WO2024050285A2 - Analogues de leptine pour le traitement de l'obésité et la gestion du poids - Google Patents

Analogues de leptine pour le traitement de l'obésité et la gestion du poids Download PDF

Info

Publication number
WO2024050285A2
WO2024050285A2 PCT/US2023/072937 US2023072937W WO2024050285A2 WO 2024050285 A2 WO2024050285 A2 WO 2024050285A2 US 2023072937 W US2023072937 W US 2023072937W WO 2024050285 A2 WO2024050285 A2 WO 2024050285A2
Authority
WO
WIPO (PCT)
Prior art keywords
leptin
lepr
amino acid
partial agonist
residues
Prior art date
Application number
PCT/US2023/072937
Other languages
English (en)
Other versions
WO2024050285A3 (fr
Inventor
Kenan Christopher GARCIA
Robert Saxton
Original Assignee
The Board Of Trustees Of The Leland Stanford Junior University
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 The Board Of Trustees Of The Leland Stanford Junior University filed Critical The Board Of Trustees Of The Leland Stanford Junior University
Publication of WO2024050285A2 publication Critical patent/WO2024050285A2/fr
Publication of WO2024050285A3 publication Critical patent/WO2024050285A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/72Receptors; Cell surface antigens; Cell surface determinants for hormones
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/5759Products of obesity genes, e.g. leptin, obese (OB), tub, fat
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70596Molecules with a "CD"-designation not provided for elsewhere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • Leptin is an adipocyte-derived protein hormone discovered in the 1990s as a critical regulator of body weight in mammals. Genetic loss of leptin results in increased food intake and severe early onset obesity in both mice and humans, and administration of recombinant leptin is sufficient to restore normal body weight in this context. Although these observations led to substantial interest in the clinical use of leptin for obesity, exogenous leptin treatment is not effective in most obese patients, nearly all of whom already exhibit significantly elevated plasma leptin levels but diminished leptin responsiveness.
  • Leptin exerts its satiety-promoting effects by activating the leptin receptor (LepR) on the surface of a subset of hypothalamic neurons. Activation of LepR in turn drives the phosphorylation and activation of the Signal Transducer and Activator of Transcription 3 (STAT3), which drives production of anorexigenic peptides that suppress food intake and increase energy expenditure.
  • Leptin is produced by adipocytes such that levels of circulating leptin are proportional to the amount of adipose tissue, thereby serving as a homeostatic feedback mechanism to correlate food intake with organismal energy stores.
  • compositions and methods are provided for preventing or treating obesity and/or overweight, and managing body weight.
  • the compositions comprise partial agonists of the leptin receptor (lepr), which partial agonists elicit sub-maximal signaling at saturating ligand concentrations, and bias the response to STAT3 signaling.
  • lepr leptin receptor
  • Human leptin protein variants for example leptin modified to increase affinity at site 2 binding site(s) and decrease binding at site 3 binding site(s), preferentially suppress phosphorylation of SHP2, ERK, and STAT1 relative to STAT3, resulting in a biased signal that selectively activates pathways associated with satiety, resulting in decreased activation of pathways associated with leptin resistance.
  • the variant proteins find use in the suppression of appetite; and can provide for therapeutic weight loss.
  • a partial agonist of lepr is a variant of leptin protein, comprising one or more amino acid modifications, e.g. amino acid substitutions, that alter the binding to the lepr receptor.
  • Partial agonists may elicit a maximal STAT3 response of from about 25% to about 75% relative to the wild-type ligand, e.g. wild-type leptin.
  • a partial agonist of interest induces STAT3-biased signaling, where maximal phosphorylation of phosphatase SHP2, ERK, or STAT1 is less than about 25% of the phosphorylation obtained with the wild-type ligand, less than about 10%, less than about 5%, less than about 1%.
  • amino acid substitutions are made at one or more “site 3” residues of human leptin, which residues are shown in FIG. 10, and are, relative to the mature human wildtype protein, SEQ ID NO:1 , K33, Q34, K35, T37, S1 17, Y1 19, S120.
  • the combined effect of amino acid modifications at site 3 residues reduce binding to the site 3 residues, e.g. by reducing from about 2-fold to about 500-fold relative to wild-type, from about 5-fold to about 100-fold, from about 5-fold to about 50-fold, from about 5-fold to about 10-fold.
  • amino acid modifications thus generate a partial agonist of lepr.
  • amino acid modifications e.g. a substitution to an amino acid other than the wild-type; or an amino acid deletion, is provided at one, two, three, four, five or more residues selected from K33, Q34, K35, T37, S117, Y1 19, and S120.
  • amino acid modifications are made at K33, including without limitation K33A, K33E, K33D, K33I, K33L, K33T, K33S, K33V.
  • amino acid modifications are made at Q34, including without limitation Q34A, Q34E, Q34D, Q34I, Q34L, Q34T, Q34S, Q34V, Q34K, Q34R.
  • amino acid modifications are made at K35; including without limitation K34A, K34E, K34D, K34I, K34L, K34T, K34S, K34V.
  • amino acid modifications are made at T37, including without limitation T37A, T37R, T37N, T37D, T37Q, T37E, T37K, T37I, T37L, T37V.
  • amino acid modifications are made at S1 17, including without limitation S117A, S1 17E, S117D, S1 17I, S1 17L, S117V, S117K, S117R, S1 17N, S117Q.
  • amino acid modifications are made at Y119, including without limitation Y1 19A, Y119E, Y1 19D, Y119I, Y1 19L, Y119V, Y1 19K, Y119R, Y1 19N, Y1 19Q, Y1 19F, Y1 19W.
  • amino acid modifications are made at S120, including without limitation S120A, S120R, S120N, S120D, S120Q, S120E, S120K, S120I, S120L, S120V.
  • amino acid modifications are made at E122, including without limitation E122A, E122R, E122K, E122I, E122L, E122T, E122S, E122V.
  • modifications are made at each of K33, Q34, K35.
  • modifications are made at S1 17; and may be made at each of K33, Q34, K35 and S1 17.
  • one or more amino acid modifications are made at one or more “site 2” residues, which residues are shown in FIG. 10, and are, relative to the human wild-type protein, SEQ ID NO:1 , K5, D9, L13, K15, T16, R20, N72, Q75, N82, D85, and L86.
  • a consideration for the therapeutic LepR agonists is that the serum levels of endogenous leptin are highly elevated in the context of human obesity. It is therefore desirable to include in protein modifications one or more modifications that enhance affinity of binding to site 2 residues, e.g.
  • amino acid modifications e.g. a substitution to an amino acid other than the wild-type; or an amino acid deletion
  • amino acid modifications is provided at one, two, three, four, five or more residues selected from K5, D9, L13, K15, T16, R20, N72, Q75, N82, D85, and L86.
  • an amino acid modification is made at D23, including without limitation D23L, which may be combined with modifications at each of K33, Q34, K35, at S1 17; or at each of K33, Q34, K35 and S117.
  • the present disclosure provides for variant leptin proteins as disclosed above, pharmaceutical formulations comprising such proteins, and for methods of using such proteins.
  • the present invention provides a pharmaceutical formulation comprising a therapeutically effective amount of a variant partial agonist leptin protein, alone or in combination with a pharmaceutically acceptable carrier.
  • the formulation may be provided in a unit dose, comprising an effective dose of the protein.
  • a formulation comprising an effective dose of a variant leptin partial agonist and a pharmaceutically acceptable excipient, where the therapeutically effective amount of the variant leptin partial agonist is in the range of from about 0.1 mg/kg to about 100 mg/kg.
  • the effective dose is at least about 0.1 mg/kg, at least about 0.5 mg/kg, at least about 1 mg/kg, at least about 5 mg/kg, at least about 10 mg/kg, at least about 20 mg/kg, at least about 50 mg/kg, up to about 100 mg/kg, in some embodiments the effective dose is from about 1 to 25 mg/kg.
  • Dosing may be daily, every 2 days, every 3 or more days, e.g. weekly, semi-weekly, bi-weekly, monthly, etc. Dosing may be parenteral, including sustained release formulations.
  • a method for weight management which may be associated with treating or delaying the progression or onset of diabetes and metabolic syndrome, especially type II diabetes, which include reducing complications of diabetes such as retinopathy, neuropathy, nephropathy and delayed wound healing, and related diseases such as insulin resistance (impaired glucose homeostasis), hyperglycemia, hyperinsulinemia, elevated blood levels of fatty acids or glycerol, hyperlipidemia including hypertriglyceridemia, Syndrome X, atherosclerosis and hypertension, wherein a therapeutically effective amount of a variant leptin partial agonist is administered to a mammalian, e.g., human, patient in need of treatment.
  • a mammalian e.g., human
  • a method for treating obesity and related diseases as defined herein wherein a therapeutically effective amount of a variant leptin partial agonist is administered to a mammalian, e.g., human, patient in need of treatment.
  • the reduced food intake observed with administration the variant leptin partial agonist is associated with weight loss, e.g. loss of 1 % body weight, 2.5%, 5%, 7.5%, 10%, 12.5%, 15%, 17.5%, 20%, 22.5%, 25%, 27.5%, 30% or more, depending on the initial weight of the subject.
  • these methods of treatment may be combined with one or more other types of therapeutic agent, such as an antidiabetic agent, a hypolipidemic agent or anti-obesity agent, e.g. metformin, sulfonylureas, e.g. glyburide, glipizide, glimepiride; glinides, e.g. repaglinide and nateglinide; thiazolidinediones, e.g. rosiglitazone, pioglitazone; DPP-4 inhibitors, e.g. sitagliptin, saxagliptin, linagliptin; GLP-1 receptor agonists, e.g.
  • an antidiabetic agent e.g. metformin, sulfonylureas, e.g. glyburide, glipizide, glimepiride
  • glinides e.g. repaglinide and
  • exenatide, liraglutide, semaglutide; SGLT2 inhibitors, e.g. canagliflozin, dapagliflozin, empagliflozin, etc. is administered to a human patient in need of treatment.
  • SGLT2 inhibitors e.g. canagliflozin, dapagliflozin, empagliflozin, etc.
  • the other therapeutic agents when employed in combination with the compounds of the present invention may be used, for example, in those amounts indicated in the Physician's Desk Reference, as in the patents set out above or as otherwise determined by one of ordinary skill in the art.
  • FIGS. 1 A-1 D Cryo-EM structure of the leptin receptor signaling complex.
  • A Schematic depicting LepR ECD domain architectures, and the regions analyzed for structure determination using cryo-EM.
  • B Cryo-EM 2-dimensional class averages of assembled leptin-bound LepR D1 D7 and LepR D3 D7 complexes.
  • C Overlaid segmented density maps of the Leptin-LepR D1 D7 and Leptin-LepR D3 D7 complexes resolved to 5.9 A and 4.5 A resolution, respectively.
  • D Three views of the leptin-LepR D1 D7 structural model, with leptin in salmon and LepR in purple.
  • FIGS. 2A-2E Structural homology between leptin and IL-6 family cytokine receptor complexes.
  • A Side and top views of leptin-bound LepR structural model (PDB ID: 8DH9), showing the leptin-binding domains (D3-D5) of LepR, with leptin in salmon and LepR in purple.
  • B Side and top views of the hexameric IL-6 receptor complex (PDB ID: 1 P9M) showing IL-6 in pink, gp130 in green, and IL6Rct in gray.
  • D Schematic showing how LepR mutants L503S/L504S (LepR- 2KO) and L370S (LepR-3KO) assemble to exclusively form an asymmetric 1 :2 leptin-LepR complex, in the same conformation as the partially open 2:2 complex observed in our structure.
  • FIGS. 3A-3K Structural basis for leptin dependent LepR dimerization.
  • A Front view of the segmented density map of the leptin-LepR D3 D7 complex resolved to 4.5 A resolution (transparent) with the focus refined map encompassing leptin and the leptin-binding domains of LepR, resolved to 3.8 A resolution (solid).
  • B, C Close-up views of the leptin-LepR site 2 binding interface. Hydrogen bonds and salt-bridges are shown as black dashed-lines.
  • E Top view of the segmented density maps shown in (A).
  • F Close-up view of the leptin-LepR site 3a binding interface.
  • G Immunoblot of lysates prepared and analyzed as in (C).
  • H Comparison of the apoleptin structure (PDB ID: 1 AX8) and LepR-bound leptin structure (this paper, PDB ID: 8DHA), showing leptin in salmon, LepR D3 in purple.
  • FIGS. 4A-4I Biased leptin analogs decouple activation of STAT3 from LepR negative regulators.
  • A Cartoon model of LepR signaling in which STAT3 recruited by phosphorylation of LepR-Y1141 , whereas LepR negative regulators are recruited by phosphorylation of LepR-Y986.
  • B Phospho-STAT3 dose-response curves for WT or mutant leptin in LepR-expressing HEK 293T cells, analyzed by flow cytometry and shown as a percent of maximal WT leptin mean fluorescent intensity (MFI; mean ⁇ SD, n>2).
  • (D) Quantification of immunoblots prepared as in (C) (100 nM leptin, mean ⁇ SEM, n 4, two-sided t-test).
  • FIGS. 5A-5D Assembly and purification of the mouse leptin receptor complex, (a-d) Sizeexclusion chromatography profiles and corresponding SDS-PAGE gels of the assembled Leptin- LepRD1-D7 complex (a), crosslinked Leptin-LepRD1-D7 complex (b), Leptin-LepRD3-D7 complex (c), and crosslinked Leptin-LepRD3-D7 complex (d).
  • Green bars indicate the Leptin- LepR complex
  • orange bars indicate free Leptin
  • red box indicates fractions that were collected for further processing and structural characterization.
  • FIGS. 6A-6F Leptin-LepRD1 -D7 complex cryo-EM data processing, (a) Representative cryo- EM micrograph of the leptin-LepRD1-D7 complex, (b) Reference free 2D class averages of the Leptin-LepRD1 -D7 complex, (c) Workflow for processing Leptin-LepRD1 -D7 complex cryo-EM dataset, (d) Local resolution of the Leptin-LepRD1 -D7 complex cryo-EM density map, ranging from 4.0 A to 7.5 A resolution, (e) Euler angle orientation distribution for Leptin-LepRDI - D7 complex cryo-EM dataset, (f) Half-set gold-standard FSC and map vs model FSC curves for Leptin-LepR D1 D7 complex.
  • FIGS. 7A-7I Leptin-LepRD3-D7 complex cryo-EM data processing, (a) Representative cryo- EM micrograph of the Leptin-LepRD3-D7 complex, (b) 2D class averages of the Leptin- LepRD3-D7 complex, (c) Workflow for processing Leptin-LepRD3-D7 complex cryo-EM dataset, (d) Local resolution of the Leptin- LepRD3-D7 complex cryo-EM density map, ranging from 2.0 A to 6.0 A resolution, (e) Euler angle orientation distribution for Leptin-LepRD3-D7 complex cryo- EM dataset, (f) Half-set gold-standard FSC and map vs model FSC curves for Leptin-LepRD3- D7 complex, (g) Local resolution cryo-EM density map of the focus refined region of the Leptin- LepRD3-D7 complex, ranging from 2.0 A to 4.5 A resolution, (h) Euler angle orientation distribution
  • FIGS. 8A-8D Analysis of cryo-EM map quality, (a) Molecular model of the leptin-LepRDI - D7 complex fit into its 5.9 A resolution cryo-EM density map. (b) Sample cryo-EM density map and fitted molecular model for key regions of the 4.5 A resolution cryo-EM density map of the leptin-LepRD3-D7 complex. (c,d) Sample cryo-EM density map and fitted molecular model for key regions at the focus refine d 3.8 A resolution map of the leptin-LepR interface, showing that secondary structure and bulky side chain s can be resolved at the current resolution.
  • FIGS. 9A-9C Analysis of “open” LepR complex, (a) Quantification of immunoblot in Figure 2e. (b) Top and side views for the Leptin-LepRD3-D7 complex reported here, highlighting the asymmetric, “open” conformation of the one LepR D3 domain, and absence of steric hindrance at the membrane proximal D7 domains, (c) Top and side view of Leptin-LepR model in which the second LepR D3 domain is closed to form a site 3 contact with the second bound Leptin, analogous the symmetric receptor complex observed in the IL-6 receptor complex structure. This rotation results in a significant steric clash between the membrane proximal D7 domains of the two LepR chains.
  • FIGS. 10A-10B Conservation of the Leptin-LepR binding interface, (a) Sequence alignment of mouse (SEQ ID NO:2) and human (SEQ ID NO:1 ) Leptin with positions according to increasing sequence identity. Residues involved in LepR binding at site 2 and site 3 are indicated, (b) Sequence alignment of the Leptin binding domains of mouse and human LepR (D3 (SEQ ID NO:3 and SEQ ID NO:6), D4 (SEQ ID NO:4 and SEQ ID NO:7), and D5 (SEQ ID NO:5 and SEQ ID NO:8) with positions colored according to increasing sequence identity. Residues involved in Leptin binding at site 2 and site 3 are indicated.
  • FIGS. 1 1 A-1 1 E Functional Characterization of Leptin and LepR mutants, (a, b) Quantification of immunoblots in Figure 3d (a) and 3g (b). (C) Coomassie-stained SDS-PAGE gel of recombinant leptin variants, (c) Coomassie-stained SDS-PAGE gel of recombinant leptin variants, (d) Quantification of immunoblots from Figure 3k. (d) Quantification of immunoblots from Figure 3K. (e) Table of calculated EC50 and Emax values for all leptin mutants tested on mLepR expressing 293T cells.
  • compositions and methods are provided for preventing or treating obesity and/or overweight; and managing body weight.
  • the compositions comprise partial agonists of the leptin receptor (lepr), which partial agonists elicit sub-maximal signaling at saturating ligand concentrations; and bias the response to STAT3 signaling.
  • lepr leptin receptor
  • Human leptin protein variants provided herein preferentially suppress phosphorylation of SHP2 and ERK relative to STAT3, resulting in a biased signal that selectively activates pathways associated with satiety, with decreased activation of pathways associated with leptin resistance.
  • the variant proteins find use in the suppression of appetite, and can provide for therapeutic weight loss.
  • Leptin is a hormone that regulates food intake, body mass, and reproductive function. It is encoded by the obese (ob) gene, and expressed by fat cells in white adipose tissue.
  • the leptin receptor is the homodimeric leptin receptor (LEP-R), which is structurally similar to the class I family of cytokine receptors.
  • Leptin regulates appetite and metabolism by inhibiting the synthesis and release of neuropeptide Y (NPY) in the arcuate nucleus (ARC). Leptin can inhibit neural pathways activated by appetite stimulants (orexigenic) to reduce energy intake and activate pathways targeted by anorexigenic to suppress appetite.
  • leptin affects the transcription of proopiomelanocortin POMC, whose a-MSH product is released into the synapse to activate neurons via binding to the melanocortin receptor (MGR) and leads to appetite-suppression.
  • MGR melanocortin receptor
  • the mature human leptin protein has the reference sequence (SEQ ID NO:1 ) VPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPILTLSKMDQTLAVYQQILTS MPSRNVIQISNDLENLRDLLHVLAFSKSCHLPWASGLETLDSLGGVLEASGYSTEVVALSRLQG SLQDMLWQLDLSPGC.
  • the mature mouse leptin protein has the reference sequence (SEQ ID NO:2) VPIQKVQDDTKTLIKTIVTRINDISHTQSVSSKQKVTGLDFIPGLHPILTLSKMDQTLAVYQQILTS MPSRNVIQISNDLENLRDLLHVLAFSKSCHLPWASGLETLDSLGGVLEASGYSTEVVALSRLQG SLQDMLWQLDLSPGC.
  • Leptin-mediated dimerization of LepR results in the JAK2-dependent phosphorylation of multiple tyrosine residues on the intracellular domain (ICD) of LepR, each of which have distinct biological roles.
  • Phosphorylation of Tyr1141 drives activation of STAT3 and the subsequent satiety promoting effects of leptin, whereas phosphorylation of Tyr986 activates the SHP2/ERK pathway and is required for recruitment of cytosolic LepR antagonists that drive leptin resistance.
  • Leptin is overexpressed at the gene level in the adipose tissue of individuals with obesity, and strong positive associations exist between plasma leptin levels and body fat percentage.
  • Leptin resistance refers to states in which leptin fails to promote its anticipated effects, frequently coexisting with hyperleptinaemia. Leptin resistance is closely associated with obesity and also observed in physiological situations such as pregnancy and in seasonal animals.
  • Leptin resensitisation refers to the reversion of leptin-resistant states and is associated with improvement in endocrine and metabolic disturbances commonly observed in obesity and a sustained decrease of plasma leptin levels, possibly below a critical threshold level.
  • leptin resensitisation can be achieved with treatments that reduce body adiposity and leptinaemia, or with some pharmacological compounds, while physiological leptin resistance reverts spontaneously.
  • leptin resistance is not fully understood, substantial evidence suggests that it is driven by chronic hyperleptinemia and the resulting desensitization of LepR to leptin, due in large part to the expression and recruitment of cytosolic LepR antagonists such as Suppressor of Cytokine Signaling 3 (SOCS3).
  • SOCS3 Cytokine Signaling 3
  • lepr sites are consistent with the art, e.g. for example see Peelman et al. (2004) Protein Structure and Folding 279(39): 41038-41046, herein specifically incorporated by reference.
  • binding site II or “site 2” residues, which comprise, relative to the human wildtype protein, SEQ ID NO:1 , K5, D9, L13, K15, T16, R20, N72, Q75, N82, D85, and L86. In general these modification increase the affinity of leptin to lepr.
  • polypeptide peptide
  • protein protein
  • amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymer.
  • sequence identity refers to the subunit sequence identity between two molecules. When a subunit position in both of the molecules is occupied by the same monomeric subunit (e.g., the same amino acid residue or nucleotide), then the molecules are identical at that position. The similarity between two amino acid or two nucleotide sequences is a direct function of the number of identical positions. In general, the sequences are aligned so that the highest order match is obtained. If necessary, identity can be calculated using published techniques and widely available computer programs, such as the GCS program package (Devereux et al., Nucleic Acids Res. 12:387, 1984), BLASTP, BLASTN, FASTA (Atschul et al., J. Molecular Biol. 215:403, 1990).
  • protein variant or “variant protein” or “variant polypeptide” herein is meant a protein that differs from a wild-type protein by virtue of at least one amino acid modification.
  • Variant polypeptide may refer to the polypeptide itself, a composition comprising the polypeptide, or the amino sequence that encodes it.
  • the variant polypeptide has at least one amino acid modification compared to the parent polypeptide, e.g. from about one to about ten amino acid modifications, and preferably from about one to about five amino acid modifications compared to the parent.
  • amino acid refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids.
  • Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, e.g., hydroxyproline, gamma- carboxyglutamate, and O-phosphoserine.
  • amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an a-carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid.
  • Amino acid mimetics refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but that functions in a manner similar to a naturally occurring amino acid.
  • peptide residue and “peptidic structure” are intended to include peptides comprised of naturally-occurring L-amino acids and the corresponding D-amino acids, as well as peptide derivatives, peptide analogues and peptidomimetics of the naturally- occurring L-amino acid, structures.
  • Approaches to designing peptide analogues, derivatives and mimetics are known in the art. For example, see Veber and Freidinger 1985 T/NS p. 392; Evans, et al. 1987 J. Med. Chem. 30:229.
  • Peptidomimetics that are structurally similar to therapeutically useful peptides may be used to produce an equivalent or enhanced therapeutic or prophylactic effect, by methods known in the art and further described in the following references: Spatola, A. F. 1983 in: Chemistry and Biochemistry of Amino Acids, Peptides, and Proteins, B. Weinstein, eds., Marcel Dekker, New York, p. 267; Holladay, et al. 1983 Tetrahedron Lett. 24:4401 -4404.
  • a “derivative” of a compound refers to a form of that compound in which one or more reactive groups in the compound have been derivatized with a substituent group.
  • peptide derivatives include peptides in which an amino acid side chain, the peptide backbone, or the amino- or carboxy-terminus has been derivatized (for example, peptidic compounds with methylated amide linkages or hydroxylated amino acids or amino acid residues).
  • amino acid structure is intended to include the amino acid, as well as analogs, derivatives and mimetics of the amino acid that maintain the functional activity of the compound.
  • phenylalanine structure is intended to include phenylalanine as well as pyridylalanine and homophenylalanine.
  • leucine structure is intended to include leucine, as well as substitution with valine, isoleucine or other natural or nonnatural amino acid having an aliphatic side chain, such as norleucine.
  • amino- and/or carboxy-terminus of the peptide compounds disclosed herein can be standard amino and carboxy termini as seen in most proteins.
  • amino- and/or carboxy-terminus of the peptide compound can be chemically altered by the addition or replacement of a derivative group.
  • Amino-derivative groups which can be present at the N- terminus of a peptide compound include acetyl, aryl, aralkyl, acyl, epoxysuccinyl and cholesteryl groups.
  • Carboxy-derivative groups which can be present at the C-terminus of a peptide compound include alcohol, aldehyde, epoxysuccinate, acid halide, carbonyl, halomethane, diazomethane groups and carboxamide.
  • modified refers to a polypeptide which retains the overall structure of a related polypeptide but which differs by at least one residue from that related polypeptide.
  • a modified C-terminus is a C-terminus of a polypeptide that has a chemical structure other than a standard peptide carboxy group, an example of such a modified C-terminus being a C-terminal carboxamide.
  • pharmaceutically acceptable carrier refers to a carrier medium which does not interfere with the effectiveness of the biological activity of the active ingredients and which is not toxic to the host or patient.
  • peptide residue and "peptidic structure” are intended to include peptides comprised of naturally-occurring L-amino acids and the corresponding D-amino acids, as well as peptide derivatives, peptide analogues and peptidomimetics of the naturally- occurring L-amino acid structures.
  • Approaches to designing peptide analogues, derivatives and mimetics are known in the art (see Farmer, P.S. in: Drug Design E.J. Ariens, ed. Academic Press, New York, 1980, vol. 10, pp. 1 19-143; Ball J.B. & Alewood, P.F. 1990 /. Mol. Recognition 3:55; Luthman, et al.
  • constrained peptides may be generated by methods known in the art (Rizo, et al. 1992 Ann. Rev. Biochem. 61 :387); for example, by adding internal cysteine residues or organic linkers capable of forming intramolecular bridges which cyclize the peptide, adding cyclic lactam bridge, or the use of flexible 6-aminohexanoic acid (Ahx), rigid aminoisobutyric acid (Aib) or D-amino acid residues to alter the stability of the helix.
  • Synthetic or non-naturally occurring amino acids refer to amino acids which do not naturally occur in vivo but which, nevertheless, can be incorporated into the peptide structures described herein.
  • polypeptides of the disclosure are modified by covalent linkage to a heterologous moiety, which moiety may comprise a polymer, an Fc, an FcRn binding ligand, immunoglobulin, albumin, a collagen-binding motif, etc.
  • a covalently linked polymer may be selected from the group consisting of a polyethyleneglycol (PEG) moiety; a polypropylenglycol (PPG) moiety; a PAS moiety; which is an amino acid sequence comprising mainly alanine and serine residues or comprising mainly alanine, serine, and proline residues, the amino acid sequence forming random coil conformation under physiological conditions [US No.
  • HES hydroxyethylstarch
  • An "Fc region” can be a naturally occurring or synthetic polypeptide that is homologous to an IgG C-terminal domain produced by digestion of IgG with papain.
  • IgG Fc has a molecular weight of approximately 50 kDa.
  • the leptin polypeptide may be fused to the entire Fc region, or a smaller portion that retains the ability to extend the circulating half- life of a chimeric polypeptide of which it is a part.
  • full-length or fragmented Fc regions can be variants of the wildtype molecule. That is, they can contain mutations that may or may not affect the function of the polypeptides; as described further below, native activity is not necessary or desired in all cases.
  • the leptin polypeptide can comprise a polypeptide that functions as an antigenic tag, such as a FLAG sequence.
  • FLAG sequences are recognized by biotinylated, highly specific, anti-FLAG antibodies, as described herein (see also Blanar et al., Science 256: 1014, 1992; LeClair et al., Proc. Natl. Acad. Sci. USA 89:8145, 1992).
  • the chimeric polypeptide further comprises a C-terminal c-myc epitope tag.
  • the PEG molecular weight may be between about 1 kDa and about 100 kDa for ease in handling and manufacturing.
  • the PEG may have an average molecular weight of about 200, 500, 1000, 2000, 4000, 8000, 16,000, 32,000, 64,000, or 100,000 kDa.
  • the PEG may have a branched structure (U.S. Pat. No. 5,643,575; Morpurgo et al. AppL Biochem. Biotechnol. 56:59-72 (1996); Vorobjev et al, Nucleosides Nucleotides 18:2745-2750 (1999); and Caliceti et al, Bioconjug.
  • modified polypeptide derivatives comprise one or more substitutions of disulfide bonds with lactam bridges to increase the metabolic stability of the peptides. Cystathiones are resistant towards thiol reduction. Therefore, substitutions of disulfides with thioethers, or selenosulfide, diselenide and ditelluride bridges can provide protection against reduction [Knerr et al., ACS Chem Biol , 6(7), 753-760, 2011 ; Muttenthaler et al. J Med Chem., 53(24), 8585-8596, 2010].
  • Peptide disulfide bond mimics based on diaminodiacids can also be used to improve the stability of analogs (Cui et al., Angew Chem, 125, 9737-9741 , 2013).
  • the disulfide bridge can also be modified either by the insertion of linkers or bridges of a different nature.
  • polypeptides are modified by the addition of one or more alkane, cholesterol, or PEG-cholesterol moieties to increase the metabolic stability of the peptides.
  • Stapled peptides via the introduction of a synthetic brace (staple), can be synthesized using ring-closing metathesis to lock a peptide in a specific conformation and reduce conformational entropy.
  • the sequence of leptin may be altered in various ways known in the art to generate targeted changes in sequence.
  • the polypeptide will usually be substantially similar to the sequences provided herein, i.e. will differ by at least one amino acid, and may differ by at least two but not more than about ten amino acids.
  • the sequence changes may be substitutions, insertions or deletions, including truncation at the corboxy or the amino terminus. Scanning mutations that systematically introduce alanine, or other residues, may be used to determine key amino acids.
  • Conservative amino acid substitutions typically include substitutions within the following groups: (glycine, alanine); (valine, isoleucine, leucine); (aspartic acid, glutamic acid); (asparagine, glutamine); (serine, threonine); (lysine, arginine); or (phenylalanine, tyrosine).
  • Modifications of interest that do not alter primary sequence include chemical derivatization of polypeptides, e.g., acetylation, amidation, acylation, or carboxylation. Also included are modifications of glycosylation, e.g. those made by modifying the glycosylation patterns of a polypeptide during its synthesis and processing or in further processing steps; e.g. by exposing the polypeptide to enzymes which affect glycosylation, such as mammalian glycosylating or deglycosylating enzymes. Also embraced are sequences that have phosphorylated amino acid residues, e.g. phosphotyrosine, phosphoserine, or phosphothreonine.
  • modifications of glycosylation e.g. those made by modifying the glycosylation patterns of a polypeptide during its synthesis and processing or in further processing steps; e.g. by exposing the polypeptide to enzymes which affect glycosylation, such as mammalian glycosylating or de
  • polypeptides that have been modified using ordinary molecular biological techniques and synthetic chemistry so as to improve their resistance to proteolytic degradation or to optimize solubility properties or to render them more suitable as a therapeutic agent.
  • the backbone of the peptide may be cyclized to enhance stability (see Friedler et al. (2000) J. Biol. Chem. 275:23783-23789).
  • Analogs of such polypeptides include those containing residues other than naturally occurring L-amino acids, e.g. D-amino acids or non-naturally occurring synthetic amino acids.
  • the present invention includes within its scope pharmaceutical compositions comprising, as an active ingredient, a therapeutically effective amount of at least one lepr partial agonist, alone or in combination with a pharmaceutical carrier or diluent.
  • a pharmaceutical carrier or diluent e.g., a pharmaceutically acceptable carrier or diluent.
  • polypeptides of the present invention can be used alone, in combination with other compounds of the invention, or in combination with one or more other therapeutic agent(s), e.g., an antidiabetic agent or other pharmaceutically active material.
  • cysteines can be used to make thioethers, histidines for linking to a metal ion complex, carboxyl groups for forming amides or esters, amino groups for forming amides, and the like.
  • Polypeptides described herein can be prepared by, for example, by using standard solid phase techniques, or by expression in a cell through recombinant methods. (See Merrifield, 1963. Am. Chem. Soc. 85:2149; J.M. Stewart and J.D. Young, 1984 Solid Phase Peptide Syntheses 2nd Ed., Pierce Chemical Company). These procedures can also be used to synthesize peptides in which amino acids other than the 20 naturally occurring, genetically encoded amino acids are substituted at one, two, or more positions of any of the modified peptides as disclosed herein. For instance, naphthylalanine can be substituted for tryptophan, facilitating synthesis.
  • Other synthetic amino acids that can be substituted into the peptides of the present embodiments include L-hydroxypropyl, L-3, 4-dihydroxy-phenylalanyl, d amino acids such as L-d-hydroxylysyl and D-d-methylalanyl, L-a-methylalanyl,
  • D amino acids and non- naturally occurring synthetic amino acids can also be incorporated into the peptides of the present embodiments (see Roberts, et al. 1983 Unusual Amino/ Acids in Peptide Synthesis 5:341 -449).
  • the naturally occurring side chains of the 20 genetically encoded amino acids, or any other side chain as disclosed herein can be transposed to the nitrogen of the amino acid, instead of the a-carbon as typically found in peptides.
  • the peptides can be synthesized in a stepwise manner on an insoluble polymer support (also referred to as "resin") starting from the C-terminus of the peptide.
  • a synthesis is begun by appending the C-terminal amino acid of the peptide to the resin through formation of an amide or ester linkage. This allows the eventual release of the resulting peptide as a C-terminal amide or carboxylic acid, respectively.
  • the C-terminal residue may be attached to 2-Methoxy-4-alkoxybenzyl alcohol resin (SASRIN.TM., Bachem Bioscience, Inc., King of Prussia, Pa.) as described herein and, after completion of the peptide sequence assembly, the resulting peptide alcohol is released with LiBH.sub.4 in THF (see J. M. Stewart and J. D. Young, supra, p. 92).
  • SASRIN.TM. 2-Methoxy-4-alkoxybenzyl alcohol resin
  • the syntheses of the peptides described herein can be carried out by using a peptide synthesizer, such as an Advanced Chemtech Multiple Peptide Synthesizer (MPS396) or an Applied Biosystems Inc.
  • peptide synthesizer (ABI 433A). If the MPS396 was used, up to 96 peptides were simultaneously synthesized. If the ABI 433A synthesizer was used, individual peptides were synthesized sequentially. In both cases the stepwise solid phase peptide synthesis was carried out utilizing an Fmoc/t-butyl protection strategy.
  • Peptides with the desired purity can be obtained by purification using preparative HPLC, for example, on a Waters Model 4000 or a Shimadzu Model LC-8A liquid chromatograph.
  • the solution of crude peptide is injected into a YMC S5 ODS column and eluted with a linear gradient of MeCN in water, both buffered with 0.1 % TFA, using a flow rate of 14-20 mL/min with effluent monitoring by UV absorbance at 220 nm.
  • the structures of the purified peptides can be confirmed by electro-spray MS analysis.
  • the polypeptides may also be isolated and purified in accordance with conventional methods of recombinant synthesis.
  • a lysate may be prepared of the expression host and the lysate purified using HPLC, exclusion chromatography, gel electrophoresis, affinity chromatography, or other purification technique.
  • the compositions which are used will be substantially pure, e.g. the peptide of interest will comprise at least 20% by weight of the desired product, more usually at least about 75% by weight, preferably at least about 95% by weight, and for therapeutic purposes, usually at least about 99.5% by weight, or more, in relation to contaminants related to the method of preparation of the product and its purification. The percentages may be based upon total protein.
  • the term "obesity” means the condition of excess body fat (adipose tissue), including by way of example in accordance with the National Institutes of Health Federal Obesity Clinical Guidelines for adults, whereby body mass index (“BMI”) calculated by dividing body mass in kilograms by height in meters squared is equal to or greater than twenty-five (25).
  • BMI body mass index
  • BMI body mass index
  • Modifications of this classification may be used for specific ethnic groups.
  • Another alternative for assessing overweight and obesity is by measuring waist circumference.
  • Another classification is based on the recommendation from the Adult Treatment Panel III where the recommended cut-offs are 102 cm for men and 88 cm for women.
  • the methods, combinations and compositions of the invention may also be used for reduction of self-diagnosed overweight and for decreasing the risk of becoming obese due to life style, genetic considerations, heredity and/or other factors.
  • obesity-related condition refers to any disease or condition that is caused by or associated with (e.g., by biochemical or molecular association) obesity or that is caused by or associated with weight gain and/or related biological processes that precede clinical obesity.
  • obesity-related conditions include, but are not limited to, type 2 diabetes, metabolic syndrome, fatty liver disease such as NASH, hyperglycemia, hyperinsulinemia, impaired glucose tolerance, impaired fasting glucose, hyperlipidemia, hypertriglyceridemia, insulin resistance, hypercholesterolemia, atherosclerosis, coronary artery disease, peripheral vascular disease, and hypertension.
  • Syndromes with associated obesity include, without limitation, 5p13 microduplication syndrome; 16p1 1 .2 deletion; Albright hereditary osteodystrophy/PHP Type 1 a; Alstrom syndrome; Bardet Biedel syndrome (BBS); Borjeson-Forssman-Lehmann Syndrome; Carpenter syndrome; CHOPS syndrome; Chudley-Lowry syndrome; Cohen syndrome; Kabuki syndrome/Niikawa-Kuroki syndrome; Kleefstra syndrome; MORM syndrome; Prader-Willi Syndrome; Rubinstein-Taybi syndrome; Shashi-X-linked mental retardation; Smith Magenis Syndrome; WAGRO syndrome; OBHD; Ulnary Mammary syndrome; Bannayan-Riley-Ruvalcaba syndrome; Beckwith-Weidemann syndrome; Klippel-Trenaunay-Weber syndrome; Parkes Weber syndrome; Proteus syndrome; Silver-Russell syndrome; Simpson-Golabi-Behmel syndrome; Sotos syndrome; Weaver syndrome
  • Diabetes is a metabolic disease that occurs when the pancreas does not produce enough of the hormone insulin to regulate blood sugar (“type 1 diabetes mellitus”) or, alternatively, when the body cannot effectively use the insulin it produces (“type 2 diabetes mellitus”).
  • Insulin resistance occurs in 25% of non-diabetic, non-obese, apparently healthy individuals, and predisposes them to both diabetes and coronary artery disease.
  • Hyperglycemia in type II diabetes is the result of both resistance to insulin in muscle and other key insulin target tissues, and decreased beta cell insulin secretion.
  • Longitudinal studies of individuals with a strong family history of diabetes indicate that the insulin resistance precedes the secretory abnormalities. Prior to developing diabetes these individuals compensate for their insulin resistance by secreting extra insulin. Diabetes results when the compensatory hyperinsulinemia fails. The secretory deficiency of pancreatic beta cells then plays a major role in the severity of the diabetes.
  • Type II diabetes mellitus as diagnosed according to criteria published in the Report of the Expert Committee on the Diagnosis and Classification of Diabetes Mellitus whereby fasting plasma glucose level is greater than or equal to 126 milligrams per deciliter, and latent autoimmune diabetes mellitus of adults. It is characterized by insulin resistance and hyperglycemia, which in turn can cause retinopathy, nephropathy, neuropathy, or other morbidities. Additionally, diabetes is a well-known risk factor for atherosclerotic cardiovascular disease.
  • Metabolic syndrome refers to a group of factors, including hypertension, obesity, hyperlipidemia, and insulin resistance (manifesting as frank diabetes or high fasting blood glucose or impaired glucose tolerance), that raises the risk of developing heart disease, diabetes, or other health problems; (Grundy et al, Circulation. 2004; 109:433-438).
  • IFG impaired fasting glucose
  • IGT two-hour glucose levels of 140 to 199 mg/dL after a 75 gram oral glucose challenge
  • metabolic syndrome refers to metabolic disorders, particularly glucose and lipid regulatory disorders, including insulin resistance and defective secretion of insulin by pancreatic beta cells, and may further include conditions and states such as abdominal obesity, dyslipidemia, hypertension, glucose intolerance or a prothrombotic state, and which may further result in disorders such as hyperlipidemia, obesity, diabetes, insulin resistance, glucose intolerance, hyperglycemia, and hypertension.
  • subject is used interchangeably herein to refer to a mammal being assessed for treatment and/or being treated.
  • the mammal is a human.
  • subject encompass, without limitation, individuals having a disease or a condition such as obesity.
  • Subjects may be human, but also include other mammals, particularly those mammals useful as laboratory models for human disease, e.g., mice, rats, etc.
  • sample with reference to a patient encompasses blood and other liquid samples of biological origin, solid tissue samples such as a biopsy specimen or tissue cultures or cells derived therefrom and the progeny thereof.
  • sample also encompasses samples that have been manipulated in any way after their procurement, such as by treatment with reagents; washed; or enrichment for certain cell populations, such as diseased cells.
  • the definition also includes samples that have been enriched for particular types of molecules, e.g., nucleic acids, polypeptides, etc.
  • biological sample encompasses a clinical sample, and also includes tissue obtained by surgical resection, tissue obtained by biopsy, cells in culture, cell supernatants, cell lysates, tissue samples, organs, bone marrow, blood, plasma, serum, and the like.
  • a “biological sample” includes a sample obtained from a patient’s diseased cell, e.g., a sample comprising polynucleotides and/or polypeptides that is obtained from a patient’s diseased cell (e.g., a cell lysate or other cell extract comprising polynucleotides and/or polypeptides); and a sample comprising diseased cells from a patient.
  • a biological sample comprising a diseased cell from a patient can also include non-diseased cells.
  • diagnosis is used herein to refer to the identification of a molecular or pathological state, disease or condition in a subject, individual, or patient.
  • treatment refers to administering an agent, or carrying out a procedure, for the purposes of obtaining an effect on or in a subject, individual, or patient.
  • the effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of effecting a partial or complete cure for a disease and/or symptoms of the disease.
  • Treatment may include treatment of obesity in a mammal, particularly in a human, and includes achievement of weight loss, or prevention of weight gain.
  • a "therapeutically effective amount” refers to that amount of the therapeutic agent sufficient to treat or manage a disease or disorder.
  • a therapeutically effective amount may refer to the amount of therapeutic agent sufficient to delay or minimize the onset of disease.
  • a therapeutically effective amount may also refer to the amount of the therapeutic agent that provides a therapeutic benefit in the treatment or management of a disease.
  • a therapeutically effective amount with respect to a therapeutic agent of the invention means the amount of therapeutic agent alone, or in combination with other therapies, that provides a therapeutic benefit in the treatment or management of a disease.
  • a partial agonist of lepr is a variant of leptin protein, comprising one or more amino acid modifications, e.g. amino acid substitutions, that alter the binding to the receptor.
  • Partial agonists may elicit a maximal STAT3 response of from about 25% to about 75% relative to the wild-type ligand, e.g. wild-type leptin.
  • a partial agonist of interest induces STAT3- biased signaling, where maximal phosphorylation of phosphatase SHP2, ERK, or STAT1 is less than about 25% of the phosphorylation obtained with the wild-type ligand, less than about 10%, less than about 5%, less than about 1%.
  • the modifications may comprise amino acid substitutions in site 3 residues that decrease affinity.
  • Site 3 binding residues comprise, relative to the human wild-type protein, SEQ ID NO:1 , K33, Q34, K35, T37, S117, Y119, S120.
  • the combined effect of amino acid modifications at site 3 residues reduce binding to the site 3 residues, e.g. by reducing from about 2-fold to about 500- fold relative to wild-type, from about 5-fold to about 100-fold, from about 5-fold to about 50-fold, from about 5-fold to about 10-fold.
  • amino acid modifications thus generate a partial agonist of lepr.
  • amino acid modifications e.g. a substitution to an amino acid other than the wild-type; or an amino acid deletion, is provided at one, two, three, four, five or more residues selected from K33, Q34, K35, T37, S117, Y1 19, S120.
  • amino acid modifications are made at K33, including without limitation K33A, K33E, K33D, K33I, K33L, K33T, K33S, K33V.
  • amino acid modifications are made at Q34, including without limitation Q34A, Q34E, Q34D, Q34I, Q34L, Q34T, Q34S, Q34V, Q34K, Q34R.
  • amino acid modifications are made at K35; including without limitation K34A, K34E, K34D, K34I, K34L, K34T, K34S, K34V.
  • amino acid modifications are made at T37, including without limitation T37A, T37R, T37N, T37D, T37Q, T37E, T37K, T37I, T37L, T37V.
  • amino acid modifications are made at S1 17, including without limitation S117A, S1 17E, S117D, S1 17I, S1 17L, S117V, S117K, S117R, S1 17N, S117Q.
  • amino acid modifications are made at Y119, including without limitation Y1 19A, Y119E, Y1 19D, Y119I, Y1 19L, Y119V, Y1 19K, Y119R, Y1 19N, Y1 19Q, Y1 19F, Y1 19W.
  • amino acid modifications are made at S120, including without limitation S120A, S120R, S120N, S120D, S120Q, S120E, S120K, S120I, S120L, S120V.
  • amino acid modifications are made at E122, including without limitation E122A, E122R, E122K, E122I, E122L, E122T, E122S, E122V.
  • one or more amino acid modifications are made at one or more “site 2” residues, which residues are shown in FIG. 9, and are, relative to the human wild-type protein, SEQ ID NO:1 , K5, D9, L13, K15, T16, R20, N72, Q75, N82, D85, and L86.
  • a consideration for the therapeutic LepR agonists is that the serum levels of endogenous leptin are highly elevated in the context of human obesity. It is therefore desirable to include in protein modifications one or more modifications that enhance affinity of binding to site 2 residues, e.g.
  • amino acid modifications e.g. a substitution to an amino acid other than the wild-type; or an amino acid deletion, is provided at one, two, three, four, five or more residues selected from K5, D9, L13, K15, T16, R20, N72, Q75, N82, D85, and L86.
  • an amino acid modification is made at D23, including without limitation D23L.
  • Selection of amino acid modifications at binding site 2 and binding site 3 may utilize methods of mutagenesis, surface display and selection.
  • yeast surface display can be used to select for desired changes at the specified residues, by generating a library of polypeptides comprising amino acid substitutions at targeted residues, displaying the library of polypeptides on the surface of yeast cells, and selecting for desired binding characteristics. Labeling with soluble ligands enables rapid and quantitative analysis of yeast- displayed libraries by flow cytometry, while cell-surface selections allow screening of libraries with insoluble or even as-yet-uncharacterized binding targets. Affinity and specificity are key parameters governing a protein's function as a diagnostic or therapeutic agent, and yeast surface display can be applied for improving or altering these binding properties.
  • Methods are provided for treating obesity and related diseases as defined herein, wherein a therapeutically effective amount of a variant leptin polypeptide as disclosed herein, e.g. a lepr partial agonist, is administered to a mammalian, e.g., human, patient in need of treatment.
  • a therapeutically effective amount of a variant leptin polypeptide as disclosed herein e.g. a lepr partial agonist
  • the reduced food intake observed with administration of a lepr partial agonist is associated with weight loss, e.g. loss of 1 % body weight, 2.5%, 5%, 7.5%, 10%, 12.5%, 15%, 17.5%, 20%, 22.5%, 25%, 27.5%, 30% or more, depending on the initial weight of the subject.
  • Methods are also provided for treating or delaying the progression or onset of diabetes and metabolic syndrome, especially type II diabetes, including complications of diabetes, including retinopathy, neuropathy, nephropathy and delayed wound healing, and related diseases such as insulin resistance (impaired glucose homeostasis), hyperglycemia, hyperinsulinemia, elevated blood levels of fatty acids or glycerol, obesity, hyperlipidemia including hypertriglyceridemia, Syndrome X, atherosclerosis and hypertension, and for increasing high density lipoprotein levels, wherein a therapeutically effective amount of a variant leptin polypeptide as disclosed herein, e.g. a lepr partial agonist is administered to a mammalian, e.g., human, patient in need of treatment, for a period of time sufficient to effect treatment.
  • a mammalian e.g., human
  • these methods of treatment are optionally combined with one or more other types of therapeutic agent, such as an antidiabetic agent, a hypolipidemic agent or anti-obesity agent, e.g. metformin, sulfonylureas, e.g. glyburide, glipizide, glimepiride; glinides, e.g. repaglinide and nateglinide; thiazolidinediones, e.g. rosiglitazone, pioglitazone; DPP-4 inhibitors, e.g. sitagliptin, saxagliptin, linagliptin; GLP-1 receptor agonists, e.g.
  • an antidiabetic agent e.g. metformin, sulfonylureas, e.g. glyburide, glipizide, glimepiride
  • glinides e.g. repaglinide
  • exenatide, liraglutide, semaglutide; SGLT2 inhibitors, e.g. canagliflozin, dapagliflozin, empagliflozin, etc. is administered to a human patient in need of treatment.
  • SGLT2 inhibitors e.g. canagliflozin, dapagliflozin, empagliflozin, etc.
  • the other therapeutic agents when employed in combination with the compounds of the present invention may be used, for example, in those amounts indicated in the Physician's Desk Reference, as in the patents set out above or as otherwise determined by one of ordinary skill in the art.
  • a lepr partial agonist e.g. a variant leptin polypeptide as disclosed herein, is effective to induce at least "minimal weight loss" when the compound induces, over a period of time from 12 to 52 weeks, a statistically significant and placebo-adjusted decrease in mean body weight of at least about 2.5%, but less than about 5.0%, in a cohort of subjects with a baseline mean BMI > 27 kg/m 2 .
  • a lepr partial agonist e.g. a variant leptin polypeptide as disclosed herein is effective in "treatment of obesity" or “to induce weight loss” when the composition induces, over a period of time from 12 to 52 weeks, a statistically significant and placebo-adjusted decrease in body weight of at least about 5.0% in a cohort of subjects with a baseline mean BMI>27 kg/m 2 .
  • an effective dose of a variant leptin may vary according to gender and body weight, and may be comparable to the dosing for metreleptin.
  • an initial dose may be from about 0.01 mg/kg/day, about 0.025 mg/kg/day, about 0.05 mg/kg/day, about 0.1 mg/kg/day, and the dose may be up to about 500 mg/kg/day, up to about 250 mg/kg/day, up to about 100 mg/kg/day, up to about 75 mg/kg/day, up to about 50 mg/kg/day, up to about 25 mg/kg/day, up to about 10 mg/kg/day.
  • Administration may be, for example daily, or extended release formulations can be utilized to provide the same effective daily dose, by released over a period of about 1 week, 2 weeks, 3 weeks, one month, etc.
  • Efficacy of treatment for obesity can be readily determined by weight loss, for example the reduced food intake observed with administration of a lepr partial agonist is associated with weight loss, e.g. loss of 1 % body weight, 2.5%, 5%, 7.5%, 10%, 12.5%, 15%, 17.5%, 20%, 22.5%, 25%, 27.5%, 30% or more, depending on the initial weight of the subject.
  • weight loss e.g. loss of 1 % body weight, 2.5%, 5%, 7.5%, 10%, 12.5%, 15%, 17.5%, 20%, 22.5%, 25%, 27.5%, 30% or more, depending on the initial weight of the subject.
  • weight loss e.g. loss of 1 % body weight, 2.5%, 5%, 7.5%, 10%, 12.5%, 15%, 17.5%, 20%, 22.5%, 25%, 27.5%, 30% or more, depending on the initial weight of the subject.
  • an improvement in insulin sensitivity can be monitored with various methods known to the art, to determine an improvement in insulin sensitivity (or decrease in insulin resistance), where
  • Hyperinsulinemic euglycemic clamp is known to be the “gold standard” for the measurement of insulin sensitivity.
  • simplified assays can be used in quantification of insulin sensitivity.
  • insulin sensitivity indices There are two major groups of insulin sensitivity indices: (1 ) Indices calculated by using fasting plasma concentrations of insulin, glucose and triglycerides, (2) indices calculated by using plasma concentrations of insulin and glucose obtained during 120 min of a standard (75 g glucose) OGTT.
  • the former group include homeostasis model assessment-insulin resistance (HOMA-IR), QUIKI INDEX, and McAuley index while latter include, Matsuda, Belfiore, Cederholm, Avignon and Stumvoll index.
  • HOMA-IR homeostasis model assessment-insulin resistance
  • QUIKI INDEX QUIKI INDEX
  • McAuley index include, Matsuda, Belfiore, Cederholm, Avignon and Stumvoll index
  • the HEC-derived index of insulin sensitivity (ISIHEC, ml/kg/min/plU ml) is obtained during a steady state period of HEC.
  • ISIHEC MCR/I me an where, l me an - average steady state plasma insulin response (plU/ml), MCR: Metabolic clearance rate of glucose (ml/kg/min).
  • MCR Mmean/(Gmean x 0.18), where Mmear,: Metabolized glucose expressed as average steady state glucose infusion rate per kg of body weight (mg/kg/min)
  • Gmean Average steady state blood glucose concentration (mmol/l) 0.18 -conversion factor to transform blood glucose concentration from mmol/l into mg/ml.
  • HOMA is a model of the relationship of glucose and insulin dynamics that predicts fasting steady-state glucose and insulin concentrations for a wide range of possible combinations of insulin resistance and p-cell function.
  • Quantitative insulin sensitivity check index is an empirically-derived mathematical transformation of fasting blood glucose and plasma insulin concentrations that provide a consistent and precise ISI with a better positive predictive power. It is a variation of HOMA equations, as it transforms the data by taking both the logarithm and the reciprocal of the glucose-insulin product, thus slightly skewing the distribution of fasting insulin values. It employs the use of fasting values of insulin and glucose as in HOMA calculations. QUICKI is virtually identical to the simple equation form of the HOMA model in all aspects, except that a log transform of the insulin glucose product is employed to calculate QUICKI.
  • the QUICKI can be determined from fasting plasma glucose (mg/dl) and insulin (plU/ml) concentrations.
  • McAuley index is used for predicting insulin resistance in normoglycemic individuals. Regression analysis was used to estimate the cut-off points and the importance of various data for insulin resistance (fasting concentrations of insulin, triglycerides, aspartate aminotransferase, basal metabolic rate (BMI), waist circumference). A bootstrap procedure was used to find an index most strongly correlating with insulin sensitivity index, corrected for fat-free mass obtained by HEC (Mffm/I).
  • Matsuda index derives an ISI from the OGTT.
  • the OGTT ISI composite
  • the composite wholebody insulin sensitivity index is based on insulin values given in microunits per milliliter (pU/mL) and those of glucose, in milligrams per deciliter (mg/L) obtained from the OGTT and the corresponding fasting values.
  • Dosage and frequency of dosing may vary depending on the half-life of the agent in the patient. It will be understood by one of skill in the art that such guidelines will be adjusted for the molecular weight of the active agent, the clearance from the blood, the mode of administration, and other pharmacokinetic parameters.
  • the dosage may also be varied for localized administration, e.g. intranasal, inhalation, etc., or for systemic administration, e.g. i.m., i.p., i.v., oral, and the like.
  • An active agent can be administered by any suitable means, including topical, oral, parenteral, intrapulmonary, and intranasal. In some embodiments administration is subcutaneous. Parenteral infusions include intramuscular, intravenous (bolus or slow drip), intraarterial, intraperitoneal, intrathecal or subcutaneous administration. An agent can be administered in any manner which is medically acceptable. This may include injections, by parenteral routes such as intravenous, intravascular, intraarterial, subcutaneous, intramuscular, intratumor, intraperitoneal, intraventricular, intraepidural, or others as well as oral, nasal, ophthalmic, rectal, or topical. Sustained release administration is also specifically included in the disclosure, by such means as depot injections or erodible implants.
  • an agent can be formulated with an a pharmaceutically acceptable carrier (one or more organic or inorganic ingredients, natural or synthetic, with which a subject agent is combined to facilitate its application).
  • a suitable carrier includes sterile saline although other aqueous and non-aqueous isotonic sterile solutions and sterile suspensions known to be pharmaceutically acceptable are known to those of ordinary skill in the art.
  • An "effective amount” refers to that amount which is capable of ameliorating or delaying progression of the diseased, degenerative or damaged condition. An effective amount can be determined on an individual basis and will be based, in part, on consideration of the symptoms to be treated and results sought. An effective amount can be determined by one of ordinary skill in the art employing such factors and using no more than routine experimentation.
  • compositions comprising a pharmaceutically acceptable excipient.
  • the preferred form depends on the intended mode of administration and therapeutic application.
  • the compositions can also include, depending on the formulation desired, pharmaceutically-acceptable, non-toxic carriers or diluents, which are defined as vehicles commonly used to formulate pharmaceutical compositions for animal or human administration.
  • the diluent is selected so as not to affect the biological activity of the combination. Examples of such diluents are distilled water, physiological phosphate-buffered saline, Ringer's solutions, dextrose solution, and Hank's solution.
  • the pharmaceutical composition or formulation may also include other carriers, adjuvants, or nontoxic, nontherapeutic, nonimmunogenic stabilizers and the like.
  • compounds which are "commercially available” may be obtained from commercial sources including but not limited to Acros Organics (Pittsburgh PA), Aldrich Chemical (Milwaukee Wl, including Sigma Chemical and Fluka), Apin Chemicals Ltd. (Milton Park UK), Avocado Research (Lancashire U.K.), BDH Inc. (Toronto, Canada), Bionet (Cornwall, U.K.), Chemservice Inc. (West Chester PA), Crescent Chemical Co. (Hauppauge NY), Eastman Organic Chemicals, Eastman Kodak Company (Rochester NY), Fisher Scientific Co. (Pittsburgh PA), Fisons Chemicals (Leicestershire UK), Frontier Scientific (Logan UT), ICN Biomedicals, Inc.
  • the active agents of the invention and/or the compounds administered therewith are incorporated into a variety of formulations for therapeutic administration.
  • the agents are formulated into pharmaceutical compositions by combination with appropriate, pharmaceutically acceptable carriers or diluents, and are formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres, and aerosols.
  • administration of the active agents and/or other compounds can be achieved in various ways, usually by oral administration.
  • the active agents and/or other compounds may be systemic after administration or may be localized by virtue of the formulation, or by the use of an implant that acts to retain the active dose at the site of implantation.
  • the active agents and/or other compounds may be administered in the form of their pharmaceutically acceptable salts, or they may also be used alone or in appropriate association, as well as in combination with other pharmaceutically active compounds.
  • the agents may be combined, as previously described, to provide a cocktail of activities.
  • the following methods and excipients are exemplary and are not to be construed as limiting the invention.
  • Formulations are typically provided in a unit dosage form, where the term "unit dosage form,” refers to physically discrete units suitable as unitary dosages for human subjects, each unit containing a predetermined quantity of active agent in an amount calculated sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier or vehicle.
  • unit dosage form refers to physically discrete units suitable as unitary dosages for human subjects, each unit containing a predetermined quantity of active agent in an amount calculated sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier or vehicle.
  • the specifications for the unit dosage forms of the present invention depend on the particular complex employed and the effect to be achieved, and the pharmacodynamics associated with each complex in the host.
  • sustained-release as in a sustained-release form, sustained-release composition or sustained-release formulation, is intended to include a form of an active ingredient, or formulation for an active ingredient, which has an extended in vivo half-life or duration of action.
  • a sustained-release form may result from modification of the active ingredient, such as modifications that extend circulation residence time, decrease rates of degradation, decrease rates of clearance or the like, or may result from formulations or compositions which provide for extended release of the active ingredient, such as use of various liposomes, emulsions, micelles, matrices and the like.
  • a controlled-release form or formulation is a type of sustained-release form or formulation.
  • a unit dose is at least about 0.1 mg/kg, at least about 0.5 mg/kg, at least about 1 mg/kg, at least about 5 mg/kg, at least about 10 mg/kg, at least about 20 mg/kg, at least about 50 mg/kg, at least about 100 mg/kg, in some embodiments the effective dose is from about 1 to 50 mg/kg.
  • Dosing may be daily, every 2 days, every 3 or more days, e.g. weekly, semi-weekly, bi-weekly, monthly, etc. Dosing may be parenteral, including sustained release formulations. Dosing may be maintained for long periods of time, e.g. months, or years, to maintain desirable glucose and fatty acid levels.
  • the pharmaceutically acceptable excipients such as vehicles, adjuvants, carriers or diluents, are commercially available.
  • pharmaceutically acceptable auxiliary substances such as pH adjusting and buffering agents, tonicity adjusting agents, stabilizers, wetting agents and the like, are commercially available.
  • Any compound useful in the methods and compositions of the invention can be provided as a pharmaceutically acceptable base addition salt.
  • “Pharmaceutically acceptable base addition salt” refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid.
  • Salts derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like.
  • Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts.
  • Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine, N-ethylpiperidine, polyamine resins and the like.
  • Particularly preferred organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline and caffeine.
  • the agents can be used alone or in combination with appropriate additives to make tablets, powders, granules or capsules, for example, with conventional additives, such as lactose, mannitol, corn starch or potato starch; with binders, such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators, such as corn starch, potato starch or sodium carboxymethylcellulose; with lubricants, such as talc or magnesium stearate; and if desired, with diluents, buffering agents, moistening agents, preservatives and flavoring agents.
  • conventional additives such as lactose, mannitol, corn starch or potato starch
  • binders such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins
  • disintegrators such as corn starch, potato starch or sodium carboxymethylcellulose
  • lubricants such as talc or magnesium stearate
  • compositions can also include large, slowly metabolized macromolecules such as proteins, polysaccharides such as chitosan, polylactic acids, polyglycolic acids and copolymers (such as latex functionalized SepharoseTM, agarose, cellulose, and the like), polymeric amino acids, amino acid copolymers, and lipid aggregates (such as oil droplets or liposomes).
  • macromolecules such as proteins, polysaccharides such as chitosan, polylactic acids, polyglycolic acids and copolymers (such as latex functionalized SepharoseTM, agarose, cellulose, and the like), polymeric amino acids, amino acid copolymers, and lipid aggregates (such as oil droplets or liposomes).
  • a carrier may bear the agents in a variety of ways, including covalent bonding either directly or via a linker group, and non-covalent associations.
  • Suitable covalent-bond carriers include proteins such as albumins, peptides, and polysaccharides such as aminodextran, each of which have multiple sites for the attachment of moieties.
  • the nature of the carrier can be either soluble or insoluble for purposes of the invention.
  • Acceptable carriers, excipients, or stabilizers are non-toxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyidimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, his
  • the active ingredients may also be entrapped in microcapsule prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsule and poly-(methylmethacylate) microcapsule, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nanoparticles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nanoparticles and nanocapsules
  • compositions can be prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection can also be prepared.
  • the preparation also can be emulsified or encapsulated in liposomes or micro particles such as polylactide, polyglycolide, or copolymer for enhanced adjuvant effect, as discussed above. Langer, Science 249: 1527, 1990 and Hanes, Advanced Drug Delivery Reviews 28: 97- 119, 1997.
  • the agents of this invention can be administered in the form of a depot injection or implant preparation which can be formulated in such a manner as to permit a sustained or pulsatile release of the active ingredient.
  • the pharmaceutical compositions are generally formulated as sterile, substantially isotonic and in full compliance with all Good Manufacturing Practice (GMP) regulations of the U.S. Food and Drug Administration.
  • GMP Good Manufacturing Practice
  • Toxicity of the active agents can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., by determining the LD50 (the dose lethal to 50% of the population) or the LD100 (the dose lethal to 100% of the population). The dose ratio between toxic and therapeutic effect is the therapeutic index.
  • the data obtained from these cell culture assays and animal studies can be used in further optimizing and/or defining a therapeutic dosage range and/or a sub-therapeutic dosage range (e.g., for use in humans). The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition.
  • the term “dosing regimen” refers to a set of unit doses (typically more than one) that are administered individually to a subject, typically separated by periods of time.
  • a given therapeutic agent has a recommended dosing regimen, which may involve one or more doses.
  • a dosing regimen comprises a plurality of doses each of which are separated from one another by a time period of the same length; in some embodiments, a dosing regimen comprises a plurality of doses and at least two different time periods separating individual doses.
  • all doses within a dosing regimen are of the same unit dose amount. In some embodiments, different doses within a dosing regimen are of different amounts.
  • a dosing regimen comprises a first dose in a first dose amount, followed by one or more additional doses in a second dose amount different from the first dose amount. In some embodiments, a dosing regimen comprises a first dose in a first dose amount, followed by one or more additional doses in a second dose amount same as the first dose amount. In some embodiments, a dosing regimen is correlated with a desired or beneficial outcome when administered across a relevant population (i.e., is a therapeutic dosing regimen).
  • each component can be administered at the same time or sequentially in any order at different points in time. Thus, each component can be administered separately but sufficiently closely in time so as to provide the desired therapeutic effect.
  • Concomitant administration means administration of one or more components, such as engineered proteins and cells, known therapeutic agents, etc. at such time that the combination will have a therapeutic effect. Such concomitant administration may involve concurrent (i.e. at the same time), prior, or subsequent administration of components. A person of ordinary skill in the art would have no difficulty determining the appropriate timing, sequence and dosages of administration.
  • a first prophylactic or therapeutic agent can be administered prior to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subsequent to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of a second prophylactic or therapeutic agent to a subject with a disorder.
  • Suitable anti-diabetic agents for use in combination with the compounds of the present invention include biguanides (e.g., metformin or phenformin), glucosidase inhibitors (e.g,. acarbose or miglitol), insulins (including insulin secretagogues or insulin sensitizers), meglitinides (e.g., repaglinide), sulfonylureas (e.g., glimepiride, glyburide, gliclazide, chlorpropamide and glipizide), biguanide/glyburide combinations (e.g., Glucovance.RTM.), thiazolidinediones (e.g., troglitazone, rosiglitazone and pioglitazone), PPAR-alpha agonists, PPAR-gamma agonists, PPAR alpha/gamma dual agonists, glycogen phosphorylase inhibitors
  • Thiazolidinediones include Mitsubishi's MCC-555 (disclosed in U.S. Pat. No. 5,594,016), Glaxo-Welcome's GL-262570, englitazone (CP-68722, Pfizer) or darglitazone (CP-86325, Pfizer, isaglitazone (MIT/J&J), JTT-501 (JPNT/P&U), L-895645 (Merck), R-119702 (Sankyo/WL), NN- 2344 (Dr. Reddy/NN), or YM-440 (Yamanouchi).
  • Suitable PPAR alpha/gamma dual agonists include AR-HO39242 (Astra/Zeneca), GW- 409544 (Glaxo-Wellcome), KRP297 (Kyorin Merck) as well as those disclosed by Murakami et al, "A Novel Insulin Sensitizer Acts As a Coligand for Peroxisome Proliferation-Activated Receptor Alpha (PPAR alpha) and PPAR gamma. Effect on PPAR alpha Activation on Abnormal Lipid Metabolism in Liver of Zucker Fatty Rats", Diabetes 47, 1841 -1847 (1998), and in U.S. application Ser. No. 09/644,598, filed Sep. 18, 2000, employing dosages as set out therein, which compounds designated as preferred are preferred for use herein.
  • Suitable aP2 inhibitors include those disclosed in U.S. application Ser. No. 09/391 ,053, filed Sep. 7, 1999, and in U.S. application Ser. No. 09/519,079, filed Mar. 6, 2000, employing dosages as set out therein.
  • Suitable DPP4 inhibitors that may be used in combination with the compounds of the invention include those disclosed in WO99/38501 , WO99/46272, WO99/67279 (PROBIODRUG), WO99/67278 (PROBIODRUG), WO99/61431 (PROBIODRUG), NVP-DPP728A (1 -[[[2-[(5- cyanopyridin-2-yl)amino]ethyl]amino]acetyl]-2-cyano-(S)-pyrro- Udine) (Novartis) as disclosed by Hughes et al, Biochemistry, 38 (36), 1 1597-1 1603, 1999, TSL-225 (tryptophyl-1 , 2,3,4- tetrahydroisoquinoline-3-carboxylic acid (disclosed by Yamada et al, Bioorg.
  • Suitable meglitinides include nateglinide (Novartis) or KAD1229 (PF/Kissei).
  • GLP-1 glucagon-like peptide-1
  • examples of other suitable glucagon-like peptide-1 (GLP-1 ,) compounds that may be used in combination with the GLP-1 mimics of the present invention include GLP-1 (1 -36) amide, GLP-1 (7-36) amide, GLP-1 (7-37) (as disclosed in U.S. Pat. No. 5,614,492 to Habener), as well as AC2993 (Amylin), LY-315902 (Lilly) and NN-2211 (NovoNordisk).
  • hypolipidemic/lipid lowering agents for use in combination with the compounds of the present invention include one or more MTP inhibitors, HMG CoA reductase inhibitors, squalene synthetase inhibitors, fibric acid derivatives, ACAT inhibitors, lipoxygenase inhibitors, cholesterol absorption inhibitors, ileal Na.sup.+/bile acid cotransporter inhibitors, upregulators of LDL receptor activity, bile acid sequestrants, cholesterol ester transfer protein inhibitors (e.g., CP-529414 (Pfizer)) and/or nicotinic acid and derivatives thereof.
  • MTP inhibitors HMG CoA reductase inhibitors
  • squalene synthetase inhibitors fibric acid derivatives
  • ACAT inhibitors lipoxygenase inhibitors
  • cholesterol absorption inhibitors ileal Na.sup.+/bile acid cotransporter inhibitors
  • upregulators of LDL receptor activity e.g., CP
  • MTP inhibitors which may be employed as described above include those disclosed in U.S. Pat. Nos. 5,595,872, 5,739,135, 5,712,279, 5,760,246, 5,827,875, 5,885,983 and 5,962,440.
  • the HMG CoA reductase inhibitors which may be employed in combination with one or more compounds of SEQ ID NO:X include mevastatin and related compounds, as disclosed in U.S. Pat. No. 3,983,140, lovastatin (mevinolin) and related compounds, as disclosed in U.S. Pat. No. 4,231 ,938, pravastatin and related compounds, such as disclosed in U.S. Pat. No. 4,346,227, simvastatin and related compounds, as disclosed in U.S. Pat. Nos. 4,448,784 and 4,450,171.
  • Other HMG CoA reductase inhibitors which may be employed herein include, but are not limited to, fluvastatin, disclosed in U.S. Pat. No.
  • Hypolipidemic agents include pravastatin, lovastatin, simvastatin, atorvastatin, fluvastatin, cerivastatin, atavastatin and ZD-4522.
  • phosphinic acid compounds useful in inhibiting HMG CoA reductase such as those disclosed in GB 2205837, are suitable for use in combination with the compounds of the present invention.
  • Squalene synthetase inhibitors suitable for use herein include, but are not limited to, a- phosphono-sulfonates disclosed in U.S. Pat. No. 5,712,396, those disclosed by Biller et al, J. Med. Chem., 1988, Vol. 31 , No. 10, pp 1869-1871 , including isoprenoid (phosphinyl- methyl)phosphonates, as well as other known squalene synthetase inhibitors, for example, as disclosed in U.S. Pat. Nos. 4,871 ,721 and 4,924,024 and in Biller, S. A., Neuenschwander, K., Ponpipom, M.
  • squalene synthetase inhibitors suitable for use herein include the terpenoid pyrophosphates disclosed by P. Ortiz de Montellano et al, J. Med. Chem., 1977, 20, 243-249, the farnesyl diphosphate analog A and presqualene pyrophosphate (PSQ-PP) analogs as disclosed by Corey and Volante, J. Am. Chem. Soc., 1976, 98, 1291 -1293, phosphinylphosphonates reported by McClard, R. W. et al, J.A.C.S., 1987, 109, 5544 and cyclopropanes.
  • the fibric acid derivatives which may be employed in combination with one or more compounds of SEQ ID NO:X include fenofibrate, gemfibrozil, clofibrate, bezafibrate, ciprofibrate, clinofibrate and the like, probucol, and related compounds, as disclosed in U.S. Pat. No.
  • bile acid sequestrants such as cholestyramine, colestipol and DEAE-Sephadex (Secholex.RTM., policexide.RTM.), as well as lipostabil (Rhone-Poulenc), Eisai E-5050 (an N-substituted ethanolamine derivative), imanixil (HOE-402), tetrahydrolipstatin (THL), istigmastanylphos-phorylcholine (SPG, Roche), aminocyclodextrin (Tanabe Seiyoku), Ajinomoto AJ-814 (azulene derivative), melinamide (Sumitomo), Sandoz 58-035, American Cyanamid CL-277,082 and CL-283,546 (disubstituted urea derivatives), nicotinic acid, acipimox, acifran, neomycin, p-aminosal
  • the ACAT inhibitor which may be employed in combination with one or more compounds of SEQ ID NO:X include those disclosed in Drugs of the Future 24, 9-15 (1999), (Avasimibe); "The ACAT inhibitor, CI-1011 is effective in the prevention and regression of aortic fatty streak area in hamsters", Nicolosi et al, Atherosclerosis (Shannon, Irel). (1998), 137 (1 ), 77-85; "The pharmacological profile of FCE 27677: a novel ACAT inhibitor with potent hypolipidemic activity mediated by selective suppression of the hepatic secretion of ApoBI OO-containing lipoprotein", Ghiselli, Giancarlo, Cardiovasc. Drug Rev.
  • ACAT inhibitors physiologic mechanisms for hypolipidemic and anti-atherosclerotic activities in experimental animals, Krause et al, Editor(s): Ruffolo, Robert R., Jr.; Hollinger, Mannfred A., Inflammation: Mediators Pathways (1995), 173-98, Publisher: CRC, Boca Raton, Fla.; "ACAT inhibitors: potential anti-atherosclerotic agents", Sliskovic et al, Curr. Med. Chem. (1994), 1 (3), 204-25; "Inhibitors of acyl-CoA:cholesterol O-acyl transferase (ACAT) as hypocholesterolemic agents. 6.
  • the first water-soluble ACAT inhibitor with lipid-regulating activity Inhibitors of acyl-CoA:cholesterol acyltransferase (ACAT). 7. Development of a series of substituted N-phenyl-N'-[(1 -phenylcyclopentyl)methyl]ureas with enhanced hypocholesterolemic activity", Stout et al, Chemtracts: Org. Chem. (1995), 8 (6), 359-62, or TS-962 (Taisho Pharmaceutical Co. Ltd).
  • the hypolipidemic agent may be an upregulator of LD2 receptor activity, such as MD-700 (Taisho Pharmaceutical Co. Ltd) and LY295427 (Eli Lilly).
  • suitable cholesterol absorption inhibitor for use in combination with the compounds of the invention include SCH48461 (Schering-Plough), as well as those disclosed in Atherosclerosis 1 15, 45-63 (1995) and J. Med. Chem. 41 , 973 (1998).
  • ileal NaVbile acid cotransporter inhibitors for use in combination with the compounds of the invention include compounds as disclosed in Drugs of the Future, 24, 425- 430 (1999).
  • the lipoxygenase inhibitors which may be employed in combination with one or more compounds of SEQ ID NO:X include 15-lipoxygenase (15-LO) inhibitors, such as benzimidazole derivatives, as disclosed in WO 97/12615, 15-LO inhibitors, as disclosed in WO 97/12613, isothiazolones, as disclosed in WO 96/38144, and 15-LO inhibitors, as disclosed by Sendobry et al "Attenuation of diet-induced atherosclerosis in rabbits with a highly selective 15-lipoxygenase inhibitor lacking significant antioxidant properties”, Brit. J. Pharmacology (1997) 120, 1199-1206, and Cornicelli et al, ”15-Lipoxygenase and its Inhibition: A Novel Therapeutic Target for Vascular Disease”, Current Pharmaceutical Design, 1999, 5, 1 1 -20.
  • 15-LO 15-lipoxygenase
  • 15-LO 15-lipoxygenase
  • Suitable anti-hypertensive agents for use in combination with the compounds of the present invention include beta adrenergic blockers, calcium channel blockers (L-type and T-type; e.g. diltiazem, verapamil, nifedipine, amlodipine and mybefradil), diuretics (e.g., chlorothiazide, hydrochlorothiazide, flumethiazide, hydroflumethiazide, bendroflumethiazide, methylchlorothiazide, trichloromethiazide, polythiazide, benzthiazide, ethacrynic acid tricrynafen, chlorthalidone, furosemide, musolimine, bumetanide, triamtrenene, amiloride, spironolactone), renin inhibitors, ACE inhibitors (e.g., captopril, zofenopril,
  • Dual ET/AII antagonist e.g., compounds disclosed in WO 00/01389
  • neutral endopeptidase (NEP) inhibitors neutral endopeptidase (NEP) inhibitors
  • vasopepsidase inhibitors dual NEP-ACE inhibitors
  • omapatrilat and gemopatrilat e.g., omapatrilat and gemopatrilat
  • Suitable anti-obesity agents for use in combination with the compounds of the present invention include a NPY receptor antagonist, a MCH antagonist, a GHSR antagonist, a CRH antagonist, a beta 3 adrenergic agonist, a lipase inhibitor, a serotonin (and dopamine) reuptake inhibitor, a thyroid receptor beta drug and/or an anorectic agent.
  • the beta 3 adrenergic agonists which may be optionally employed in combination with compounds of the present invention include AJ9677 (Takeda/Dainippon), L750355 (Merck), or CP331648 (Pfizer,) or other known beta 3 agonists, as disclosed in U.S. Pat. Nos. 5,541 ,204, 5,770,615, 5,491 ,134, 5,776,983 and 5,488,064, with AJ9677, L750,355 and CP331648 being preferred.
  • lipase inhibitors which may be optionally employed in combination with compounds of the present invention include orlistat or ATL-962 (Alizyme), with orlistat being preferred.
  • Leptin is an adipocyte-derived protein hormone that promotes satiety and energy homeostasis by activating the leptin receptor (LepR)-STAT3 signaling axis in a subset of hypothalamic neurons.
  • Leptin signaling is dysregulated in obesity, however, where appetite remains elevated despite high levels of circulating leptin, due to leptin resistance.
  • therapeutic use of leptin as an anti-obesity medication has been limited to rare cases of leptin deficiency.
  • the structure reveals an asymmetric complex architecture in which a single leptin induces LepR dimerization via two distinct receptor-binding sites.
  • Analysis of the leptin-LepR binding interfaces reveals the molecular basis for human obesity-associated mutations in both leptin and LepR.
  • Structure-based design of leptin mutants that destabilize the LepR dimer yielded both partial and biased agonists that decouple stimulation of STAT3 from activation of LepR negative regulators, even in the presence of wild-type leptin. Together, these results reveal the structural basis for leptin signaling and provide therapeutic LepR agonists that overcome leptin resistance in the context of obesity.
  • the low affinity site 3 interface by contrast is formed by the immunoglobulin (Ig) domain of LepR (D3), which interacts with the top of leptin helix D and loop AB.
  • Ig immunoglobulin
  • D3 canonical “site 1 ” interface, which participates in high affinity receptor binding in most cytokine receptor complexes, is unoccupied for both leptin molecules in the leptin-LepR complex (Fig. 1d).
  • the membrane distal CHR1 domains (D1 and D2) bend upwards, resulting in a highly elongated structure with a total estimated length of approximately 180 A projecting from the cell surface (Fig. 1 c,d).
  • the two- membrane proximal fibronectin domains (FNIII, D6 and D7) of LepR bend inwards towards one another, forming an approximately 90° angle between D6 and D7 which is predicted to bring to TM domains of each LepR monomer in close association.
  • FNIII, D6 and D7 the two- membrane proximal fibronectin domains of LepR bend inwards towards one another, forming an approximately 90° angle between D6 and D7 which is predicted to bring to TM domains of each LepR monomer in close association.
  • receptor dimerization is ligand-dependent, as is the case for other class 1 cytokine receptors.
  • the Ig domains of both gp130 subunits bend back in to engage the adjacent IL-6 molecules, forming two site 3 interactions in a symmetric, closed receptor conformation (Fig. 2b).
  • the Ig domain of only a single LepR engages leptin at site 3, whereas the Ig domain of the second LepR projects outward, away from the second bound leptin which is unoccupied at site 3 (Fig. 2a).
  • the architecture of the leptin-bound LepR complex more closely resembles heterodimeric IL-6 family receptor complexes, such as IL-27, in which a single ligand dimerizes two different receptors, gp130 and IL27Ra, to form a similarly open and asymmetric receptor complex (Fig. 2c).
  • the Ig domain of LepR engages the top of helix D and the CD loop of leptin (Fig. 3, D and E), burying 775 A 2 .
  • the primary contact appears to be mediated by Tyr1 19 within the CD loop of leptin, which inserts into an aromatic cluster consisting of mouse LepR residues Phe403, Tyr405, Tyr409, and Tyr420 (“site 3a,” Fig. 3e).
  • a second site 3 contact with LepR is formed by the leptin AB loop, which engages the LepR Ig domain via a primarily backbone-mediated p-sheet interaction formed between LepR residues 415-417 and leptin residues 35-37 (“site 3b”, Fig. 3g).
  • site 3b primarily backbone-mediated p-sheet interaction formed between LepR residues 415-417 and leptin residues 35-37
  • site 3b primarily backbone-mediated p-sheet interaction formed between LepR residues 415-417 and leptin residues 35-37
  • site 3b primarily backbone-mediated p-sheet interaction formed between LepR residues 415-417 and leptin residues 35-37
  • site 3b primarily backbone-mediated p-sheet interaction formed between LepR residues 415-417 and leptin residues 35-37
  • the side chains of neighboring leptin residues Gln34 and Lys33 appear also form additional contacts with Cy
  • Leptin-mediated LepR dimerization is primarily driven by the aromatic and polar contacts formed by leptin residues Tyr1 19 and S120 at site 3a, with the binding-induced conformational change of the leptin AB loop playing a secondary role to further stabilize the LepR dimer and enable maximal STAT3 activation.
  • mice expressing a Y1138S LepR mutant are extremely obese and hyperphagic due to loss of leptin- mediated STAT3 activation.
  • mice expressing a Y985F LepR mutant are lean and do not become leptin resistant, phenocopying loss of hypothalamic SOCS3 expression.
  • these partial agonists also induced STAT3-biased signaling, with minimal phosphorylation of the phosphatase SHP2 or its downstream effector ERK when compared to WT leptin, suggesting reduced phosphorylation of LepR Y98S relative to LepR Y1141 (Fig. 4c).
  • LepR agonists An important consideration for the development of therapeutic LepR agonists is that the serum levels of endogenous leptin are highly elevated in the context of human obesity, ranging from 5 ng/ml (0.3 nM) in lean individuals to as high as 100 ng/ml (6 nM) in obesity. Therefore, we next assessed how the leptin partial agonists KQK-AAA and S1 17N impacted LepR signaling in the presence of high levels of WT leptin. To enhance the ability of these variants to compete with WT leptin for LepR binding, we incorporated an additional mutation at site 2, D23L, which reduces the off rate of leptin for LepR (Fig. 4d).
  • cryo-EM structures support a two-step mechanism of LepR activation in which leptin first binds LepR via the high affinity site 2 interface to form a 1 :1 complex on the cell membrane. Subsequent trans-interaction between leptin and the LepR Ig domain forms the site 3 interface, which dimerizes LepR to form signaling competent 2:2 complexes.
  • our cryo-EM data reveals that a partially open, asymmetric 2:2 complex in which only one leptin forms a site 3 contact with LepR is the most stable dimer conformation in solution.
  • LepR complexes that can form only one site 3 interaction exhibit full signaling activity in cells, suggesting that engagement of a single leptin at site 3 is sufficient for receptor activation.
  • the asymmetric receptor homodimerization observed here is reminiscent of the asymmetry seen in some homodimeric receptor tyrosine kinases, including Insulin Receptor and Epithelial growth factor receptor (EGFR), but is unique among known cytokine receptor complexes.
  • EGFR Epithelial growth factor receptor
  • mouse LepR D1 D7 (M. musculus, residues 23-839) and LepR D3 D7 (M musculus, residues 330-839) were cloned into a pD649 mammalian expression vector containing an N-terminal HA signal peptide, C-terminal GCN4 leucine zipper (EELLSKNYHLENEVARLKK), and C-terminal 6xHis-tag.
  • DNA was transiently transfected into Expi-293F cells (Thermo) using Expifectamine transfection reagent (Thermo).
  • Expi293F cells were grown in serum free Expi293 expression media (Thermo) and maintained at 37°C with 5% CO2 with gentle agitation. 72 hours after transfection, cell supernatant was harvested and proteins were purified with Ni-NTA resin (Qiagen) followed by size-exclusion chromatography (SEC) on a Superdex 200 column (GE) in Phosphate-buffered saline (PBS, 135 mM NaCI, 2.5 mM KOI, 8.0 mM Na 2 HPO 4 , 30 mM KH 2 PO 4 , pH 7.2).
  • PBS Phosphate-buffered saline
  • wild-type or mutant leptin (/-/. sapiens, residues 23-167) was cloned into a pET28a E. coli expression vector containing a C-terminal 6xHis-tag.
  • DNA was transformed into BL21 (DE3) competent cells and grown at 37°C in LB media supplemented with Kanamycin (40 pg/mL) until the culture reached log-phase growth.
  • Protein expression was induced by adding IPTG at a final concentration of 1 mM. The culture was shaken at 37°C for 4 hours. Cells were harvested by centrifugation at 6000xg for 6 minutes, and frozen at -20°C.
  • Inclusion bodies were washed 3x in Wash Buffer 1 (50 mM T ris-HCI pH 8.0, 0.5% T ritonX-100, 100 mM NaCI, 1 mM Na EDTA, 1 mM DTT) and 1 x in Wash Buffer 2 (50 mM Tris-HCI pH 8.0, 100 mM NaCI, 1 mM Na EDTA, 1 mM DTT), with centrifugation at 10,000xg for 15 min between each wash. Inclusion bodies were then solubilized by rotating in Denaturing Buffer (20 mM Tris-HCI pH 8.0, 8 M Urea, 1 mM DTT) at room temperature for 24 hours. Solubilized samples were then centrifuged at 30,000xg for 30 minutes, and supernatant was frozen at -80 for further processing.
  • Wash Buffer 1 50 mM T ris-HCI pH 8.0, 0.5% T ritonX-100, 100 mM NaCI, 1 mM Na
  • an energy filter was used during imaging of the LepR D1 D7 complex. Movies were collected at a calibrated magnification corresponding to 0.653 A and 0.8521 A per physical pixel, for LepR D1 D7 and LepR D3 D7 complexes, respectively. The dose was set to a total of 53 electrons per A 2 over an exposure of 1 .518 and 2.545 seconds, for LepR D1 D7 and LepR D3 D7 complexes, respectively. Automated data collection was carried out using SerialEM with a nominal defocus range set from 0.8 to 2.0 pM. 1 1 ,292 movies were collected for the complex with LepR D1 D7 and 21 ,1 12 movies were collected for the complex with LepR D3 D7 .
  • Cryo-EM Data processing and 3D reconstruction All movies were processed using cryoSPARC v3.1 .0 unless otherwise specified. Movies were motion corrected, had contrast transfer functions (CTFs) determined, and particles picked using the cryoSPARC live processing functions. During this processing, micrographs were binned to the physical pixel size.
  • CTFs contrast transfer functions
  • Lentiviruses were produced by transfection of HEK-293T cells with pLV-EF1 a-IRES-Puro vector containing an N-terminal Myc- tag in combination with the pMD2G envelope and psPax2 packaging plasmids. Forty-eight hours after transfection, the virus-containing supernatant was collected and centrifuged for 5 minutes at 300xg to remove cell debris. Virus was concentrated by incubation with 1 x PEG-IT (SBI) at 4° C for 12-24 hours. The solution was then centrifuged a 1 ,500xg for 30 min and virus pellet was resuspended with 10% of the initial virus volume in serum free DMEM.
  • SBI 1 x PEG-IT
  • Target cells were plated in 6-well plates containing DMEM supplemented with 10% v/v fetal bovine serum, penicillinstreptomycin, 1 mM sodium pyruvate, 10 nM HEPES and 2 mM GlutaMAXTM (Gibco). Concentrated virus was added to the media together with 8 pg/mL polybrene. Forty-eight hours later, the media was changed to fresh media containing puromycin for selection.
  • HEK-293T cells were plated in six-well culture dishes coated with fibronectin (Millipore) at 0.7x10 6 cells per well in DMEM (Dulbecco’s Modified Eagle Medium) supplemented with 10% v/v fetal bovine serum, penicillinstreptomycin, 1 mM sodium pyruvate, 10 nM HEPES and 2 mM GlutaMAXTM (Gibco). Forty-eight hours later, cells were transfected using FuGene 6 (Promega) with pD649 vector containing HA- or FLAG-tagged full-length human LepR, WT or mutant.
  • DMEM Dulbecco’s Modified Eagle Medium
  • HEK-293T cells stably expressing Myc-tagged full-length human LepR were plated in six-well culture dishes coated with fibronectin (Millipore) at 1 x10 6 cells per well in DMEM (Dulbecco’s Modified Eagle Medium) supplemented with 10% v/v fetal bovine serum, penicillin-streptomycin, 1 mM sodium pyruvate, 10 nM HEPES and 2 mM GlutaMAXTM (Gibco). Twenty-Four hours later, cells were treated with recombinant human Leptin for 20 minutes at 37°C.
  • DMEM Dulbecco’s Modified Eagle Medium
  • Antibodies used for immunoblots were obtained from Cell Signaling Technologies and include: Phospho-STAT3 (Y705, Antibody #9131 ), Phospho-SHP2 (Y542, Antibody #3751 ), Phospho- ERK1/2 (T202/Y204, Antibody #9101 ), STAT3 (clone 79D7), SHP2 (Antibody #3752), ERK1/2 (clone 137F5), HA (clone C29F4).
  • HEK-293T cells stably expressing Myc- tagged full-length human LepR were plated in 96-well plates and stimulated with WT or mutant Leptin for 20 min at 37°C, followed by fixation with paraformaldehyde (Electron Microscopy Sciences) for 10 min at room temperature.
  • the cells were permeabilized for intracellular staining by treatment with ice cold methanol (Fisher) for 30 min at -20°C.
  • the cells were then incubated with Alexa Fluor 647 conjugated Anti-Stat3 (pY705) antibody (1 :100, BD, clone 4/P-STAT3) and anti-c-Myc-Alexa Fluor 488 (1 :100, CST, clone 9B1 1 ) for 1 hour at room temperature in autoMACS buffer (Miltenyi). Data was acquired using CytoFlex, flow cytometer instrument (Beckman Coulter). The MFI values were background subtracted and represented as a percent of the maximal WT Leptin value within each experiment and plotted in Prism 8 (GraphPad). The dose-response curves were generated using the “sigmoidal dose-response” analysis.
  • HEK-293T cells stably expressing Myc-tagged full-length human LepR were treated with PBS or Leptin variants for 5 hours.
  • Cells were isolated and lysed using Qiashredder columns (Qiagen) per the manufacturer’s instructions.
  • RNA was isolated using RNAeasy plus mini kit (Qiagen) per the manufacturer’s instructions. 1 pg RNA for each sample was used for cDNA generation with iScript Reverse Transcription Supermix (BioRad). Relative gene expression was measured by SYBR-green based qPCR using the comparative AC t method and normalized to GAPDH expression. All samples were run in triplicate.
  • mice qPCR primers were obtained from IDT: GAPDH: (5’GGA AAC TTG CTG TGG GTG A3’, 5’CAA GGA CGG AGA CTT CGA TTC3’), SOCS3: (5TGT AGT TGA GGT CAA TGA AGG G3’, 5’ACA TCG CTC AGA CAC CAT G3’).
  • REGION D4 (SEQ ID NO:7) DVNINISCETDGYLTKMTCRWSTSTIQSLAESTLQLRYHRSSLYCSDIPSIHPISEPKDCYLQSD
  • tissue protective functions of interleukin-22 can be decoupled from pro-inflammatory actions through structure-based design.

Landscapes

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

Abstract

L'invention concerne des compositions et des procédés pour la prévention ou le traitement de l'obésité et/ou du surpoids, et la gestion du poids corporel. Les compositions comprennent des agonistes partiels du récepteur de leptine (lepr), y compris des variants de polypeptides de leptine, les agonistes partiels déclenchant une signalisation sous-maximale à des concentrations de ligand de saturation, et sollicitant la réponse à la signalisation STAT3. Des variants de protéine de leptine humaine, par exemple la leptine modifiée pour augmenter l'affinité au niveau du ou des sites de fixation de site 2 et diminuer la liaison au niveau du ou des sites de fixation de site 3, suppriment de préférence la phosphorylation de SHP2, ERK et STAT1 par rapport à STAT3, entraînant un signal polarisé qui active sélectivement des voies associées à la satiété, avec une activation réduite des voies associées à la résistance à la leptine. Les protéines variantes trouvent une utilisation dans la suppression de l'appétit, et peuvent compenser la perte de poids thérapeutique.
PCT/US2023/072937 2022-08-31 2023-08-25 Analogues de leptine pour le traitement de l'obésité et la gestion du poids WO2024050285A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263374149P 2022-08-31 2022-08-31
US63/374,149 2022-08-31

Publications (2)

Publication Number Publication Date
WO2024050285A2 true WO2024050285A2 (fr) 2024-03-07
WO2024050285A3 WO2024050285A3 (fr) 2024-04-18

Family

ID=90098746

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/072937 WO2024050285A2 (fr) 2022-08-31 2023-08-25 Analogues de leptine pour le traitement de l'obésité et la gestion du poids

Country Status (1)

Country Link
WO (1) WO2024050285A2 (fr)

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7307142B2 (en) * 2004-11-26 2007-12-11 Yissum Research And Development Company Of The Hebrew University Of Jerusalem Leptin antagonists
US20120108503A1 (en) * 2008-08-20 2012-05-03 The Board Of Regents Of The University Of Texas System Methods for treating type i diabetes with leptin and leptin agonists
BR112012026718A2 (pt) * 2010-04-22 2017-12-19 Medical Res Infrastructure & Health Services Fund Tel Aviv Medical Ct antagonista de leptina sintético, molécula de dna isolada, composição farmacêutica, método para o tratamento de uma doença ou condição, rato transgênico e método de varredura de uma substância tendo atividade terapêutica para uma doença ou distúrbio.

Also Published As

Publication number Publication date
WO2024050285A3 (fr) 2024-04-18

Similar Documents

Publication Publication Date Title
US20190192630A1 (en) Mutated fibroblast growth factor (fgf) 1 and methods of use
US20100041612A1 (en) Fragments of the Glucagon-Like Peptide-1 and Uses Thereof
US7238670B2 (en) Human glucagon-like-peptide-1 mimics and their use in the treatment of diabetes and related conditions
CN104662038B (zh) 胰高血糖素类似物
US7960349B2 (en) N-terminally modified GLP-1 receptor modulators
RU2733720C2 (ru) Модуляторы активности комплемента
US7238671B2 (en) Human glucagon-like-peptide-1 mimics and their use in the treatment of diabetes and related conditions
AU2002348469A1 (en) Human glucagon-like-peptide-1 mimics and their use in the treatment of diabetes and related conditions
MXPA06015193A (es) Moduladores del peptido 1 similar al glucagon humano y su uso en el tratamiento de la diabetes y condiciones relacionadas.
US20200040051A1 (en) Fibroblast growth factor (fgf) 1 with mutation in the heparin binding domain and methods of use to reduce blood glucose
US20070238669A1 (en) Human glucagon-like-peptide-1 modulators and their use in the treatment of diabetes related conditions
JP2024056838A (ja) 治療用途のためのヒトcペプチドとヒトエラスチン受容体との相互作用のペプチドモジュレーター
JP2022532332A (ja) 代謝性疾患を治療するための融合タンパク質
TW201348249A (zh) 多胜肽、編碼該多胜肽之核酸分子、以及該多胜肽之應用
WO2024050285A2 (fr) Analogues de leptine pour le traitement de l'obésité et la gestion du poids
JP2015514773A (ja) ナトリウムチャネル感受性コノペプチドおよび類似体を含む組成物ならびにその方法
WO2024030214A2 (fr) Compositions peptidiques dérivées de brinp2 pour le traitement de l'obésité et de la gestion du poids
Brandt Synthesis and characterization of insulin receptor partial agonists as a route to improved diabetes therapy
CN111788217A (zh) 白介素-15活性的拮抗剂肽
CN108676071A (zh) 一种抗Aβ蛋白聚集的七肽及其应用与编码该合成多肽的基因
AU2014202625B2 (en) Hepatic insulin sensitizing substance and test meal for insulin sensitization
JP2004182683A (ja) 新規な炎症性疾患改善剤
JP2022522461A (ja) 代謝性障害の処置のための組換えタンパク質の使用
Taylor et al. Yang3, Joost Oppenheim3, Dusan Uhrin2, John RW Govan4, Dominic J. Campopiano2, Derek MacMillan 5, Perdita E. Barran 2 and Julia R. Dorin MRC Human Genetics Unit, Edinburgh EH4 2XU, Scotland, UK 2School of Chemistry, University of Edinburgh, Edinburgh EH9 3JJ, UK

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 23861456

Country of ref document: EP

Kind code of ref document: A2