrolidone, poly-1,3-dioxolane, poly-1,3,6. -trioxane, ethylene / maleic anhydride copolymer, polyamino acids (either homopolymers or random or non-random copolymers), and dextran or poly (n-vinylpyrrolidone) polyethylene oxide / ethylene oxide copolymers, polyoxyethylated polyols, polystyrene maleate and polyvinyl alcohol. Polyethylene glycol propionaldehyde may have advantages in manufacturing due to its stability in water. Fusion proteins can be prepared by fixing polyamino acids on an OB (or analogous) protein portion. For example, the polyamino acid can be a carrier protein that serves to increase the half-life of protein circulation. For the therapeutic or cosmetic purposes present, said polyamino acid must be a polyamino acid that does not elicit an antibody neutralization response, or other adverse response. Said polyamino acid may be selected from the group consisting of serum albumin (e.g., human serum albumin), an antibody or portion thereof (e.g. • constant antibody region, sometimes referred to as "Fc") or other polyamino acids. Don compliance with 10 indicated below, the polyamino acid binding location can be found at the N-terminus of the OB protein portion, or elsewhere, and can also be connected by a chemical "linker" portion to the OB protein. In the case of an OB-Fc fusion, the OB is typically fused at its C-terminal to the N-terminal. However, the OB can be fused at its N-terminal to the C-terminus of the Fc-molecule. Typically, in such fusions, the fused protein will retain at least the domains • 20 fusionally active CH2 and CH3 hinges of the constant region of the immunoglobulin heavy chain. The fusions may also be carried out at the C terminal of the Fc portion of a constant or immediately N-terminal domain relative to the CH1 domain of the heavy chain or 25 well the corresponding region of the light chain. The place Exactly in which the fusion takes place is not critical. The fusion proteins can comprise multimers of the Fc-OB fusion. The polymer can be of any molecular weight and can be branched or unbranched. In the case of polyethylene glycol, the preferred molecular weight is between 2 kDa and about 100 kDa (the term "about" indicates that in polyethylene glycol preparations, certain molecules will have a greater weight, or after molecules in less weight, than the established molecular weight. ) to facilitate handling and manufacturing. Other sizes can be used, depending on the desired therapeutic profile (for example, the duration of the desired prolonged release, the effects, possibly on the biological activity, the ease of handling, the degree of antigenicity or the lack of antigenicity and other known effects. of polyethylene glycol on a protein or analog). The number of polymer molecules fixed in this way can vary and a person skilled in the art can determine the effect on the function. It can be mono derivatized or a di-, tri-, tetra-, or another derivation combination can be provided with the same chemical portions or with different chemical portions (for example, polymers, for example different weights of polyethylene glycols) . The ratio between polymer molecules and molecules of protein (or peptide) will vary, as well as their concentrations in the reaction mixture. In general, the optimum ratio (in terms of reaction efficiency where there is no excess of unreacted protein or polymer) will be determined by factors such as the desired degree of derivation (eg, mono, di-, tri-, etc.), the molecular weight of the selected polymer, where the polymer is branched or unbranched, and the reaction conditions. The chemical portions must be bound to the protein taking into account the effects on the functional or antigenic domains of the protein. There are numerous fixation methods available to the person skilled in the art. For example EP 401 384 which is incorporated herein by reference (coupling of PEG on G-CSF), see also Malik, Exp. Hematol. 20: 1028-1035 (1992) (reported pegylation of GM-CSF using tresyl chloride). For example, polyethylene glycol can be covalently linked through amino acid residues through a reactive group, for example a free amino or carboxyl group. Reactive groups are groups in which an activated polyethylene glycol molecule can be attached. The amino acid residues having a free amino group can include licina residues and the N-terminal amino acid residue. Those having a free carboxyl group can include, aspartic acid residues, glutamic acid residues and C-terminal amino acid residues.
Sulfhydryl groups can also be used as reactive groups to hold polyethylene glycol molecules. For therapeutic purposes, a binding to an amino group is preferred, for example an N-terminal or lysine-binding. Fixation on residues important for binding with the receiver should be avoided if a connection with the receiver is desired. N-terminally chemically modified OB proteins or polypeptides can be specifically desired. Using polyethylene glycol as an illustration of the compositions useful in the practice of the present invention, one can select from among several molecules of polyethylene glycol (by molecular weight, branching, etc.), the ratio between polyethylene glycol molecules and protein molecules in the mixture of polyethylene glycol. the reaction, the type of pegylation reaction to be performed, and the method for obtaining the selected N-terminally pegylated protein. The method for obtaining the N-terminally pegylated preparation (ie, the separation of this portion from other monopegylated proportions, if necessary) can be by purification of the N-terminally pegylated material from a population of pegylated protein molecules. Selective N-terminal chemical modification can be achieved by reductive alkylation that exploits the differential reactivation of different types of primary amino groups (lysine versus ...., .--. * -i ... t i..is? .í • «IcttllaBai-. .... ^ -? A., I ....... *. »__ .." -. ........
N-terminal) available for derivation in the particular protein. Under the appropriate reaction conditions, a substantially selective derivation of the N-terminal protein with a polymer-containing carbonyl group is achieved. For example, the protein can be N-terminally pegylated selectively by effecting the reaction at a pH which allows it to take advantage of the pKa differences between the e-amino group of the lysine residues and the a-amino group of the N-terminal residue of the protein. Through a selective derivation of this type, the binding of a water soluble polymer on a protein is controlled: the conjugation with the polymer is carried out predominantly at the N-terminus of the protein and no significant modification of other reactive groups occurs, for example in the amino groups of side chain of lysine using the reductive alkylation, the water-soluble polymer may be of the type described above, and must have simple aldehyde for coupling with the protein. Polyethylene glycol propionaldehyde containing a simple reactive aldehyde can be employed. An N-terminally monopegylated derivative is preferred to facilitate the production of a therapeutic agent, an N-terminal pegylation ensures a homogeneous product as product characterization is simplified in relation to di-, tri- or multi-pegylated products. The use of the above reductive alkylation process for the preparation of an N-terminal product is preferred by the ease of commercial manufacture. As described in the following Examples, the administration of compositions that interact with an adrenoreceptor (preferably adrenergic agonists) either before or concurrently with the administration of a leptin significantly increases the amount of leptin that crosses the blood-brain barrier in the brain. The compositions and methods of the invention are also useful for increasing the transport of endogenous leptin through the blood-brain barrier. These results are illustrated through the following Examples where a labeled radio leptin was administered and then measured in mice that received compositions that interact with an adrenoreceptor. Compositions containing epinephrine (which works with adrenoreceptor) were the most effective to increase leptin transport. Other compositions including those containing amino acids or hormones were also tested and in some cases were effective in increasing the transport of leptin through the blood-brain barrier. The invention is described through the following Examples by way of illustration and said Examples are not to be construed as limiting the invention set forth in the appended claims.
Example 1 describes the effects of epinephrine on the transport of leptin through the blood-brain barrier. Example 2 describes the effect of various dosages of epinephrine on the transport of leptin through the blood-brain barrier. In Example 3, the effect of epinephrine on the integrity of the blood-brain barrier is examined. In Example 4, the effect of the amino acids tyrosine and phenylalanine on the transport of leptin through the blood-brain barrier is studied. In Example 5, the effect of arginine, phenylalanine, tryptophan, and tyrosine on the transport of leptin through the blood-brain barrier is studied. In Example 6, the effects of neurotransmitters including dopamine, histamine, serotonin and epinephrine on the transport of leptin are described. Example 7 describes the defect of coadministration of adrenoreceptor agonists / antagonists of cirazoline hydrochloride, UK14304, albuterol, CGP-12177A, and benoxatine hydrochloride on the transport of leptin through the blood-brain barrier. In Example 8, the effect of the co-administration of certain adrenoreceptor agonists such as isoproterrenol, clonidine, arterenol, and phenylephrine on the l? ak _¿_ab_ * .. *. A ^ &amp ... ~. 4 »t. • *. * fc% & .-t.f i transport of leptin through the blood-brain barrier. In Example 9 the effect of the adrenoreceptor antagonists phentolamine, D, L-propanolol, yohimbine, and prasozine on the transport of leptin through the blood-brain barrier is tested. Example 10 describes the effect of tumor necrosis factor on the transport of leptin through the blood-brain barrier. Example 11 describes the effect of purinergic and glutaminergic agonists on the transport of leptin through the blood-brain barrier. Example 1 Effects of the Administration of Epinephrine on the Transport of Leptin through the Hemato-encephalic barrier in Mice. In this Example, the effect of the administration of epinephrine on the transport of leptin through the blood-brain barrier in mice is studied. In these Experiments, six groups of five male ICR mice (Blue Spruce Farms, Altamont, NY) weighing approximately 17-22 g were anesthetized with ethyl carbamate (4 g / kg) and then their jugular vein was surgically exposed. carotid artery. Mice received an intraperitoneal (i.p.) injection of epinephrine (33 μg / 200 μl) in a Ringer solution lactated with 1% bovine serum albumin. The hour of these injections was considered as zero hour. After time intervals of 10 minutes (min), 30 min., 45 min., 1 hour (h), and 2 h after epinephrine injection, radiolabeled leptin (125 I, 1.65 x 10 d) was administered in solution of Ringer lactate with bovine serum albumin at 1% mice through intravenous (iv) injection into the jugular vein. The control mice did not receive epinephrine only a Ringer solution laced with 1% bovine serum albumin and received an injection of 125 I-leptin only after the 10 minute time interval. All the mice were decapitated and their blood was collected after 10 minutes after the injection of leptin. The brain (except pineal and pituitary bodies) was removed and counted in gamma counter (Micromedic 4/200, Horsham, PA) for 3 minutes. Blood was collected from a cut in the right carotid artery, centrifuged at 2000 g for 10 minutes at 4 ° C, and then 0.1 ml was counted in a gamma counter. The brain / blood ratios were expressed as algebraic counts at μl / g of brain over counts / min. / μl of arterial blood. Table 1. Administration of Epinephrine followed by Leptin Time after 125I-leptin, brain / serum ± Administration of standard deviation (cpg / g) SA to * ^ *, jJhü - «, ^ -é * ¿- ^. j a.fc », -. ^^ "« * _ ..; »-á. *: £ *" * ^ tjt,. ^ ¿Temj & »A, * -. *! Áha &Uk w & Epinephrine (cpm / μl) Control (10 min., Without 15.68 ± 2.28 Epinephrine) 10 min. 23.69 ± 3.90 30 min. 23.27 ± 3.45 45 min. 18.68 + 2.19 1 h 20.73 ± 2.68 2 h 24.86 ± 4.02 The results of this experiment indicate that the administration of epinephrine before the administration of leptin increases the absorption of leptin by the brain in mice. More specifically, in the control mice that did not receive epinephrine, the amount of radiolabelled leptin in the brain after i.v. it was 15.68 ± 2.28 count / min. / g of brain over counts / min. / μl of arterial blood compared to the amount of leptin in the mice that received epinephrine before leptin administration was 23.69 + 3.90 counts / min. / g of brain over count / min. / μl of arterial blood. This represents an increase in the absorption of leptin by the brain of approximately 51%. The other time points of 30 min., 45 min., 1 h, and 2 h illustrate that epinephrine continues to exert its positive effects on the absorption of leptin by the brain even after the passage of a time interval of 2 h.
Example 2 Effects of the Administration of Epinephrine on the Barrier Hematoencephalic in Mice In this Example, the effect of several dosages of epinephrine on the transport of leptin through the blood-brain barrier was studied. As in Example 1, the mice were anesthetized with ethyl carbamate. The mice received an i.v. of a solution containing radiolabelled leptin (1 51.2 lxlO6 cpm) in lactated Ringer's solution with 1% bovine serum albumin and various amounts of epinephrine (133.33 μg, 400 nM, 66.6 μg, 200 nM, 33.3 μg, 100 nM; 13.3 μg, 40 nM, 0.667 μg, 2 nM) in 200 μl. Blood and brain samples were collected according to the previous Examples 10 minutes after the injection of leptin. Table 2. Co-administration of Epinephrine with Leptin Time after 125I, brain / serum ± Administration of standard deviation (cpg / g) Epinephrine (10 min.) (Cpm / μl) Control (no epinephrine) 24.54 + 5.6 + epinephrine 2 nM 28.68 ± 10.7 + epinephrine 40 nM 62.73 + 27.9 + epinephrine 100 nM 74.18 + 20.3 + epinephrine 200 nM 64.75 ± 21.3 + epinephrine 400 nM all mice died Instantly The results of this experiment indicate that the co-administration of leptin plus epinephrine increases the absorption of leptin by the brain. In the same way, at 40 nM epinephrine, the absorption was increased by approximately 155%, to 100 nM epinephrine the absorption was increased by approximately 200%, to 200 nM epinephrine, the absorption was increased by approximately 163% but two of the five mice in this group died, and 400 nM epinephrine 10 all mice died. These data were corrected for the amount of residual blood in the brain after the removal of capillaries by gradient centrifugation. Example 3 Effects of the Administration of Epinephrine on Integrity 15 of the Hematoencephalic Barrier in Mice In this Example, the effects of the administration of epinephrine on the integrity of the blood-brain barrier in mice were evaluated. Radiolabelled albumin is the traditional standard to administer and 20 monitor for the purpose of testing the integrity of the blood-brain barrier (Davson, H. (1967) Physi olgy of the Cerebrospinal Fl uid, pp. 82-103, J &A. Churchill, London). In accordance with what was described above, mice were 25 anesthetized with ethyl carbamate and then received a i.v. injection either of a solution containing radiolabelled leptin (125I, 1.54xl06 cpm) and albumin (99Tc, 3.4x06 cpm) (labeled solution) in Ringer's solution lactated with 1% bovine serum albumin in 200 μl or the labeled solution plus epinephrine (33 μg). All the mice were decapitated and the blood and testes were collected after 10 minutes after the injection of leptin. The brain (except pituitary and pineal) was removed and counted in a gamma counter (Micromedic 4/200, Horsham, PA) for 3 minutes. The blood was also collected from a cut in the right carotid artery, centrifuged for 2000 g for 10 minutes, at a temperature of 4 ° C, and then 50 μl was counted in a gamma counter. The brain / blood and testes / blood ratios were expressed as counts / min. / g of brain or testicle on counts / min. / μl of arterial blood. Table 3. Co-administration of Epinephrine with Leptin and Albumin Time brain / brain / testicles / testicle: Blood blood blood back to ad- (cpm / g) (cpm / g) (cpm / g) (cpm / g) ministra- (cpm / μl) (cpm / μl) (cpm / μl) (cpm / μl) treatment of epinephrine Control (without 125I-lep- "Tc-al- 125I-lep-" Tc-albumin-Epinephrine) tub tina mine 1 min. 12.57 10.02 8.03 4.20 2 min. 15.66 10.22 7.71 4.52 3 min. 16.80 9.87 10.64 4.45 4 min. 22.18 10.14 23.76 8.23 5 min. 22.84 10.85 23.92 8.85 7.5 min. 23.46 10.38 29.55 9.06 10 min. 27.48 11.60 39.02 10.74 12.5 min. 24.36 10.47 44.36 14.42 + 40 nM epinephrine 1 min. 19.50 10.38 5.37 2.90 2 min. 22.11 11.28 9.55 4.70 3 min. 27.51 10.84 9.98 4.09 4 min. 36.82 10.46 18.42 6.56 5 min. 28.97 10.62 29.72 7.78 7.5 min. 36.55 10.27 58.68 17.52 10 min. 38.38 11.82 25.38 8.42 12.5 min. 50.53 10.52 152.38 34.09 The results of this experiment indicate that the co-administration of epinephrine with leptin induced an increased absorption of leptin by the brain in mice. The data of these time points of 1 min., 2 min., 3 min., 4 min., 5 min., 7.5 min., 10 min., And 12 min. They illustrate that the effects of epinephrine on the absorption of leptin by the brain increase over time as shown in this experiment iJ, 12 minutes. The co-administration of epinephrine did not increase the albumin absorption in the brain of mice. This shows that the increased absorption of leptin by the brain when epinephrine is administered is not the result of a damaged blood-brain barrier, since the amount of 99Tc-albumin that crosses the blood-brain barrier remains almost the same in the presence or absence of Epinephrine In other studies, it was shown that epinephrine increased the absorption of leptin and albumin by the testes. While these data indicate that epinephrine can act by disrupting the blood-testicle barrier, they provide nevertheless evidence that the absorption of leptin into other tissues that the brain can be increased by using cytokines, peptides, neurotransmitters and other molecules in accordance with the present invention. EXAMPLE 4 EFFECTS OF Amino Acid Administration on the Transport of Leptin Through the Hematoencephalic Barrier in Mice In this Example, the effect of the administration of several amino acids on the transport of leptin through the hematocrit barrier was studied. encephalic As in the previous Examples, the mice were anesthetized with ethyl carbamate. The mice then received an i.v. either of a marked solution that .J < tí.iÉ-_t-A »« M * - «- Ma AuJ_, fc. l. «rt .. . i -. . . . * - < m * .f- * - > -,. %. -. ,. _. ?*TO.?* . k-contained radiolabelled human leptin (1251.2.lxl O6 cpm) in Ringer's solution lactated with 1% bovine serum albumin in 200 μl or the labeled solution plus one of the following amino acids (tyrosine or phenylalanine, 10 μg). Blood and brain samples were collected according to what was described in the previous Examples at the following time points after the injection of leptin (1 min., 2 min., 3 min., 4 min., 5 mm., 7.5 min., 10 min., 12.5 min., and 15 min.) Table 4. Co-administration of Amino Acids with Leptin Time after the brain / blood (cpm / g) (cpm / μl; control amino acid administration (without amino - 12b I-leptin acids 1 min 12.98 2 min 16.34 3 min 15.64 4 min 16.71 5 min 18.38 7.5 min 17.42 10 min 19.00 12.5 min 22.83 15 min 26.33 + tyrosine ... > .,. & »... _- ». Á- r < c "¿.. ..?? staUt" ", ...- • -jas? -« á-saj ji. í 1 min 13.34 2 min 14.67 3 min 15.04 4 min 16.35 5 min 17.55 7.5 min 18.26 10 min 22.90 12.5 min 24.11 15 min 23.86 + phenylalanine 1 min 12.24 4 min 16.15 5 min 14.39 10 min 16.67 1122.55 mmiinn .. 20.64 15 min 23.55 The results of this experiment indicate that the co-administration of tyrosine with leptin increases the absorption of leptin by the brain, however, phenylalanine has no such effect.The increase in leptin uptake by tyrosine depended on the time in the tested interval of 15 minutes. EXAMPLE 5 EFFECTS OF THE ADMINISTRATION OF OTHER Amino Acids on the Transport of Leptin through the Hematogenous Barrier i. to. _ < . t- .. i ...,. £ ^% ^^ -L encephalic in Mice In this Example, the effect of other amino acids on the transport of leptin through the ato-encephalic barrier was studied. As in the previous Examples, the mice were anesthetized with ethyl carbamate. The mice received an i.v. either of a solution containing radiolabelled leptin (125I, 1.68xl06 cpm) in Ringer's solution lactated with 1% bovine serum albumin in 200 μl of the solution plus one of the following amino acids (arginine, phenylalanine, tryptophan, or tyrosine, 1 mg). Blood and brain samples were collected according to what was described in the previous Examples 10 minutes after the injection of leptin. Table 5. Co-administration of Amino Acids with Leptin Time after 125 I-leptin, brain / blood Administration of standard leprosy (cpm / g) Tub (10 min.) / (Cpm / μl) Control (without epinephrine) 15.30 ± 5.05 + arginine 18.82 + 2.58 + phenylalanine 19.83 + 5.29 + tryptophan 17.97 + 4.33 + tyrosine 23.73 ± 8.84 The results of this experiment indicate that the co-administration of arginine, phenylalanine, or tryptophan with Leptin does not affect the absorption of leptin by the brain. However, administration of tyrosine significantly increases the absorption of leptin by the brain. Similar studies also show neither leucine, nor threonine, nor glycine have an effect on the transport of leptin through the blood-brain barrier. Example 6 Effects of Neurotransmitter Administration on the transport of Leptin through the Hematoencephalic Barrier in Mice In this Example, the effect of certain neurotransmitters on the transport of leptin through the blood-brain barrier is studied. As described above, groups of mice were anesthetized with ethyl carbamate. The mice then received an intracerebroventricular (icv) injection of a solution containing a neurotransmitter, for example: acetylcholine, 98 μg; dopamine, 103 μg, epinephrine, 55 μg, histamine, 117 μg; or serotonin, 130 μg. After 10 minutes the mice received an i.v. containing radiolabelled leptin (125I, 1.77x106 cpm) in a Ringer solution laced with 1% bovine serum albumin in 100 μl. Blood and brain samples were collected according to what was described in the previous Examples 10 minutes after the injection of leptin. _- «.__ Table 6. Administration of Neurotransmitters with Leptin Time after 125I-leptin, brain / blood Administration of standard leprosy (cpm / g) Tub (10 min.) / (Cpm / μl) 5 Control (without epinephrine, 12.95 ± 3.31 + acetylcholine 13.78 ± 4.73 + dopamine 13.17 ± 2.94 • + epinephrine 15.37 + 3.12 + histamine 10.85 ± 4.22 10 + serotonin 11.32 ± 3.38 The results of this experiment indicate that the ivc administration of acetylcholine neurotransmitters, # Dopamine, histamine, and serotonin with leptin, did not increase the absorption of leptin by the brain. This shows that the The site in which epinephrine acts to modify the transport of leptin is found on the blood side of the blood-brain barrier. In another series of studies, neurotransmitters were injected intravenously with 200 nmol / mouse either from 20 acetylcholine, dopamine, epinephrine, histamine or serotonin. The study results indicate that only epinephrine could increase the transport of leptin through the blood-brain barrier. Example 7 25 Effects of Adrenoreceptor Agonist Administration .. * < _ .. *. ufe. < ¿Afaé .t * & * on the Transport of Leptin through the Hematoencephalic Barrier in Mice In this Example, the effects of certain adrenoreceptor agonists on the transport of leptin through the blood-brain barrier are studied. As in the previous Examples, mice were anesthetized with ethyl carbamate. The mice received an injection • encephalic As in the previous Examples, the mice were 10 anesthetized with ethyl carbamate. The mice received an i.v. either of a labeled solution containing radiolabeled leptin (125I, 1.98xl06 cpm) in solution • Lactated Ringer with 1% bovine serum albumin in 100 μl of the solution plus one of the following agonists: 15 isoproterenol, 25.30 μg; clonidine, 18.66 μg; Epinephrine, 14.26 μg; L-phenylephrine, 14.26 μg; or arterenol 22.35 μg). Blood and brain samples were collected according to what was described above 10 minutes after the injection of leptin. 20 Table 7. Co-administration of Adrenoreceptor Agonists with Leptin. Time after 125I-leptin, brain / blood Administration ± standard deviation (cpm / g) (10 min.) / (Cpm / μl) 25 control (without agonist) 14.07 ± 1.88 + isoproterenol 20.61 ± 4.05 + clonidine 15.05 ± 1.54 + arterenol (norepinephrine) 23.78 ± 6.35 + L-phenylephrine 19.19 ± 4.57 5 The results of this experiment indicate that the coadministration of adrenoreceptor agonists, isoproterenol and arterenol with leptin increased the absorption of Leptin through the brain. However, clonidine and L-phenylephrine had no effect on the transport of 10 leptin. Example 8 Effects of Administration of Adrenergic Antagonists on the Transport of Leptin Through the Hematoencephalic Barrier in Mice In this Example, the effects of coadministration of epinephrine with adrenoreceptor antagonists on the transport of leptin through of the hemato-encephalic barrier. In accordance with what was described above, the mice were • 20 anesthetized with ethyl carbamate. The mice then received an i.v. either of a labeled solution containing radiolabelled leptin (125I, 1.48xl06 cpm) in Ringer's solution lactated with 1% bovine serum albumin and epinephrine (3.33 μl) in 100 μl or labeled leptin or 25 epinephrine plus one of the following antagonists ¿Si3 8 ^ * FÁ **? Fc¿t ** & ™ * í- & ~ * ', - =. ^ • L «. * -. - - * -,! '.. * • .. - - -,.__, _- to. -. . . .. uaaa JK * t i *. (phentolamine, 528.4 μg, D, L-propanolol, 4.41 μg, yohimbine, 547.3 μg, or prasozin, saturated solution of 587.8 μg). Blood and brain samples were collected according to what was described above 10 minutes after the injection of leptin. Table 8. Co-administration of Adrenoreceptor Antagonists and Epinephrine with Leptin Time after 125I-leptin, brain / blood Standard epinephrine administration (cpm / g) nephrin (10 min.) / (Cpm / μl) control (epinephri26.78 + 4.62 na alone) + phentolamine 18. 86 ± 4. 27 + D, L-propanolol 25. 68 ± 4. 02 + yohimbine 17. 38 ± 3. 30 + prasozina 18. 06 ± 1. 89 The results of this experiment indicate that the coadministration of the adrenoreceptor antagonists plus epinephrine either had no effect on the absorption of leptin by the brain or reduced the absorption of leptin by the brain. Specifically, D, L-propanolol (an ß antagonist) had no effect while phentolamine, yohimbine and prasozine (to antagonists) had a negative effect compared to the control. Example 9 -...? Jm Effects of the Administration of Agonists / Adrenergic Antagonists on the Transport of Leptin through the Hematoencephalic Barrier in Mice In this Example, the effect of agonists and antagonists on the transport of leptin through the blood-brain barrier was studied . As in the previous Examples, mice were anesthetized with ethyl carbamate. The mice then received an i.v. either of a solution containing radiolabelled leptin (125 I, 1.2xlOd cpm) in Ringer's solution with 1% bovine serum albumin in 100 μl or the labeled solution plus one of the following agonists (cirazoline hydrochloride, 25 μg; albuterol , 50 μg, UK 14304, epinephrine, 13.3 μg) or antagonists (benoxatine hydrochloride, 250 μg + epinephrine, 13.3 μg, CGP-12177A (250 μg) and epinephrine, 13.3 μg). Blood and brain samples were collected according to what was described in the previous Examples 10 minutes after the epinephrine injection. Table 9. Co-administration of Agonists / Antagonists of i, a2, ß adrenoreceptors with leptin. Time after 125 I-leptin, brain / blood Standard epinephrine administration (cpm / g) nephrin (10 min.) / (Cpm / μl) control (without epinephrine) 17.0 ± 2.74 + DMSO 25.1 ± 2.13 + epinephrine 47.1 ± 17.3 + cirazoline (ai agonist) 45.5 + 11.3 + DMSO + UK14304 (a2 agonist) 14.7 + 3.98 + albuterol (ß agonist) 18.4 ± 2.39 5 + epinephrine + CGP12177A 39.7 ± 8.70 (ß antagonist) + epinephrine + benoxatin 19.7 + 5.01 • (antagonist) The results of the experiment indicate that the administration of ai-agonist cirazoline with leptin increased the absorption of leptin by the brain. The data also show that the benoxatine antagonist blocked the • effect of epinephrine increase. Example 10 15 Modulation of Leptin Transport by Tumor Necrosis Factor a (TNF-a) TNF-a (cachexin) is a cytokine that is approximately the same size as leptin that is transported through the blood-brain barrier and that It also has effects 20 about feeding. This suggests the possibility that TNF can modulate the transport of leptin through the blood-brain barrier. Similarly, leptin may play a role in the transport of TNF through the blood-brain barrier. This hypothesis was tested 25 using two experimental paradigms, an acute model and a _aa_._ chronic model. Acute Model This experiment determined whether the acute administration of TNF could noticeably affect the entry of radiolabeled leptin 5 (I-leptin) into the brain. Three groups of mice were tested. One group received labeled leptin (in accordance with that described in Example • presented above) alone; a second group received labeled leptin plus 1 μl / mouse of mouse TNF; a third group 10 received one μl / mouse of human TNF and labeled leptin. After the injection, brain and blood samples were obtained at times similar to those used in the Examples • presented above in time. (It will be noted that it is a high dose of TNF). No differences were observed between these 15 groups as to the amount of labeled leptin transported in the brain of the mice. These data show that TNF and leptin do not share the same transport (there is no competition or inhibition of transport of leptin) nor regulates in an important way the transporter of 20 leptin (no increase in leptin transporter). Chronic Model This experiment was carried out in genetically altered mice in such a way that both receptors for TNF were "knocked out" and therefore did not express receptors of 25 active TNF. As such, these mice were insensitive to ÜS-É, 2 ». & __É_¿Í «_La y .. _., ..-_ _-S-É_6 -? _. ^. > . * i. -_... sik___. _. . S.ni * _.- *. .. ~. ».. ** &&. > .
TNF. The rate of leptin-labeled absorption in these mice was then determined in comparison to the controls. The amount of unlabeled leptin that was necessary to inhibit the leptin transporter in these mice was also determined. The input rate of 0.477 μl / g-min for labeled leptin is similar to that typically found in normal mice. However, the entry of I-leptin into the brain was not inhibited by 0.1 or 0.3 μl / mouse of unlabeled leptin and 1.0 μl / mouse inhibited the entry regimen by only 40%. In normal mice 0.3 μl inhibits entry by 50% and one μg / mouse inhibits entry by 95%. These data suggest that mice knocked out for TNF receptor have an altered transporter for leptin through the blood-brain barrier. Chronic exposure to TNF, perhaps especially during development, is probably required for the normal functioning of the leptin transporter. This therefore represents another class of compounds (cytokines) in addition to the adrenergic agonist, amino acids and other compositions described above that can modulate the transport of leptin through the blood-brain barrier. Example 11 Modulation of Leptin Absorption by Adenosine and Glutamate t.i__t. .-í, .Ít. i. j .i ..
Studies were conducted to determine if compounds that interact with urinary receptors or with glutamate receptors (glutaminergics) are able to regulate the absorption of leptin in the brain. In particular, these studies were conducted to determine whether acute administration of adenosine, arginine or glutamate could affect the entry of radiolabeled leptin (I-leptin) into the brain. Four groups of mice were tested. One group received labeled leptin (as described in the Examples presented above) alone, a second group received marked leptin plus adenosine (0.4 mmol / kg), a third group received labeled leptin plus arginine (10 mg / mouse) and a fourth group received marked leptin plus glutamate in the 10 mg / mouse dosage. L-arginine is an essential amino acid included below as a control. After the injection, brain and blood samples were obtained 10 minutes after the injection of leptin in accordance with what was described. The brain / serum rations have been corrected for vascular space subtracting 10 μl / g. Table 10. Co-administration of Adenosine, Arginine, Glutamate with Leptin Compound 125I-leptin, brain / blood ± standard deviation (cpm / g) (cpm / μl) -Í..Í.; > ..JI- A.5 I-leptin alone (control) 21.6 ± 1.96 Adenosine 14.9 ± 4.79 Arginine 21.8 ± 5.29 Glutamate 9.67 ± 2.3 The results of this experiment shown in Table 10 indicate that the co-administration of purinergic adenosine agonist or the glutamate agonist significantly decreased the transport of leptin to the brain. The co-administration of arginines, an essential amino acid, had no effect on the transport of leptin. The results presented above show that purinergic agonists such as adenosine are useful for decreasing the absorption of leptin in the brain and can therefore act as therapeutic agents under pathological conditions or else under other circumstances where a decrease in the absorption of leptin in the brain it is desired Similarly, the results shown in Table 10 show that agonists (e.g., glutamate) that interact with glutamate receptors including ionotropic receptors (e.g., AMPA and N-methyl-D-aspartate receptors) and metrabotropic receptors can also be useful in the same context as the purinergic agonists described above. While the present invention has been described in terms of aS ?. - * _._. _i..É_i .._ t¿. fca-t .. specific modalities, it is understood that experts in the field may implement variations and modifications. All references are incorporated herein by reference. • 10 fifteen twenty 25 _ .. ».. -..-- r .--,, _i .., > fc.,: Aüti i-i., LIST OF SEQUENCES < 110 > Banks, William A. < 120 > MODULATION OF TRANSPORTATION OF LEPTINE THROUGH THE BARRIER HEMATO-ENCEFÁLICA < 130 > 01017/35040 < 140 > < 141 > < 160 > 6 < 170 > Patentln Ver. 2.0 < 210 > 1 < 211 > 2793 < 212 > DNA < 213 > Murino < 220 > < 223 > CDNA of murine ob (leptin) < 220 > < 221 > CDS < 222 > (57) .. (557) < 220 > < 221 > sig_peptide < 222 > (57) .. (59) < 220 > < 221 > mat_peptide < 222 > (60) .. (557) < 400 > 1 ggatccctgc tccagcagct gcaaggtgca agaagaagaa gatcccaggg aggaaa atg 59 Met -1 tgc tgg aga ccc ctg tgt cgg ttc ctg tgg ctt tgg tec tat ctg tet 107 Cys Trp Arg Pro Leu Cys Arg Phe Leu Trp Leu Trp Ser Tyr Leu Ser 1 5 10 15 tat gtt caca gca gtg cct ate cag aaa gtc cag gat gac acc aaa acc 155 Tyr Val Gln Ala Val Pro He Gln Lys Val Gln Asp Asp Thr Lys Thr 20 25 30 etc ate aag acc att gtc acc agg ate aat gac att tea falls acg cag 203 Leu He Lys Thr He Val Thr Arg He Asn Asp He Ser His Thr Gln 35 40 45 tcg gta tec gcc aag cag agg gtc act ggc ttg gac ttc att cct ggg 251 Ser Val Ser Ala Lys Gln Arg Val Thr Gly Leu Asp Phe He Pro Gly 50 55 60 ctt falls ccc att ctg agt ttg tec aag atg gac cag act ctg gca gtc 299 Leu His Pro He Leu Ser Leu Ser Lys Met Asp Gln Thr Leu Ala Val 65 70 75 80 tat ca gg gtc etc acc age ctg cct tec caat aat gtg ctg cag ata 347 Tyr Gln Gln Val Leu Thr Ser Leu Pro Ser Gln Asn Val Leu Gln He 85 90 95 gcc aat gac ctg gag aat etc cga gac etc etc cat ctg ctg gcc ttc 395 Wing Asn Asp Leu Glu Asn Leu Arg Asp Leu Leu His Leu Leu Ala Phe 100 105 110 tec aag age tgc tec ctg cct cag acc agt ggc ctg cag aag cea gag 443 Ser Lys Ser Cys Ser Leu Pro Gln Thr Ser Gly Leu Gln Lys Pro Glu 115 120 125 age ctg gat ggc gtc ctg gaa gcc tea etc tac tec ac gag gtg gtg 491 Ser Leu Asp Gly Val Leu Glu Ala Ser Leu Tyr Ser Thr Glu Val Val 130 135 140 gct ttg age agg ctg cag ggc tet ctg cag gac att ctt caa cag ttg 539 Wing Leu Being Arg Leu Gln Gly Being Leu Gln Asp He Leu Gln Gln Leu 145 150 155 160 gat gtt age cct gaa tgc tgaagtttca aaggccacca ggctcccaag 587 Asp Val Ser Pro Glu Cys 165 aatcatgtag agggaagaaa ccttggcttc caggggtctt caggagaaga gagccatgtg 647 cacacatcca teatteattt ctctccctcc tgtagaccac ccatccaaag gcatgactcc 707 acaatgettg actcaagtta tccacacaac ttcatgagca caaggagggg ccagcctgca 767 gaggggactc teaectagtt cttcagcaag tagagataag agccatccca tcccctccat 827 gtcccacctg ctccgggtac atgttcctcc gtgggtacac gcttcgctgc ggcccaggag 887 gggatgggta aggtgaggta gagcctttgg gctgtctcag agtctttggg agcaccgtga 947 aggctgcatc cacacacagc tggaaactcc caagcagcac acgatggaag caettattta 1007 tttattctgc attctatttt ggatggatct gaagcaaggc ateagetttt tcaggctttg 1067 ggggtcagcc aggatgagga aggctcctgg ggtgctgctt tcaatectat tgatgggtct 1127 gcccgaggca aacctaattt ttgagtgact ggaaggaagg ttgggatctt ccaaacaaga 1187 gtctatgcag gtagcgctca agattgacct ctggtgactg gttttgtttc tattgtgact 1247 gactctatcc aaacacgttt gcagcggcat tgccgggagc ataggctagg ttattatcaa 1307 aagcagatga attttgtcaa gtgtaatatg tatctatgtg cacctgaggg tagaggatgt 1367 gttagaggga gggtgaagga tccggaagtg ttctctgaat tacatatgtg tggtaggctt 1427 i. ~ j «a. - »_ > JtJk? . ttctgaaagg gtgaggcatt ttcttacctc tgtggccaca tagtgtggct ttgtgaaaag 1487 gacaaaggag ttgactcttt ccggaacatt tggagtgtac caggcaccct tggaggggct 1547 aaagctacag gccttttgtt ggcatattgc tgagctcagg gagtgagggc cccacatttg 1607 agacagtgag ccccaagaaa agggtccctg gtgtagatct ccaaggttgt ccagggttga 1667 tctcacaatg cgtttcttaa gcaggtagac gtttgcatgc caatatgtgg ttctcatctg 1727 attggttcat ccaaagtaga accctgtctc ccacccattc tgtggggagt tttgttccag 1787 tgggaatgag aaatcactta gcagatggtc ctgagccctg ggccagcact gctgaggaag 1847 tgccagggcc ccaggccagg ctgccagaat tgcccttcgg gctggaggat gaacaaaggg 1907 gcttgggttt ttccatcacc cctgcaccct atgtcaccat caaactgggg ggcagatcag 1967 cttgatggaa tgagaggaca agcaatacac tttaagactg agcacagttt cgtgctcagc 2027 tctgtctggt gctgtgagct agagaagctc accacataca tataaaaatc agaggctcat 2087 gtccctgtgg ttagacccta ctcgcggcgg tgtactccac cacagcagca ccgcaccgct 2147 ggaagtacag tgctgtcttc aacaggtgtg aaagaacctg agctgagggt gacagtgccc 2207 aggggaaccc tgcttgcagt ctattgcatt tacataccgc atttcagggc acattagcat 2267 ccactc CTAT ggtagcacac tgttgacaat aggacaaggg ataggggttg actatccctt 2327 atccaaaatg cttgggacta gaagagtttt ggattttaga gtcttttcag gcataggtat 2387 atttgagtat atataaaatg agatatcttg gggatggggc ccaagtataa acatgaagtt 2447 catttatatt tcataatacc gtatagacac tgcttgaagt gtagttttat acagtgtttt 2507 aaataacgtt gtatgcatga aagacgtttt tacagcatga acctgtctac tcatgccagc 2567 actcaaaaac cttggggttt tggagcagtt tggatcttgg gttttctgtt aagagatggt 2627 tagcttatac ctaaaaccat aatggcaaac aggctgcagg accagactgg atcctcagcc 2687 ctgaagtgtg cccttccagc caggtcatac cctgtggagg tgagcgggat caggttttgt 2747 ggtgctaaga gaggagttgg aggtagattt tggaggatct gagggc 2793 < 210 > 2 < 211 > 167 .-slS? 3 & , A Á > & 8bÁ &l -? T? UtrJ? *. ri * < . - ?? < 212 > PRT < 213 > Murino < 400 > 2 Met Cys Trp Arg Pro Leu Cys Arg Phe Leu Trp Leu Trp Ser Tyr Leu -1 1 5 10 15 Be Tyr Val Gln Ala Val Pro He Gln Lys Val Gln Asp Asp Thr Lys 20 25 30 • Thr Leu He Lys Thr He Val Thr Arg He Asn Asp He Ser His Thr 35 40 45 10 Gln Ser Val Be Ala Lys Gln Arg Val Thr Gly Leu Asp Phe He Pro 50 55 60 Gly Leu His Pro He Leu Ser Leu Ser Lys Met Asp Gln Thr Leu Wing 65 70 75 Val Tyr Gln Gln Val Leu Thr Ser Leu Pro Ser Gln Asn Val Leu Gln 15 80 85 90 95 He Wing Asn Asp Leu Glu Asn Leu Arg Asp Leu Leu His Leu Leu Wing 100 105 110 Phe Ser Lys Ser Cys Ser Leu Pro Gln Thr Ser Gly Leu Gln Lys Pro 115 120 125 20 Glu Ser Leu Asp Gly Val Leu Glu Ala Ser Leu Tyr Ser Thr Glu Val 130 135 140 Val Wing Leu Ser Arg Leu Gln Gly Ser Leu Gln Asp He Leu Gln Gln 145 150 155 Leu Asp Val Ser Pro Glu Cys 25 160 165 < 210 > 3 < 211 > 700 < 212 > DNA < 213 > Homo sapiens < 220 > < 221 > CDS < 222 > (46) .. (546) < 220 > < 221 > sig_peptide < 222 > (46) .. (48) < 220 > < 221 > mat_peptide < 222 > (49) .. (546) < 220 > < 223 > or of being human (leptin) wherein N represents adenine or guanine or cytokine or thymine < 400 > 3 nnngnngttg caaggcccaa gaagcccann ntcctgggaa ggaaa atg cat tgg gga 57 Met His Trp Gly -ii acc ctg tgc gga ttc ttg tgg ctt tgg ccc tat ctt ttc tat gtc caa 105 Thr Leu Cys Gly Phe Leu Trp Leu Trp Pro Tyr Leu Phe Tyr Val Gln 5 10 15 gct gtg ccc ate caa aaa gtc ca g gat gac acc aaa acc etc ate aag 153 Wing Val Pro He Gln Lys Val Gln Asp Asp Thr Lys Thr Leu He Lys 20 25 30 35 here att gtc acc agg ate aat gac att tea falls acg cag tea gtc tec 201 Thr He Val Thr Arg He Asn Asp He Ser His Thr Gln Ser Val Ser 40 45 50 5 tec aaa cag aaa gtc acc ggt ttg gac ttc att cct ggg etc drops ccc 249 Ser Lys Gln Lys Val Thr Gly Leu Asp Phe He Pro Gly Leu His Pro 55 60 65 ate ctg acc tta tec aag atg gac cag here ctg gca gtc tac ca ca cag 297 He Leu Thr Leu Ser Lys Met Asp Gln Thr Leu Ala Val Tyr Gln Gln 10 70 75 80 ate etc acc agt atg cct tec aga gtg ate ate ata tec aac gac 345 He Leu Thr Ser Met Pro Ser Arg Asn Val He Gln He Ser As Asp 85 90 95 ctg gag aac etc cgg gat ctt ctt falls gtg ctg gcc ttc tet aag age 393 15 Leu Glu Asn Leu Arg Asp Leu Leu His Val Leu Wing Phe Ser Lys Ser 100 105 110 115 tgc falls ttg ccc tgg gcc agt ggc ctg gag acc ttg gac age ctg ggg 441 Cys His Leu Pro Trp Wing Ser Gly Leu Glu Thr Leu Asp Ser Leu Gly 120 125 130 20 ggt gtc ctg gaa gct tea ggc tac tec gg gtg gtg gcg gcc gtc age 489 Gly Val Leu Glu Wing Ser Gly Tyr Ser Thr Glu Val Val Ala Leu Ser 135 140 145 agg ctg cag ggg tet ctg cag gac atg ctg tgg cag ctg gac etc age 537 Arg Leu Gln Gly Ser Leu Gln Asp Met Leu Trp Gln Leu Asp Leu Ser 25 150 155 160 cct ggg tgc tgaggccttg aaggtcactc ttcctgcaag gactnacgtt 586 Pro Gly Cys 165 aagggaagga actctggttt ccaggtatct ccaggattga agagcattgc atggacaccc 646 cttatccagg actctgtcaa tttccctgac tcctctaagc cactcttcca aagg 700 < 210 > 4 < 211 > 167 < 212 > PRT < 213 > Homo sapiens < 400 > 4 Met His Trp Gly Thr Leu Cys Gly Phe Leu Trp Leu Trp Pro Tyr Leu -1 1 5 10 15 Phe Tyr Val Gln Ala Val Pro He Gln Lys Val Gln Asp Asp Thr Lys 20 25 30 Thr Leu He Lys Thr He Val Thr Arg He Asn Asp He Ser His Thr 35 40 45 Gln Ser Val Ser Ser Lys Gln Lys Val Thr Gly Leu Asp Phe He Pro 50 55 60 Gly Leu His Pro He Leu Thr Leu Ser Lys Met Asp Gln Thr Leu Wing 65 70 75 Val Tyr Gln Gln He Leu Thr Ser Met Pro Ser Arg Asn Val He Gln 80 85 90 95 He Be As Asp Leu Glu Asn Leu Arg Asp Leu Leu His Val Leu Wing 100 105 110 Phe Ser Lys Ser Cys His Leu Pro Trp Wing Ser Gly Leu Glu Thr Leu 115 120 125 Asp Ser Leu Gly Gly Val Leu Glu Wing Ser Gly Tyr Ser Thr Glu Val 130 135 140 Val Ala Leu Ser Arg Leu Gln Gly Ser Leu Gln Asp Met Leu Trp Gln 145 150 155 Leu Asp Leu Ser Pro Gly Cys 160 165 < 210 > 5 < 211 > 146 < 212 > PRT < 213 > Mus musculus < 220 > < 223 > mature mouse ob (leptma) < 400 > 5 Val Pro He Gln Lys Val Gln Asp Asp Thr Lys Thr Leu He Lys Thr 1 5 10 15 He Val Thr Arg He Asn Asp He Ser His Thr Gln Ser Val Ser Wing 20 25 30 Lys Gln Arg Val Thr Gly Leu Asp Phe He Pro Gly Leu His Pro He 35 40 45 Leu Ser Leu Ser Lys Met Asp Gln Thr Leu Wing Val Tyr Gln Gln Val 50 55 60 Leu Thr Ser Leu Pro Ser Gln Asn Val Leu Gln He Wing Asn Asp Leu 65 70 75 80 Glu Asn Leu Arg Asp Leu Leu His Leu Leu Ala Phe Ser Lys Ser Cys . Í *.? Í 85 90 95 Ser Leu Pro Gln Thr Be Gly Leu Gln Lys Pro Glu Ser Leu Asp Gly 100 105 110 Val Leu Glu Ala Ser Leu Tyr Ser Thr Glu Val Val Ala Leu Ser Arg 115 120 125 Leu Gln Gly Ser Leu Gln Asp He Leu Gln Gln Leu Asp Val Ser Pro 130 135 140 Glu Cys 145 < 210 > 6 < 211 > 146 < 212 > PRT < 213 > Homo sapiens < 220 > < 223 > or mature human being (leptin) < 400 > 6 Val Pro He Gln Lys Val Gln Asp Asp Thr Lys Thr Leu He Lys Thr 1 5 10 15 He Val Thr Arg He Asn Asp He Ser His Thr Gln Ser Val Ser Ser 20 25 30 Lys Gln Lys Val Thr Gly Leu Asp Phe He Pro Gly Leu His Pro He 35 40 45 Leu Thr Leu Ser Lys Met Asp Gln Thr Leu Wing Val Tyr Gln Gln He 50 55 60 Leu Thr Ser Met Pro Ser Arg Asn Val He Gln He Ser As Asp Leu 65 70 75 80 Glu Asn Leu Arg Asp Leu Leu His Val Leu Wing Phe Ser Lys Ser Cys 85 90 95 His Leu Pro Trp Wing Ser Gly Leu Glu Thr Leu Asp Ser Leu Gly Gly 100 105 110 Val Leu Glu Wing Ser Gly Tyr Ser Thr Glu Val Val Ala Leu Ser Arg 115 120 125 Leu Gln Gly Ser Leu Gln Asp Met Leu Trp Gln Leu Asp Leu Ser Pro 130 135 140 Gly Cys 145

Claims (1)

  1. CLAIMS 1. A method for modulating the transport of leptin through the blood-brain barrier of a mammal, the method comprising: administering to the mammal an effective amount of one or more selected compositions within the group consisting of adrenergic agonists , adrenergic antagonists, neurotransmitters, cytokines, amino acids, opioid peptides, purinergic agonists, glutaminergic agonists and metabolites thereof. 2. The method according to claim 1, in • wherein said composition or said various compositions are administered to the mammal through a pathway of Administration selected from the group consisting of intravenous, intraarterial, intramuscular, intraperitoneal, subcutaneous, topical, intraocular, intracerebroventricular, intracisternal, intrathecal, intradermal, transdermal, nasal, oral and 20 pulmonary. 3. The method according to claim 1, further comprising co-administering to the mammal a leptin selected from the group of leptins comprising the amino acid sequence presented. 25 as SED ID NO: 2, SED ID NO: 4, SED ID NO: 5, SED ID ? ¡¡¡¡¡¡¡¡¡¡¡¡¡¡¡¡¡¡¡BÍi? i¿it¿t.?. ^. a._i ... . . _..,.." -, 4 . . «._. S-Aam. " »«. > «» ... mJk? I * NO: 6, consensus leptins, variants, analogues, leptin fusion proteins, chemically modified derivatives of leptin, and fragments thereof, said leptin optionally having an N-terminal methionine. The method according to claim 2, further comprising co-administering to the mammal a leptin selected from the group consisting of the amino acid sequence presented SED ID NO: 2, SED ID NO: 4, SED ID NO : 5, SED ID NO: 6, consensus leptins, variants, analogues, leptin fusion proteins, chemically modified derivatives of leptin, and fragments thereof, said leptin optionally having an N-terminal methionine. 5. The method according to claims 1, 2, 3, or 4, wherein the adrenergic agonist or the various adrenergic agonists are selected from the group consisting of epinephrine, isoproterenol, arterenol, and cirazoline. 6. The method according to claims 1, 2, 3 or 4, wherein the adrenergic antagonist or the various adrenergic antagonists are selected from the group consisting of yohimbine, phentolamine, prazosin and benoxatine. 7. The method of compliance with any of the -ct »_ .. i. - ,. - .-., ..__.,. t__ Reinvidicaciones 1, 2, 3 or 4, where the cytokine is TNF-a. The method according to claims 1, 2, 3, or 4, wherein the amino acid is tyrosine. The method according to Claims 1, 2, 3, or 4, wherein the purinergic agonist is tyrosine. The method according to Claims 1, 2, 3, or 4, wherein the glutaminergic agonist is glutamate. A method for modulating the weight of a mammal, the method comprises: administering to the mammal an effective amount of one or several compositions selected from the group consisting of adrenergic agonists, neurotransmitters, cytokines, amino acids, opioid peptides, purinergic agonists, glutaminergic agonists and metabolites thereof. The method according to claim 11 wherein said composition or said various compositions are administered to the mammal through a route of administration selected from the group consisting of intravenous, intraarterial, intramuscular, intraperitoneal, subcutaneous, topical administration, intraocular, intracerebroventricular, intracisternal, . Isaá ^ .¿ .._ id.Eat-t3 _.- s,. . . . . ^ '* z .. _ • £, ~ -. . '^. 3. . .- • -. Faith .i t I »a > l intrathecal, intradermal, topical transdermal, nasal, oral, and pulmonary. 13. The method according to claim 11 further comprising co-administering to the mammal a leptin selected from the group of leptins comprising the amino acid sequence SEQ ID NO. 2, SEQ ID No. 4, SEQ ID No. 5, SEQ ID No. 6, consensus leptins, variants, analogs, leptin fusion proteins, chemically modified derivatives of leptin, and fragments thereof, said leptin optionally having a N-terminal methionine. The method according to Claim 12 further comprising co-administering to the mammal a leptin selected from the group of leptins comprising the amino acid sequence presented as SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, consensus leptins, variants, analogs, leptin fusion proteins, chemically modified derivatives of leptin, and fragments thereof, said leptin optionally having an N-terminal methionine. 15. The method according to claim 11, wherein the modulation of body weight is a decrease in body weight. 16. The method of Claims 11, 12, 13, 14, or 15, wherein the adrenergic agonist or the various adrenergic agonists is selected from the group consisting of epinephrine, isoproterenol, arterenol, and cirazoline. 17. The method of Claims 11, 12, 13, 14, or 15, wherein the amino acid is tyrosine. 18. The method of Claims 11, 12, 13, or 14, in • where the cytokine is TNF-a. 19. The method of Claim 11, wherein the modulation of body weight is an increase in body weight. 20. The method of Claims 11, 12 or 19, wherein • the adrenergic antagonist or the various adrenergic antagonists are selected from the group 15 consists of yohimbine, phentolamine, prasozine, and benoxatiano. 21. The method of Claims 11, 12 or 19, wherein the purinergic agonist is adenosine. 22. The method of Claims 11, 12 or 19, wherein the glutaminergic agonist is glutamate. 23. A method for modulating the appetite in the mammal, the method comprising: administering to the mammal an effective amount of one or several selected compositions within the group that 25 consists of adrenergic agonists, antagonists adrenergics, neurotransmitters, cytokines, amino acids, opioid peptides, purinergic agonists, glutaminergic agonists and metabolites thereof. The method of Claim 23, wherein said composition or said various compositions are administered to the mammal through a route of administration selected from the group consisting of intravenous, intraarterial, intramuscular, intraperitoneal, subcutaneous, topical, intraocular, intracerebroventricular, intracisternal, intrathecal, intradermal, topical transdermal, nasal, oral, and pulmonary. The method of Claim 23, further comprising co-administering to the mammal a leptin selects from the group of leptins comprising the amino acid sequence presented as SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 5 , SEQ ID NO: 6, consensus leptins, variants, analogs, leptin fusion proteins, chemically modified derivatives of leptin, and fragments thereof, said leptin optionally having an N-terminal methionine. The method of Claim 24, further comprising co-administering to the mammal a leptin selected from the group of leptins comprising the amino acid sequence set forth as SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 5 , SEQ ID NO: 6, consensus leptins, variants, analogs, leptin fusion proteins, chemically modified derivatives of leptin, and fragments thereof, said leptin optionally having an N-terminal methionine. 27. The method of Claims 23, 24, 25 or 26 wherein the adrenergic agonist or the various adrenergic agonists is selected from the group consisting of epinephrine, isoproterenol, arterenol, and cirazoline. 28. The method of Claims 23, 24, 25 or 26 wherein the adrenergic antagonist or the various adrenergic antagonists is selected from the group consisting of yohimbine, phentolamine, prasozine, and benoxatine. 29. The method of Claims 23, 24, 25 or 26 wherein the cytokine is TNF-a. 30. The method of Claims 23, 24, 25 or 26 wherein the amino acid is tyrosine. 31. The method of Claims 23, 24, 25 or 26 wherein said purinergic agonist is adenosine. 32. The method of Claims 23, 24, 25 or 26 wherein said glutaminergic agonist is glutamate. 33. A pharmaceutical composition useful for modulating body weight, the composition comprising a leptin selected from the group of leptins comprising the amino acid sequence presented in SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, consensus leptins, variants, analogs, leptin fusion proteins, chemically modified derivatives of leptin, and fragments thereof, said leptin optionally having an N-terminal methionine, in combination with an effective amount of an agonist adrenergic, adrenergic antagonist, neurotransmitters, cytokine, amino acid, opioid peptide, purinergic agonists, glutaminergic agonists and metabolites thereof. The pharmaceutical composition according to Claim 33, wherein the adrenergic agonist or the various adrenergic agonists is selected from the group consisting of epinephrine., isoproterenol, arterenol, and cirazoline. The pharmaceutical composition according to Claim 33, wherein the adrenergic antagonist or the various adrenergic antagonists is selected from the group consisting of yohimbine, phentolamine, prasozine, and benoxatiano. The pharmaceutical composition according to Claim 33, wherein the cytokine is TNF-a. The pharmaceutical composition according to Claim 33, wherein the amino acid is tyrosine. The pharmaceutical composition according to Claim 33, wherein said purinergic agonist is adenosm. The pharmaceutical composition according to Claim 33, wherein the glutaminergic agonist is glutamate. The use of one or more adrenergic agonists or metabolites thereof for the preparation of a drug to modulate the transport of leptin through the blood-brain barrier. The use according to Claim 40, wherein the adrenergic agonist or the various adrenergic agonists is selected from the group consisting of epinephrine, isoproterenol, arterenol, and cirazoline. The use of an adrenergic antagonist or various adrenergic antagonists or metabolites thereof (thereof) for the preparation of a drug to modulate the transport of leptin through the blood-brain barrier. The use according to Claim 42, wherein the adrenergic antagonist or the various adrenergic antagonists is selected from the group consisting of yohimbine, phentolamine, prasozine, and benoxatine. 44. The use of one or more neurotransmitters or metabolites thereof (of the same) for the preparation of a drug to modulate the transport of leptin through the blood-brain barrier. 45. The use of one or more peptide hnes for the preparation of a drug to modulate the transport of leptin through the blood-brain barrier. 46. The use of one or several cytokines for the preparation of a drug to modulate the transport of leptin through the blood-brain barrier. 47. The use according to Claim 46, wherein the cytokine is TNF-a. 48. The use of one or several amino acids for the preparation of a drug to modulate the transport of leptin through the blood-brain barrier. 49. The use according to Claim 48 wherein the amino acid is tyrosine. 50. The use of one or more opioid peptides for the preparation of a drug to modulate the transport of leptin through the blood-brain barrier. 51. The use of one or several purinergic agonists for the preparation of a drug to modulate the transport of leptin through the blood-brain barrier. 2. The use according to claim 51, wherein said purinergic agonist is adenosine. * h? - - I 53. The use of a glutaminergic agonist for the preparation of a drug to modulate the transport of leptin through the blood-brain barrier. 54. The use according to Claim 53, wherein the glutaminergic agonist is glutamate. 55. The uses according to any of claims 38 to 54 further comprising the use of leptin for the preparation of said drug to modulate the transport of leptin through the blood-brain barrier. 56. The use according to Claim 55 wherein said leptin is selected from the group consisting of leptin comprising the amino acid sequence presented as SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, consensus leptins, variants, analogs, leptin fusion proteins, chemically modified derivatives of leptin, and fragments thereof, said leptin optionally having an N-terminal methionine. 57. The use of one or more adrenergic agonists or metabolites thereof (of the same) for the preparation of a drug to modulate the transport of leptin through the blood-brain barrier. 58. The use according to Claim 55, wherein the adrenergic agonist or the various agonists Adrenergic drugs are selected from the group consisting of epinephrine, isoproterenol, arterenol, and cirazoline. The use of one or more adrenergic antagonists or metabolites thereof (thereof) for the preparation of a drug to modulate the body weight of a mammal. The use according to claim 59 wherein the adrenergic antagonist or the various adrenergic antagonists are selected from the group consisting of yohimbine., phentolamine, prasozine, and benoxatiano. The use of one or more neurotransmitters or metabolites thereof for the preparation of a drug to modulate the body weight of a mammal. The use of one or more peptide hormones for the preparation of a drug to modulate the body weight of a mammal. The use of one or more cytokines is the preparation of a drug to modulate the body weight of a mammal. The use according to Claim 63 wherein the cytokine is TNF-a. The use of one or more amino acids for the preparation of a drug to modulate the body weight of a mammal. 66. The use according to Claim 65, wherein the amino acid is tyrosine. 67. The use of one or more opioid peptides for the preparation of a drug to modulate the body weight of a mammal. 68. The use of one or more purinergic agonists for the preparation of a drug to modulate the body weight of a mammal. 69. The use according to Claim 68, wherein said purinergic agonist is adenosine. 70. The use of a glutaminergic agonist for the preparation of a drug to modulate the body weight of a mammal. 71. The use according to Claim 70, wherein the glutaminergic agonist is glutamate. 72. The use according to any of Claims 57, 58, 65, or 66, further comprising the use of leptin for the preparation of said drug to modulate the body weight of a mammal. 73. The use according to Claim 72, wherein said leptin is selected from the group consisting of leptin comprising the amino acid sequence presented as SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, consensus leptins, variants, analogs, fusion proteins of íiíÁí if-tJ? TO. leptin, chemically modified derivatives of leptin, and fragments thereof, said leptin optionally having an N-terminal methionine. 74. The use of any of Claims 57, 58, 65, 66 or 72, wherein the modulation of body weight is the reduction of body weight. 75. The use according to Claim 72, wherein the modulation of body weight is the reduction of body weight. 76. The use according to any of Claims 57, 59, 60, 68, 69, 70 or 71 wherein the modulation of body weight is the increase in body weight. fifteen • twenty fifteen
MXPA02001882A 1999-08-23 2000-08-23 Modulation of the blood brain barrier transporter for leptin. MXPA02001882A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US15030099P 1999-08-23 1999-08-23
PCT/US2000/023110 WO2001013935A2 (en) 1999-08-23 2000-08-23 Modulation of the blood-brain barrier transporter for leptin

Publications (1)

Publication Number Publication Date
MXPA02001882A true MXPA02001882A (en) 2002-08-20

Family

ID=22533916

Family Applications (1)

Application Number Title Priority Date Filing Date
MXPA02001882A MXPA02001882A (en) 1999-08-23 2000-08-23 Modulation of the blood brain barrier transporter for leptin.

Country Status (5)

Country Link
EP (1) EP1250131A2 (en)
AU (1) AU7066400A (en)
CA (1) CA2382666A1 (en)
MX (1) MXPA02001882A (en)
WO (1) WO2001013935A2 (en)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007080404A2 (en) * 2006-01-12 2007-07-19 Asterion Limited Leptin ligand
CN102014959B (en) * 2008-03-10 2016-01-20 康奈尔大学 The adjustment of blood-brain barrier permeability
AU2009248914A1 (en) 2008-05-21 2009-11-26 Neurotez, Inc. Methods for Treating Neurodegenerative Disorders Related to Neurofibrillary Tangles
KR20120024529A (en) 2008-11-04 2012-03-14 니콜라오스 테자프시디스 Leptin compositions and methods for treating progressive cognitive function disorders resulting from accumulation of neurofibrillary tangles and amlyoid beta
JP2013540748A (en) * 2010-09-16 2013-11-07 コーネル ユニバーシティー Use of adenosine receptor signaling to regulate blood-brain barrier permeability
EP2601946A1 (en) * 2011-12-06 2013-06-12 Pécsi Tudomànyegyetem Treatment and prevention of diseases related to oxidative stress
GB2571696B (en) 2017-10-09 2020-05-27 Compass Pathways Ltd Large scale method for the preparation of Psilocybin and formulations of Psilocybin so produced
JP6944221B2 (en) 2017-11-20 2021-10-06 ノブメタファーマ カンパニー リミテッド Composition for prevention, amelioration or treatment of bone loss disease containing CHP (cyclo-hispro)
KR20220008824A (en) 2019-04-17 2022-01-21 컴퍼스 패쓰파인더 리미티드 How to treat anxiety disorders, headache disorders and eating disorders with psilocybin
KR102115353B1 (en) * 2019-05-17 2020-05-26 주식회사 노브메타파마 Composition comprising chp (cyclo-his pro) and pth for preventing, improving or treating of bone loss related disease

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4525359A (en) * 1982-12-10 1985-06-25 Greenway Frank L Iii Treatment for selective weight control
DK339289D0 (en) * 1989-07-07 1989-07-07 Dak Lab As PHARMACEUTICAL PREPARATION

Also Published As

Publication number Publication date
WO2001013935A3 (en) 2002-08-15
EP1250131A2 (en) 2002-10-23
CA2382666A1 (en) 2001-03-01
AU7066400A (en) 2001-03-19
WO2001013935A2 (en) 2001-03-01

Similar Documents

Publication Publication Date Title
JP5114381B2 (en) Amylin and amylin agonists for the treatment of psychiatric disorders and disorders
Clapham et al. Anti-obesity drugs: a critical review of current therapies and future opportunities
RU2519073C1 (en) Insulin conjugate with application of immunoglobulin fragment
JP6204369B2 (en) Calcium mimetics and methods of use
AU5990896A (en) Appetite regulating compositions
US20140066368A1 (en) Methods For Affecting Body Composition
US8748375B2 (en) Methods for affecting body composition using amylin agonists
AU2007360979A2 (en) Methods for treating obesity and obesity related diseases and disorders
CZ294983B6 (en) Use of amylin or amylin agonist for the preparation of a medicament intended for treating or preventing obesity in a human subject
MXPA02001882A (en) Modulation of the blood brain barrier transporter for leptin.
JP2023065401A (en) Methods for delivering neuroprotective polypeptide to central nervous system
EP2818181B1 (en) Compositions and medical uses thereof
EP1954313A1 (en) Treatment of obesity and related disorders
CN109718363B (en) Peptide for preventing, relieving or treating Alzheimer disease and application thereof
WO2011050008A2 (en) Combination therapy comprising administration of an amylinomimetic and a pyy peptidomimetic for effecting weight loss and for treating obesity and related metabolic conditions and disorders
US5677279A (en) Methods and compositions for treating pain with amylin or agonists thereof
JP2002522512A (en) Dextran-leptin conjugates, pharmaceutical compositions and related methods
JPH08502969A (en) How to prevent or treat obesity
EP2113256A1 (en) PEGylated rhGH compounds
Finn PEGylation of human growth hormone: strategies and properties
Parkinson et al. The place of pegvisomant in the management of acromegaly
WO2022115369A1 (en) Compositions and methods of inhibiting the binding of plasma igg autoantibodies to serotonin 2a receptor
WO2004089283A2 (en) Compositions and methods for treating viral infections
WO2021140174A1 (en) Glucagon and glp-1 co-agonists for the treatment of chronic kidney disease and diabetic kidney disease in type 2 diabetes
US20030207811A1 (en) Method of treating retinopathy of prematurity using somatostatin analogs

Legal Events

Date Code Title Description
FA Abandonment or withdrawal