US20210363213A1 - Glucagon-glp-1-gip triple agonist compounds - Google Patents

Glucagon-glp-1-gip triple agonist compounds Download PDF

Info

Publication number
US20210363213A1
US20210363213A1 US17/388,328 US202117388328A US2021363213A1 US 20210363213 A1 US20210363213 A1 US 20210363213A1 US 202117388328 A US202117388328 A US 202117388328A US 2021363213 A1 US2021363213 A1 US 2021363213A1
Authority
US
United States
Prior art keywords
aib
peg3
isoglu
ser
hexadecanoyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US17/388,328
Inventor
Rasmus Just
Ditte Riber
Anne Pernille Tofteng SHELTON
Torben Østerlund
Kate HANSEN
Lene JESSEN
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Zealand Pharma AS
Original Assignee
Zealand Pharma AS
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 Zealand Pharma AS filed Critical Zealand Pharma AS
Priority to US17/388,328 priority Critical patent/US20210363213A1/en
Publication of US20210363213A1 publication Critical patent/US20210363213A1/en
Assigned to ZOOLANDER SA LLC reassignment ZOOLANDER SA LLC PATENT SECURITY AGREEMENT Assignors: ZEALAND PHARMA A/S
Assigned to ZEALAND PHARMA A/S reassignment ZEALAND PHARMA A/S ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: JESSEN, Lene, RIBER, DITTE, ØSTERLUND, Torben, SHELTON, Anne Pernille Tofteng, HANSEN, Kate, JUST, Rasmus
Assigned to ZEALAND PHARMA A/S reassignment ZEALAND PHARMA A/S RELEASE BY SECURED PARTY (SEE DOCUMENT FOR DETAILS). Assignors: ZOOLANDER SA LLC
Assigned to ZEALAND PHARMA A/S reassignment ZEALAND PHARMA A/S RELEASE BY SECURED PARTY (SEE DOCUMENT FOR DETAILS). Assignors: ZOOLANDER SA LLC
Abandoned legal-status Critical Current

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
    • C07K14/575Hormones
    • C07K14/605Glucagons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/26Glucagons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/48Drugs for disorders of the endocrine system of the pancreatic hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the invention relates to compounds having agonist activity at the glucagon, GIP and GLP-1 receptors, and to their use in the treatment of metabolic disorders.
  • CVD cardiovascular diseases
  • obstructive sleep apnea obstructive sleep apnea
  • stroke peripheral artery disease
  • microvascular complications CAD
  • osteoarthritis CAD
  • CVD cardiovascular diseases
  • Many have additional cardiovascular risk factors including high/aberrant LDL and triglycerides and low HDL.
  • Cardiovascular disease accounts for about 50% of the mortality in people with diabetes, and the morbidity and mortality rates relating to obesity and diabetes underscore the medical need for efficacious treatment options.
  • Pre-proglucagon is a 158 amino acid precursor polypeptide that is processed in different tissues to form a number of different proglucagon-derived peptides, including glucagon, glucagon-like peptide-1 (GLP-1 or GLP1), glucagon-like peptide-2 (GLP-2) and oxyntomodulin (OXM), which are involved in a wide variety of physiological functions, including glucose homeostasis, insulin secretion, gastric emptying, and intestinal growth, as well as the regulation of food intake.
  • GLP-1 or GLP1 glucagon-like peptide-1
  • GLP-2 glucagon-like peptide-2
  • OXM oxyntomodulin
  • Glucagon is a 29-amino acid peptide that corresponds to amino acids 33 through 61 of pre-proglucagon, while GLP-1 is produced as a 37-amino acid peptide that corresponds to amino acids 72 through 108 of pre-proglucagon.
  • glucagon a hormone produced by the pancreas, signals the liver to break down glycogen and release glucose, causing blood glucose levels to rise toward a normal level.
  • glucagon reduces body weight probably through inhibition of food intake and stimulation of energy expenditure and/or lipolysis.
  • GLP-1 has different biological activities compared to glucagon. Its actions include stimulation of insulin synthesis and secretion, inhibition of glucagon secretion, and inhibition of food intake. GLP-1 has been shown to reduce hyperglycemia (elevated glucose levels) in diabetic patients.
  • Exendin-4 a peptide from lizard venom that shares about 50% amino acid identity with GLP-1, activates the GLP-1 receptor and likewise has been shown to reduce hyperglycemia in diabetic patients.
  • Glucose-dependent insulinotropic polypeptide is a 42-amino acid gastrointestinal regulatory peptide that, like GLP-1, stimulates insulin secretion from pancreatic ⁇ (beta) cells in the presence of elevated blood glucose levels. It is derived by proteolytic processing from a 133-amino acid precursor, preproGlP.
  • glucagon-GLP-1 dual acting receptor agonists are currently in pre-clinical development (see, e.g., WO2011/006497).
  • glucagon-GLP-1 dual agonists are associated with more profound and sustained body weight loss in animal models on top of the improvements in glycemic control.
  • glucagon based drugs may have promise for the treatment of type 2 diabetes mellitus and/or obesity.
  • Incretins are gastrointestinal hormones that regulate blood glucose by enhancing glucose-stimulated insulin secretion (Drucker, D J and Nauck, M A, Lancet 368: 1696-705 (2006)).
  • Two of the above mentioned peptides are known as incretins: GLP-1 and GIP.
  • the discovery of the incretins has led to the development of two new classes of drugs for the treatment of diabetes mellitus.
  • GLP-1 receptor agonists and small molecule compounds (oral DPP-4 inhibitors) that inhibit enzymatic inactivation of both endogenous GLP-1 and GIP
  • GLP-1 receptor agonists ByettaTM, BydureonTM LixisenatideTM and LiraglutideTM
  • DPP-4 inhibitors JanuviaTM GalvusTM OnglyzaTM and TrajentaTM
  • the two peptides also have long term effects.
  • GLP-1 R agonists protect pancreatic ⁇ -cells by inhibiting apoptosis and enhancing proliferation. For instance, the study by Farilla et al.
  • GLP-1 had anti-apoptotic effects in human islets (Farilla, L, Endocrinology 144: 5149-58 (2003)). Such effects have not been reported for GIP until recently.
  • Weidenmaier et al. reported that a DPP-4 resistant GIP analogue has anti-apoptotic effects (Weidenmaier, S D, PLOS One 5(3): e9590 (2010)).
  • the combination of the GLP-1 receptor agonist Liraglutide and an acylated GIP analogue show superior effects compared to treatment with Liraglutide or GIP analogue alone (Gault, V A, Clinical Science 121: 107-117 (2011)).
  • glucagon By combining glucagon, GLP-1 and GIP receptor agonism in novel inventive peptides it is anticipated that superior glycemic control and body weight loss can be achieved.
  • Such peptides are likely to have strong incretin actions and improved ⁇ -cell preservation from the GLP-1 and GIP components, and have improved body weight loss from all three components by stimulating energy expenditure, lipolysis and reducing food intake.
  • the present invention concerns Glucagon-GLP-1-GIP triple agonists (referred to in this specification as “triple agonists”) which comprise one or more substitutions as compared to wild-type glucagon and which may have the property of an altered, preferably increased GIP and GLP-1 receptor activity, e.g. as assessed in in vitro efficacy assays.
  • triple agonists Glucagon-GLP-1-GIP triple acting receptor agonists are superior to existing and marketed GLP-1 analogues because the triple agonists offer improved glycemic control, possible islet and ⁇ -cell preservation and enhanced body weight loss.
  • the Glucagon-GLP-1-GIP triple agonists could be used as therapeutics for both type 2 diabetes mellitus, obesity and related disorders.
  • the invention provides a triple agonist having the general formula I:
  • R 1 is H— (i.e., hydrogen), C 1-4 alkyl, acetyl, formyl, benzoyl, trifluoroacetyl or pGlu;
  • X2 is Aib, Gly, Ala, D-Ala, Ser, N-Me-Ser, Ac3c, Ac4c or Ac5c;
  • X10 is Tyr or Leu
  • X12 is Lys, Ile or ⁇
  • X13 is Ala, Tyr or Aib
  • X15 is Asp or Glu
  • X16 is Ser, Glu, Lys or ⁇
  • X17 is Lys or ⁇
  • X19 is Gln or Ala
  • X20 is Lys, His, Arg or ⁇ ;
  • X21 is Ala, Asp or Glu
  • X23 is Val or Ile
  • X24 is Asn, Glu or ⁇ ;
  • X27 is Leu, Glu or Val
  • X28 is Ala, Ser, Arg or ⁇ ;
  • X29 is Aib, Ala, Gln or Lys
  • X30 is Lys, Gly, or is absent
  • Y1 is (SEQ ID NO: 53) Gly-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser, (SEQ ID NO: 54) Gly-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Ser, (SEQ ID NO: 55) Lys-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser, (SEQ ID NO: 56) Lys-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Ser, (SEQ ID NO: 57) Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser or (SEQ ID NO: 58) Pro-Ser-Ser-Gly-Ala-Pro-Pro-Ser, or is absent;
  • is a residue of Lys, Arg, Orn or Cys in which the side chain is conjugated to a lipophilic substituent
  • R 2 is —NH 2 or —OH
  • is present at one of positions X12, X16 and X17.
  • the compound contains only one residue ⁇ , which may be present at any one of positions X12, X16, X17, X20, X24 or X28. For example, it may be present at one of X12, X16 and X17.
  • the compound may possess one or more of the following sets of residues:
  • L27, R28, and A29 L27, S28, and A29; L27, A28, and Q29; E27, S28, and A29; or V27, and A28, and Aib29;
  • Positions 1 to 29 may have the sequence
  • positions 1 to 29 may differ at up to 4 positions, e.g. at 1, 2, 3 or 4 positions, from any of the specific sequences shown above, within the constraints of Formula I.
  • positions 1 to 29 may differ at up to 4 positions, e.g. at 1, 2, 3 or 4 positions from one of the following sequences:
  • Positions 1 to 29 may have the sequence:
  • the peptide backbone of Formula I may have the sequence:
  • the peptide backbone sequence may differ at up to 5 positions from one of the sequences shown above, within the constraints of Formula I.
  • a sequence satisfying the definition of Y1 is regarded as a single position.
  • the compound differs from the reference sequence at only 4 positions in X1 to X29.
  • one of those positions is generally X30 or Y1.
  • the peptide backbone sequence may differ at up to 5 positions from one of the sequences:
  • the peptide backbone of Formula I may have the sequence:
  • Certain of the Y1 groups when present, may provide increased stability in vivo, e.g. in serum, and so may contribute to the half life of the GIP analogue. Without wishing to be bound by theory, it is believed that these groups may help to stabilize the three dimensional conformation of the molecule and/or provide resistance to proteolytic degradation.
  • the Y1 sequences Gly-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser (SEQ ID NO:53), Gly-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Ser (SEQ ID NO: 54), Lys-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser (SEQ ID NO: 55), Lys-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Ser (SEQ ID NO: 56), Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser (SEQ ID NO: 57) and Pro-Ser-Ser-Gly-Ala-Pro-Pro-Ser (SEQ ID NO: 58) have homology with a C-terminal portion of the Exendin-4 molecule and appear to contribute to the stability of the molecule without concomitantly providing significant levels of GLP-1 agonist activity.
  • the invention further provides a nucleic acid encoding a peptide having the sequence X1-X30 of Formula I.
  • an expression construct also known as an expression vector
  • the invention also provides a host cell comprising a nucleic acid or expression construct and capable of expressing, and optionally secreting, the peptide.
  • the peptide may itself be a compound of the invention, e.g. when the peptide contains only naturally occurring amino acids (i.e. proteinogenic amino acids), does not contain a residue ⁇ , and where R 1 and R 2 are H— and —OH respectively.
  • the peptide may be a precursor of a compound of the invention.
  • the invention further provides a pharmaceutical composition
  • a pharmaceutical composition comprising a triple agonist as described herein, or a pharmaceutically acceptable salt or solvate thereof, in admixture with a carrier, preferably a pharmaceutically acceptable carrier.
  • the triple agonist may, for example, be a pharmaceutically acceptable acid addition salt.
  • the pharmaceutical composition may be formulated as a liquid suitable for administration by injection or infusion, or which is formulated to cause slow release of said triple agonist.
  • the invention further provides a therapeutic kit comprising a triple agonist as described herein, and a device comprising a triple agonist as described herein.
  • the invention further provides a triple agonist as described herein, or a pharmaceutically acceptable salt or solvate thereof, for use in a method of medical treatment, e.g. for use in the treatment and/or prevention of a metabolic disorder.
  • the invention further provides the use of a triple agonist as described herein, or a pharmaceutically acceptable salt or solvate thereof, in the preparation of a medicament for the treatment and/or prevention of a metabolic disorder.
  • the invention further provides a method of prevention and or/treatment of a metabolic disorder in a subject, comprising administering a triple agonist as described herein, or a pharmaceutically acceptable salt or solvate thereof, to the subject.
  • the metabolic disorder may be diabetes or a diabetes related disorder, or obesity or an obesity related disorder.
  • diabetes or a diabetes related disorder, or obesity or an obesity related disorder.
  • the link between obesity and diabetes is well known, so these conditions are not necessarily separate or mutually exclusive.
  • Diabetes related disorders include insulin resistance, glucose intolerance, increased fasting glucose, pre-diabetes, type 1 diabetes, type 2 diabetes, gestational diabetes hypertension, dyslipidemia, bone related disorders and combinations thereof.
  • Diabetes related disorders also include atherosclerosis, arteriosclerosis, coronary heart disease, peripheral artery disease and stroke; or conditions associated with atherogenic dyslipidemia, blood fat disorders, elevated blood pressure, hypertension, a prothrombotic state and a proinflammatory state.
  • Bone related disorders include, but are not limited to osteoporosis and increased risk of bone fracture.
  • the blood fat disorder may be selected from high triglycerides, low HDL cholesterol, high LDL cholesterol, and plaque buildup in artery walls, or a combination thereof.
  • the prothrombotic state may be selected from high fibrinogen levels in the blood and high plasminogen activator inhibitor-1 levels in the blood.
  • the proinflammatory state may be an elevated C-reactive protein level in the blood.
  • Obesity related disorders include obesity linked inflammation, obesity linked gallbladder disease and obesity induced sleep apnea, or may be associated with a condition selected from atherogenic dyslipidemia, blood fat disorders, elevated blood pressure, hypertension, a prothrombotic state, and a proinflammatory state, or a combination thereof.
  • patient may be used interchangeably and refer to either a human or a non-human animal.
  • mammals such as humans, primates, livestock animals (e.g., bovines, porcines), companion animals (e.g., canines, felines) and rodents (e.g., mice and rats).
  • livestock animals e.g., bovines, porcines
  • companion animals e.g., canines, felines
  • rodents e.g., mice and rats.
  • solvate in the context of the present invention refers to a complex of defined stoichiometry formed between a solute (in casu, a peptide conjugate or pharmaceutically acceptable salt thereof according to the invention) and a solvent.
  • the solvent in this connection may, for example, be water, ethanol or another pharmaceutically acceptable, typically small-molecular organic species, such as, but not limited to, acetic acid or lactic acid.
  • a solvate is normally referred to as a hydrate.
  • agonist refers to a substance (ligand) that activates the receptor type in question.
  • sequences disclosed herein are sequences incorporating an “H—” moiety at the amino terminus (N-terminus) of the sequence, and either an “—OH” moiety or an “—NH 2 ” moiety at the carboxy terminus (C-terminus) of the sequence.
  • an “H—” moiety at the N-terminus of the sequence in question indicates a hydrogen atom (i.e. R 1 ⁇ H—), corresponding to the presence of a free primary or secondary amino group at the N-terminus
  • an “—OH” or an “—NH 2 ” moiety at the C-terminus of the sequence i.e. R 2 ⁇ —OH or —NH 2
  • R 1 groups are possible at the N-terminus, including pyroglutamic acid (pGlu; (S)-( ⁇ )-2-pyrrolidone-5-carboxylic acid), C 1-4 alkyl, acetyl, formyl, benzoyl and trifluoroacetyl.
  • pGlu pyroglutamic acid
  • S S-( ⁇ )-2-pyrrolidone-5-carboxylic acid
  • C 1-4 alkyl acetyl, formyl, benzoyl and trifluoroacetyl.
  • the compounds described herein are Glucagon-GIP-GLP 1 dual receptor agonists. That is to say, they have agonist activity at all three of the glucagon receptor, the GIP receptor and the GLP-1 receptor.
  • agonist refers to a substance (ligand) that is capable of binding to a particular receptor and activating signaling by that receptor.
  • a GIP receptor agonist is capable of binding to the GIP receptor (designated GIP-R) and activating signaling by that receptor, e.g. by generation of cAMP or inducing Ca 2+ release.
  • Agonist activity at the GIP receptor may therefore be measured by assessing GIP receptor signalling, which may, for example, be measured via cAMP production or Ca 2+ release.
  • the cDNA sequence encoding the human GIP receptor has GenBank accession no. BC101673.1 (GI:75516688).
  • the encoded amino acid sequence (including signal peptide) is:
  • the compounds have agonist activity at the GLP-1 receptor (GLP-1-R), i.e. they are capable of binding to the GLP-1 receptor and activating signaling by that receptor, e.g. by generation of cAMP or inducing Ca 2+ release.
  • Agonist activity at the GLP-1 receptor may therefore be measured by assessing GLP-1 receptor signalling, which may, for example, be measured via cAMP production or Ca 2+ release.
  • the GLP-1 receptor may have the sequence of the human glucagon-like peptide 1 receptor (GLP-1 R) having primary accession number P43220.
  • the precursor protein (including signal peptide) has primary accession number NP_002053.3; GI:166795283 and has sequence:
  • the compounds have agonist activity at the glucagon receptor (Glu-R), i.e. they are capable of binding to the glucagon receptor and activating signaling by that receptor, e.g. by generation of cAMP or inducing Ca 2+ release.
  • Agonist activity at the glucagon receptor may therefore be measured by assessing glucagon receptor signalling, which may, for example, be measured via cAMP production or Ca 2+ release.
  • the glucagon receptor may have the sequence of the human glucagon receptor (Glu-R) having primary accession number P47871.
  • the precursor protein (including signal peptide) has primary accession number NP_000151.1; GI:4503947, and has the sequence:
  • the compounds of the present invention have at least one GIP, one glucagon, and one GLP-1 biological activity, in particular in treatment of metabolic diseases such as diabetes and obesity. This can be assessed, e.g., in in vivo assays, for example as described in the examples, in which the blood glucose level or another biological activity is determined after a test animal has been treated or exposed to a triple agonist.
  • compounds of the invention may be capable of improving glycaemic control when adminstered to a diabetic subject. Additionally or alternatively, they may be capable of reducing body weight when administered to an overweight or obese subject. In either case, the effect may be superior to that obtained with an equivalent quantity (by mass, or molar ratio) of wild type human GIP or GLP-1 in comparable subjects when given according to a comparable dosing regime.
  • Activity in in vitro assays may also be used as a measure of the compounds' activity.
  • the compounds typically have activity at the glucagon, GLP-1 and GIP receptors (designated GCG-R, GLP-1-R and GIP-R respectively).
  • EC 50 values may be used as a numerical measure of agonist potency at a given receptor.
  • An EC 50 value is a measure of the concentration of a compound required to achieve half of that compound's maximal activity in a particular assay.
  • a compound having EC 50 [GLP-1R] lower than the EC 50 [GLP-1 R] of native GIP in a particular assay may be considered to have higher potency at the GLP-1R than GIP.
  • the EC 50 GLP-1-R and/or EC 50 GIP-R and/or EC 50 GCG-R is below 1.0 nM, below 0.9 nM, below 0.8 nM, below 0.7 nM, below 0.6 nM, below 0.5 nM, below 0.4 nM, below 0.3 nM, below 0.2 nM, below 0.1 nM, below 0.09 nM, below 0.08 nM, below 0.07 nM, below 0.06 nM, below 0.05 nM, below 0.04 nM, below 0.03 nM, below 0.02 nM, below 0.01 nM, below 0.009 nM, below 0.008 nM, below 0.007 nM, below 0.006 nM, or below 0.005 nM, e.g. when assessed using the assay described in Example 2.
  • the compound of the invention may comprise a residue ⁇ , i.e. a residue selected from Lys, Arg, Orn and Cys in which the side chain is conjugated to a lipohilic substituent.
  • the substituent binds plasma proteins (e.g. albumin) in the blood stream, thus shielding the compounds of the invention from enzymatic degradation and renal clearance and thereby enhancing the half-life of the compounds. It may also modulate the potency of the compound, e.g. with respect to the GIP receptor, the glucagon receptor and/or the GLP-1 receptor.
  • plasma proteins e.g. albumin
  • the substituent is conjugated to the functional group at the distal end of the side chain from the alpha-carbon.
  • the normal ability of the Lys, Arg, Orn or Cys side chain to participate in interactions mediated by that functional group may therefore be reduced or completely eliminated by the presence of the substituent.
  • the overall properties of the compound may be relatively insensitive to changes in the actual amino acid present as residue ⁇ . Consequently, it is believed that any of the residues Lys, Arg, Orn and Cys may be present at any position where ⁇ is permitted.
  • it may be advantageous that the amino acid component of ⁇ is Lys.
  • is a residue of Lys, Arg, Orn or Cys in which the side chain is conjugated to a substituent having the formula —Z 1 or —Z 2 —Z 1 .
  • —Z 1 is a fatty chain having at a terminus a connection —X— to ⁇ or to Z 2 ;
  • —X— is a bond, —CO—, —SO—, or —SO 2 —;
  • Z 1 has a polar group at the end of the chain distal from connection —X—; said polar group comprising a carboxylic acid or a carboxylic acid bioisostere, a phosphonic acid, or a sulfonic acid group;
  • each Y is independently —NH, —NR, —S or —O, where R is alkyl, a protecting group or forms a linkage to another part of the spacer Z 2 ;
  • each X is independently a bond, CO—, SO—, or SO 2 —;
  • each V is independently a bivalent organic moiety linking Y and X;
  • n 1-10.
  • Z 1 is a fatty chain having a connection to ⁇ or to Z 2 , referred to herein as —X—.
  • —X— may be, for example, a bond, acyl (—CO—), sulfinyl (—SO—), or sulfonyl (—SO 2 —).
  • —X— is acyl (—CO—), sulfinyl (—SO—), or sulfonyl (—SO 2 —).
  • —X— is acyl (—CO—).
  • Z 1 may further have a polar group, said polar group being located at the end of the chain distal from the connection —X—.
  • the connection is located at the ⁇ -position with respect to the polar group.
  • the polar group may be bound directly to the terminus of the fatty chain, or may be bound via a linker.
  • the polar group is an acidic or weakly acid group, for example a carboxylic acid or a carboxylic acid bioisostere, a phosphonate, or a sulfonate.
  • the polar group may have a pK a of between ⁇ 2 and 12 in water, more preferably between 1 and 7, more preferably between 3 and 6.
  • Certain preferred polar groups have a pK a of between 4 and 5.
  • the polar group may comprise a carboxylic acid (—COOH) or a carboxylic acid bioisostere, a phosphonic acid (—P(O)(OH) 2 ), or a sulfonic acid (—SO 2 OH) group.
  • the polar group if present, comprises a carboxylic acid or carboxylic acid bioisostere.
  • Suitable carboxylic acid bioisosteres are known in the art.
  • the bioisostere has a proton having a pK a similar to the corresponding carboxylic acid.
  • suitable bioisoteres may include, not by way of limitation, tetrazole, acylsulfomides, acylhydroxylamine, and squaric acid derivatives, as shown below (--- indicates the point of attachment):
  • Fatty chain refers to a moiety comprising a chain of carbon atoms, the carbon atoms being predominantly substituted with hydrogen or hydrogen-like atoms, for example, a hydrocarbon chain.
  • Such fatty chains are often referred to as lipophilic, although it will be appreciated that substitution may alter the lipophilic properties of the overall molecule.
  • the fatty chain may by aliphatic. It may be entirely saturated or may include one or more double or triple bonds. Each double bond, if present, may be in the E or Z configuration.
  • the fatty chain may also have one or more cycloalkylene or heterocycloalkylene moieties in its length, and additionally or alternatively may have one or more arylene or heteroarylene moieties in its length.
  • the fatty chain may incorporate a phenylene or piperazinylene moiety in its length as, for example, shown below (wherein --- represents the points of attachment within the chain).
  • the fatty chain may be derived from a fatty acid, for example, it may be derived from a medium-chain fatty acid (MCFA) with an aliphatic tail of 6-12 carbon atoms, a long-chain fatty acid (LCFA) with an aliphatic tail of 13-21 carbon atoms, or a very long-chain fatty acid (LCFA) with an aliphatic tail of 22 carbon atoms or more.
  • MCFA medium-chain fatty acid
  • LCFA long-chain fatty acid
  • LCFA very long-chain fatty acid
  • linear saturated fatty acids from which suitable fatty chains may be derived include tridecylic (tridecanoic) acid, myristic (tetradecanoic) acid, pentadecylic (pentadecanoic) acid, palmitic (hexadecanoic) acid, and margaric (heptadecanoic) acid.
  • linear unsaturated fatty acids from which suitable fatty chains may be derived include myristoleic acid, palmitoleic acid, sapienic acid and oleic acid.
  • the fatty chain may be connected to ⁇ or to Z 2 by an amide linkage, a sulfinamide linkage, a sulfonamide linkage, or by an ester linkage, or by an ether, thioether or amine linkage. Accordingly, the fatty chain may have, a bond to ⁇ or to Z 2 or an acyl (—CO—), sulfinyl (—SO—), or sulfonyl (—SO 2 —) group.
  • the fatty chain has a terminus having an acyl (—CO—) group and is connected to ⁇ or Z 2 by an amide or ester linkage.
  • Z 1 is a group of formula:
  • A is hydrogen or a carboxylic acid, a carboxylic acid bioisostere, a phosphonic acid, or a sulfonic acid group;
  • B is a bond or a linker
  • X is a bond, acyl (—CO—), sulfinyl (—SO—), or sulfonyl (—SO 2 —);
  • Alk is a fatty chain that may be optionally substituted with one or more substituents.
  • the fatty chain is preferably 6 to 28 carbon atoms in length (e.g. a C 6-28 alkylene), more preferably, 12 to 26 carbons in length (e.g. a C 12-26 alkylene), more preferably, 16 to 22 carbons in length (e.g. C 16-22 alkylene), and may be saturated or unsaturated.
  • Alk is saturated, that is, preferably Alk is alkylene.
  • Optional substituents on the fatty chain may be independently selected from fluoro, C 1-4 alkyl, preferably methyl; trifluoromethyl, hydroxymethyl, amino, hydroxyl, C 1-4 alkoxy, preferably methoxy; oxo, and carboxyl, and may be independently located at any point along the chain.
  • each optional substituent is selected from fluoro, methyl, and hydroxyl. Where more than one substituent is present, substituents may be the same or different.
  • the number of substituents is 0 to 3; more preferably the fatty chain is unsubstituted.
  • B may be a bond or a linker.
  • B When B is a linker, it may be a cycloalkylene, heterocycloalkylene, C 6 arylene, or C 5-6 heteroarylene, or C 6 arylene-O— or C 5-6 heteroarylene-O—.
  • B When B is phenylene it may, for example, be selected from 1,2-phenylene, 1,3-phenylene, 1,4-phenylene, preferably 1,4-phenylene (so that A-B— is a 4-benzoic acid substituent or 4-benzoic acid bioisostere).
  • B When B is phenylene-O—, it may, for example, be selected from 1,2-phenylene-O—, 1,3-phenylene-O—, 1,4-phenylene-O—, preferably 1,4-phenylene-O.
  • Each phenylene of B may be optionally substituted with one or more substituents selected from fluoro, methyl, trifluoromethyl, amino, hydroxyl, and C 1-4 alkoxy, preferably methoxy.
  • B may be C 5-6 heteroarylene, for example, pyridinylene or thiofuranylene, and may be optionally substituted as described.
  • A-B— may be selected from:
  • A-B-Alk- is an alkyl chain of formula H 3 C—(CH 2 ) n —.
  • Z 1 is an acyl group of formula:
  • Z 1 is an acyl group of formula:
  • A is —COOH and B is a bond.
  • certain preferred Z 1 are derived from long-chain saturated ⁇ , ⁇ -dicarboxylic acids of formula HOOC—(CH 2 ) 12-22 —COOH, preferably, long-chain saturated ⁇ , ⁇ -dicarboxylic acids having an even number of carbon atoms in the aliphatic chain.
  • A is H and B is a bond.
  • certain preferred Z 1 are derived from long-chain saturated carboxylic acids of formula HOOC—(CH 2 ) 12-22 —CH 3 , preferably, long-chain saturated carboxylic acids having an even number of carbon atoms in the aliphatic chain.
  • Z 1 may be:
  • the carboxylic acid group if present, may be replaced by a bioisotere as detailed herein.
  • Z 2 is an optional spacer that connects Z 1 to the side chain of the amino acid component of ⁇ .
  • Z 2 if present, is a spacer bound at one terminus by Y, which may be a nitrogen, oxygen or sulfur atom, and at the other terminus by X, which may be a bond or an acyl (—CO—), sulfinyl (—SO—), sulfonyl (—SO 2 —) or absent.
  • Z 2 may be a spacer of formula (--- indicate points of attachment):
  • Y may be —NH, —NR, —S or —O, where R may be alkyl, a protecting group or may form a linkage to another part of the spacer, with the remaining valency forming a linkage to Z 1 ;
  • X may be a bond, CO—, SO—, or SO 2 —, with the remaining valency forming a linkage to the side chain of the amino acid component of Y;
  • V is a bivalent organic moiety linking Y and X;
  • n may be 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10. Where n is 2 or more, each Y, V, and X is independent of every other Y, V, and X.
  • Z 2 may be bound at each side by amide, sulfinamide, sulfonamide, or ester linkages or by amino, ether, or thioether linkages depending upon the nature of Y and X and the corresponding linking groups on Z 1 and the side chain.
  • each V may also be bound to each adjacent V by linkages as described.
  • linkages are amides, esters or sulfonamides, most preferably amides.
  • each Y is —NH or —NR and each X is CO— or SO 2 —.
  • —X— is acyl (—CO—).
  • Z 2 is a spacer of formula —S A —, —S B —, —S A —S B — or —S B —S A —, wherein S A and S B are as defined below.
  • Z 2 is selected from —S A — or —S B —S A —, that is, [side chain]—Z 2
  • Z 1 is [side chain]—S A —Z 1 or [side chain]—S B —S A —Z 1 .
  • S A may be a single amino acid residue or a residue of an amino acid derivative, especially an amino acid derivative residue having a sulfinyl or sulfonyl in place of the carboxy moiety at the C terminus. Additionally or alternatively, the single amino acid residue may have an oxygen or sulfur atom in place of the nitrogen atom at the N terminus.
  • S A is a single amino acid residue.
  • the amino acid may be selected from ⁇ -Glu, ⁇ -Glu, ⁇ -Asp, ⁇ -Asp, Ala, ⁇ -Ala (3-aminopropanoic acid), Dapa (2,3-diaminopropanoic acid), Dab (2,4-diaminobutanoic acid), and Gaba (4-aminobutanoic acid).
  • connection may be at any moiety as appropropriate. Any carboxylic acid or amino resides not bound within the residue may be free, that is, present as a free carboxylic acid or primary amine, or may be derivatised. Suitable derivatisation is known in the art.
  • carboxylic acid moieties may be present in S A amino acid residues as esters, for example, as methyl esters.
  • Amino moieties may be present as alkylated amines, for example, methylated, or may be protected as amide or carbamate moieties.
  • Other suitable amino acids include ⁇ -Ala (3-aminopropanoic acid) and Gaba (4-aminobutanoic acid) and similar w amino acids.
  • amino acids may be D or L, or a racemic or enantioenriched mixture.
  • the amino acid is an L-amino acid.
  • the amino acid is a D-amino acid.
  • S A has a carboxylic acid substituent, with ⁇ -Glu, ⁇ -Glu, ⁇ -Asp, and ⁇ -Asp, and sulfinyl and sulfonyl derivatives thereof, being preferred. Accordingly, in some embodiments, the amino acid residue is:
  • —X— is —CO—, —SO—, —SO 2 —, preferably —CO—, and a is 1 or 2, preferably 2.
  • the carboxylic acid is an ester, and the amino acid residue is:
  • —X— is —CO—, —SO—, —SO 2 —, preferably —CO—, and a is 1 or 2, preferably 2, and R is C 1-4 alkyl or C 6 aryl.
  • R is C 1-4 alkyl, preferably methyl or ethyl, more preferably ethyl.
  • a preferred S A group bearing a carboxylic acid is ⁇ -Glu.
  • S A is selected from Dapa or ⁇ -Glu. Most preferably, S A is ⁇ -Glu.
  • S B may be a linker of general formula:
  • each polymeric unit P U may be bound at each side by amide, sulfinamide, sulfonamide, or ester linkages or by amino, ether, or thioether linkages depending upon the nature of Y and X and the corresponding linking groups on Z 1 , S A , and Lys.
  • each P U may be independently a unit of formula:
  • Y may be —NH, —NR, —S or —O, wherein R may be alkyl, a protecting group or may form a linkage to another part of the spacer, with the remaining valency forming a linkage to Z 1 ;
  • X may be a bond, CO—, SO—, or SO 2 —, with the remaining valency forming a linkage to the ⁇ side chain;
  • V is a bivalent organic moiety linking Y and X.
  • V is the ⁇ -carbon of a natural or unnatural amino acid, that is V is —CHR AA —, wherein R AA is an amino acid side chain; or V is an optionally substituted C 1-6 alkylene, or V is a chain comprising one or more units of ethylene glycol in series, also known as PEG chain, for example, —CH 2 CH 2 —(OCH 2 CH 2 ) m —O—(CH 2 ) p —, where m is 0, 1, 2, 3, 4, or 5, and p is 1, 2, 3, 4, or 5; when X is CO—, p is preferably 1, 3, 4, or 5.
  • Optional alkylene substituents include fluoro, methyl, hydroxy, hydroxymethy, and amino.
  • Preferred P U units include:
  • S B may comprise one or more of each of P U i , P U ii , and P U ii in any order, or may comprise one or more units of P U i , P U ii , and P U i only, or one of more units selected from P U i and P U ii , P U i and P U iii , or P U ii and P U iii .
  • Each P U i may be independently selected from any natural or unnatural amino acid residue and, for example, may be selected from Gly, Pro, Ala, Val, Leu, lie, Met, Cys, Phe, Tyr, Trp, His, Lys, Arg, Gln, Asn, ⁇ -Glu, ⁇ -Glu, Asp, Ser Thr, Dapa, Gaba, Aib, ⁇ -Ala, 5-aminopentanoyl, 6-aminohexanoyl, 7-aminoheptanoyl, 8-aminooctanoyl, 9-aminononanoyl, and 10-aminodecanoyl.
  • P U i amino acid residues are selected from Gly, Ser, Ala, Thr, and Cys, more preferably from Gly and Ser.
  • S B is —(P U i ) n —, wherein n is 1 to 8, more preferably 5 to 7, most preferably 6. In some preferred embodiments, S B is —(P U i ) n —, n is 6 and each P U i is independently selected from Gly or Ser, with a preferred sequence being -Gly-Ser-Gly-Ser-Gly-Gly- (SEQ ID NO: 114).
  • P U ii dipeptide residues Each P U i may be independently selected from any dipeptide residue comprising two natural or unnatural amino acid residues bound by an amide linkage.
  • Preferred P U ii dipeptide residues include Gly-Gly, Gly-Ser, Ser-Gly, Gly-Ala, Ala-Gly, and Ala-Ala, more preferably Gly-Ser and Gly-Gly.
  • S B is —(P U ii ) n —, wherein n is 2 to 4, more preferably 3, and each P U ii is independently selected from Gly-Ser and Gly-Gly.
  • S B is —(P U ii ) n —, n is 3 and each P U i is independently selected from Gly-Ser and Gly-Gly, with a preferred sequence being -(Gly-Ser)-(Gly-Ser)-(Gly-Gly) (SEQ ID NO: 114).
  • Amino acids having stereogenic centres within P U i and P U ii may be racemic, enantioenriched, or enantiopure.
  • the or each amino acid is independently an L-amino acid.
  • the or each amino acid is independently a D-amino acid.
  • Each P U iii may be independently a residue of general formula:
  • m is 0, 1, 2, 3, 4, or 5, preferably 1 or 2
  • p is 1, 3, 4, or 5, preferably 1.
  • m is 1 and p is 1, that is, P U iii is a residue of 8-amino-3,6-dioxaoctanoic acid (also known as ⁇ 2-[2-aminoethoxy]ethoxy ⁇ acetic acid and H 2 N-PEG 3 -COOH). This residue is referred to herein as -PEG 3 -.
  • PEG chains are also known in the art.
  • 11-amino-3,6,9-trioxaundecanoic acid also known as H 2 N-PEG 4 -COOH or -PEG 4 -.
  • S B is —(P U iii ) n —, wherein n is 1 to 3, more preferably 2.
  • S B is -PEG 3 -PEG 3 -.
  • the compounds of the invention may provide an attractive treatment option for metabolic diseases including obesity and diabetes mellitus (diabetes).
  • Diabetes comprises a group of metabolic diseases characterized by hyperglycemia resulting from defects in insulin secretion, insulin action, or both. Acute signs of diabetes include excessive urine production, resulting compensatory thirst and increased fluid intake, blurred vision, unexplained weight loss, lethargy, and changes in energy metabolism. However, symptoms are often not severe or may be absent.
  • the chronic hyperglycemia of diabetes is associated with long-term damage, dysfunction, and failure of various organs, notably the eyes, kidneys, nerves, heart and blood vessels. Diabetes is classified into type 1 diabetes, type 2 diabetes and gestational diabetes on the basis on pathogenetic characteristics. Type 1 diabetes accounts for 5-10% of all diabetes cases and is caused by auto-immune destruction of insulin-secreting pancreatic ⁇ -cells.
  • Type 2 diabetes accounts for 90-95% of diabetes cases and is a result of a complex set of metabolic disorders. However, symptoms are often not severe or may be absent. Type 2 diabetes is the consequence of endogenous insulin production becoming insufficient to maintain plasma glucose levels below diagnostic thresholds.
  • Gestational diabetes refers to any degree of glucose intolerance identified during pregnancy.
  • Pre-diabetes includes impaired fasting glucose and impaired glucose tolerance and refers to those states that occur when blood glucose levels are elevated but below the levels that are established for the clinical diagnosis for diabetes.
  • a large proportion of people with type 2 diabetes and pre-diabetes are at increased risk of morbidity and mortality due to the high prevalence of additional metabolic risk factors, including abdominal obesity (excessive fat tissue around the abdominal internal organs), atherogenic dyslipidemia (blood fat disorders including high triglycerides, low HDL cholesterol and/or high LDL cholesterol, which foster plaque buildup in artery walls), elevated blood pressure (hypertension) a prothrombotic state (e.g. high fibrinogen or plasminogen activator inhibitor- 1 in the blood), and/or a proinflammatory state (e.g., elevated C-reactive protein in the blood).
  • abdominal obesity excessive fat tissue around the abdominal internal organs
  • atherogenic dyslipidemia blood fat disorders including high triglycerides, low HDL cholesterol and/or high LDL cholesterol, which foster plaque buildup in artery walls
  • elevated blood pressure hypertension
  • a prothrombotic state e.g. high fibrinogen or plasminogen activator inhibitor- 1 in the blood
  • obesity confers an increased risk of developing pre-diabetes, type 2 diabetes as well as, e.g., certain types of cancer, obstructive sleep apnea and gall-bladder disease.
  • Dyslipidemia is associated with increased risk of cardiovascular disease.
  • High Density Lipoprotein (HDL) is of clinical importance since an inverse correlation exists between plasma HDL concentrations and risk of atherosclerotic disease.
  • the majority of cholesterol stored in atherosclerotic plaques originates from LDL and hence an elevated concentration of Low Density Lipoproteins (LDL) is closely associated with atherosclerosis.
  • LDL Low Density Lipoproteins
  • the HDL/LDL ratio is a clinical risk indictor for atherosclerosis and coronary atherosclerosis in particular.
  • glucagon-GIP-GLP1 triple agonists act as glucagon-GIP-GLP1 triple agonists.
  • the triple agonist may combine the effect of glucagon, e.g., on fat metabolism with the effect of GIP on improved glycemic control and the effect of GLP-1 e.g., on blood glucose levels and food intake. They may therefore act to accelerate elimination of excessive adipose tissue, induce sustainable weight loss, and improve glycemic control.
  • Triple glucagon-GIP-GLP1 agonists may also act to reduce cardiovascular risk factors such as high cholesterol and such as high LDL-cholesterol.
  • the triple agonist compounds of the present invention may therefore be used (alone or in combination) as pharmaceutical agents for preventing weight gain, promoting weight loss, reducing excess body weight or treating obesity (e.g., by control of appetite, feeding, food intake, calorie intake, and/or energy expenditure and lipolysis), including morbid obesity, as well as associated diseases and health conditions including but not limited to obesity linked inflammation, obesity linked gallbladder disease and obesity induced sleep apnea.
  • the compounds may also be used for treatment of insulin resistance, glucose intolerance, pre-diabetes, increased fasting glucose, type 2 diabetes, hypertension, dyslipidemia (or a combination of these metabolic risk factors), atherosclerosis, arteriosclerosis, coronary heart disease, peripheral artery disease and stroke. These are all conditions which may be associated with obesity. However, the effects of the compounds employed in the context of the invention on these conditions may be mediated in whole or in part via an effect on body weight, or may be independent thereof.
  • the triple agonist compounds may thus be used (alone or in combination) for the treatment and/or prevention of any of the diseases, disorders, or conditions described herein, including insulin resistance, glucose intolerance, increased fasting glucose, pre-diabetes, type 1 diabetes, type 2 diabetes, gestational diabetes hypertension, dyslipidemia, or a combination thereof.
  • the diabetes related disorder is selected from atherosclerosis, arteriosclerosis, coronary heart disease, peripheral artery disease and stroke; or associated with a condition selected from atherogenic dyslipidemia, blood fat disorders, elevated blood pressure, hypertension, a prothrombotic state, and proinflammatory state, or a combination thereof.
  • the blood fat disorder is selected from high triglycerides, low HDL cholesterol, high LDL cholesterol, plaque buildup in artery walls, or a combination thereof.
  • the prothrombotic state is selected from high fibrinogen levels in the blood and high plasminogen activator inhibitor-1 levels in the blood.
  • the proinflammatory state is an elevated C-reactive protein level in the blood.
  • the obesity related disorder is selected from obesity linked inflammation, obesity linked gallbladder disease and obesity induced sleep apnea.
  • the triple agonist compounds may also be used for the treatment and/or prevention of any of the diseases, disorders, or conditions associated with diabetes related osteoporosis including increased risk of bone fractures.
  • the observed increase in fracture risk is likely to be related to impaired bone quality rather than to bone mineral density.
  • the related mechanisms due at least in part to hyperglycemia, neuropathy, and higher incidence of hypovitaminosis D, are not yet fully understood.
  • the invention provides the use of a triple agonist compound as described, in the manufacture of a medicament for any of the clinical applications described in this specification. Reference to a compound for use in any such method should be construed accordingly.
  • the invention also provides a therapeutic kit comprising a triple agonist of the invention, optionally in combination with a pharmaceutically acceptable carrier.
  • the invention provides a device comprising a triple agonist of the invention for delivery of the triple agonist to a subject.
  • the triple agonist compounds of the present invention, or salts or solvates thereof may be formulated as pharmaceutical compositions prepared for storage or administration, which typically comprise a therapeutically effective amount of a compound employed in the context of the invention, or a salt or solvate thereof, in a pharmaceutically acceptable carrier.
  • the pharmaceutical composition is formulated as a liquid suitable for administration by injection or infusion, or which is formulated to cause slow release of the triple agonist compound
  • the therapeutically effective amount of a compound of the present invention will depend, e.g., on the route of administration, the type of mammal being treated, and the physical characteristics of the specific mammal under consideration. These factors and their relationship to determining this amount are well known to skilled practitioners in the medical arts. This amount and the method of administration can be tailored to achieve optimal efficacy, and may depend on such factors as weight, diet, concurrent medication and other factors, well known to those skilled in the medical arts. The dosage sizes and dosing regimen most appropriate for human use may be guided by the results obtained by the present invention, and may be confirmed in properly designed clinical trials.
  • An effective dosage and treatment protocol may be determined by conventional means, starting with a low dose in laboratory animals and then increasing the dosage while monitoring the effects, and systematically varying the dosage regimen as well. Numerous factors may be taken into consideration by a clinician when determining an optimal dosage for a given subject. Such considerations are known to the skilled person.
  • pharmaceutically acceptable carrier includes any of the standard pharmaceutical carriers. Pharmaceutically acceptable carriers for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remington's Pharmaceutical Sciences, Mack Publishing Co. (A. R. Gennaro edit. 1985). For example, sterile saline and phosphate- buffered saline at slightly acidic or physiological pH may be used.
  • Suitable pH buffering agents may be, e.g., phosphate, citrate, acetate, lactate, maleate, tris/hydroxymethyl)aminomethane (TRIS), N-Tris(hydroxymethyl)methyl-3-aminopropanesulphonic acid (TAPS), ammonium bicarbonate, diethanolamine, histidine, which in certain embodiments is a preferred buffer, arginine, lysine, or acetate or mixtures thereof.
  • TIS tris/hydroxymethyl)aminomethane
  • TAPS N-Tris(hydroxymethyl)methyl-3-aminopropanesulphonic acid
  • ammonium bicarbonate diethanolamine
  • histidine which in certain embodiments is a preferred buffer, arginine, lysine, or acetate or mixtures thereof.
  • the term further encompasses any agents listed in the US Pharmacopeia for use in animals, including humans.
  • salts include pharmaceutically acceptable salts, such as, e.g., acid addition salts and basic salts.
  • acid addition salts include hydrochloride salts, citrate salts and acetate salts.
  • basic salts include salts where the cation is selected from alkali metals, such as sodium and potassium, alkaline earth metals such as calcium, and ammonium ions + N(R 3 ) 3 (R 4 ), where R 3 and R 4 independently designate optionally substituted C 1-6 -alkyl, optionally substituted C 2-6 -alkenyl, optionally substituted aryl, or optionally substituted heteroaryl.
  • Treatment is an approach for obtaining beneficial or desired clinical results.
  • beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable.
  • Treatment may also mean prolonging survival as compared to expected survival if not receiving treatment.
  • Treatment is an intervention performed with the intention of preventing the development or altering the pathology of a disorder. Accordingly, “treatment” refers to both therapeutic treatment and prophylactic or preventative measures in certain embodiments.
  • Those in need of treatment include those already with the disorder as well as those in which the disorder is to be prevented.
  • treatment is meant inhibiting or reducing an increase in pathology or symptoms (e.g. weight gain, hyperglycemia) when compared to the absence of treatment, and is not necessarily meant to imply complete cessation of the relevant condition.
  • compositions of the invention may be in unit dosage form.
  • the composition is divided into unit doses containing appropriate quantities of the active component.
  • the unit dosage form can be a packaged preparation, the package containing discrete quantities of the preparations, for example, packeted tablets, capsules, and powders in vials or ampoules.
  • the unit dosage form can also be a capsule, cachet, or tablet itself, or it can be the appropriate number of any of these packaged forms. It may be provided in single dose injectable form, for example in the form of an injection pen.
  • Compositions may be formulated for any suitable route and means of administration.
  • Pharmaceutically acceptable carriers or diluents include those used in formulations suitable for oral, rectal, nasal or parenteral (including subcutaneous, intramuscular, intravenous, intradermal, and transdermal) administration.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Subcutaneous or transdermal modes of administration may be particularly suitable for certain of the compounds described herein.
  • a compound of the invention may be administered as part of a combination therapy with at least one other agent for treatment of diabetes, obesity, dyslipidemia, or hypertension.
  • the at least two active agents may be given together or separately, and as part of the same pharmaceutical formulation or as separate formulations.
  • the triple agonist compound (or the salt or solvate thereof) may be used in combination with an antidiabetic agent including but not limited to metformin, a sulfonylurea, a glinide, a DPP-IV inhibitor, a glitazone, or insulin.
  • the compound or salt or solvate thereof is used in combination with insulin, DPP-IV inhibitor, sulfonylurea or metformin, particularly sulfonylurea or metformin, for achieving adequate glycemic control.
  • the compound or salt or solvate thereof is used in combination with insulin or an insulin analogue for achieving adequate glycemic control.
  • insulin analogues include but are not limited to LANTUS®, NOVORAPID®, HUMALOG®, NOVOMIX®, ACTRAPHANE HM®, LEVEMIR® and APIDRA®.
  • the triple agonist compound or salt or solvate thereof may further be used in combination with one or more of an anti-obesity agent, including but not limited to a glucagon-like peptide receptor 1 agonist, peptide YY or analogue thereof, cannabinoid receptor 1 antagonist, lipase inhibitor, melanocortin receptor 4 agonist, or melanin concentrating hormone receptor 1 antagonist.
  • an anti-obesity agent including but not limited to a glucagon-like peptide receptor 1 agonist, peptide YY or analogue thereof, cannabinoid receptor 1 antagonist, lipase inhibitor, melanocortin receptor 4 agonist, or melanin concentrating hormone receptor 1 antagonist.
  • the triple agonist compound or salt or solvate thereof may be used in combination with an anti-hypertension agent, including but not limited to an angiotensin-converting enzyme inhibitor, angiotensin II receptor blocker, diuretics, beta-blocker, or calcium channel blocker.
  • an anti-hypertension agent including but not limited to an angiotensin-converting enzyme inhibitor, angiotensin II receptor blocker, diuretics, beta-blocker, or calcium channel blocker.
  • the triple agonist compound or salt thereof may be used in combination with an anti-dyslipidemia agent, including but not limited to a statin, a fibrate, a niacin and/or a cholesterol absorption inhibitor.
  • an anti-dyslipidemia agent including but not limited to a statin, a fibrate, a niacin and/or a cholesterol absorption inhibitor.
  • the invention provides a nucleic acid encoding a peptide having the sequence X1-X30 of Formula I. Also provided is an expression construct (also known as an expression vector) comprising a nucleic acid of the invention in operable linkage with suitable regulatory elements to direct expression of the peptide, e.g. transcription and translation.
  • an expression construct also known as an expression vector
  • the invention also provides a host cell comprising a nucleic acid or expression construct and capable of expressing, and optionally secreting, the peptide.
  • the invention provides a method of producing a compound of the invention, the method comprising culturing the host cells described above under conditions suitable for expressing the compound and purifying the compound thus produced.
  • the invention also provides a nucleic acid molecule, an expression vector, or a host cell, as described above, for use in a method of medica treatment, and in particular for treatment of the metabolic disorders discussed elsewhere in this specification.
  • a nucleic acid molecule may encode a compound of the invention, a peptide having the amino acid sequence X1-X30 of Formula I, or a peptide which is a precursor of a compound of the invention.
  • nucleic acid sequences will be provided as expression constructs wherein the encoding nucleic acid is in functional linkage with appropriate control sequences to direct its expression.
  • the invention provides a method of producing a triple agonist of the invention, the method comprising expressing an amino acid precursor of the triple agonist and modifying the precursor to provide the triple agonist.
  • the modification may comprise chemical modification of a Lys, Arg or Cys residue present at a position ⁇ to introduce the lipophilic moiety, modification of the N- or C-terminus, and/or modification of any other amino acid side chains in the molecule (e.g. to introduce a non-naturally occurring amino acid residue).
  • the compounds of the invention may also be manufactured by standard peptide synthetic methods, e.g. by standard solid-phase or liquid-phase methodology, either stepwise or by fragment assembly, and isolating and purifying the final peptide compound product, or by any combinations of recombinant and synthetic methods.
  • Piperidine/NMP (20%; 10 ml) was added to the resin for initial deprotection and the mixture was heated by microwaves (30 sec; 40° C.). The reaction vessel was drained and a second portion of piperidine/DMF (20%; 10 ml) was added and heated (75° C.; 3 min.) again. The resin was then washed with NMP (6 ⁇ 10 ml).
  • Fmoc-Lys(ivDde)-OH or alternatively another amino acid with an orthogonal side chain protective group was introduced at the position of the acylation.
  • the N-terminal of the peptide backbone was then Boc-protected using Boc2O or alternatively by using a Boc-protected amino acid in the last coupling. While the peptide was still attached to the resin, the orthogonal side chain protective group was selectively cleaved using freshly prepared hydrazine hydrate (2-4%) in NMP for 2 ⁇ 15 min.
  • the unprotected lysine side chain was first coupled with Fmoc-Glu-OtBu or another spacer amino acid, which was deprotected with piperidine and acylated with a lipophilic moiety using the peptide coupling methodology as described above.
  • Abbreviations employed are as follows:
  • COMU 1-[(1-(cyano-2-ethoxy-2-oxoethylideneaminooxy)-dimethylamino-morpholinomethylene)]methanaminium hexafluorophosphate
  • ivDde 1-(4,4-dimethyl-2,6-dioxocyclohexylidene)3-methyl-butyl
  • DIPEA diisopropylethylamine
  • TIS triisopropylsilane
  • the resin was washed with EtOH (3 ⁇ 10 ml) and Et 2 O (3 ⁇ 10 ml) and dried to constant weight at room temperature (r.t.).
  • the crude peptide was cleaved from the resin by treatment with TFA/TIS/water (95/2.5/2.5; 40 ml, 2 h; r.t.). Most of the TFA was removed at reduced pressure and the crude peptide was precipitated and washed three times with diethylether and dried to constant weight at room temperature.
  • the crude peptide was purified to greater than 90% by preparative reverse phase HPLC using a PerSeptive Biosystems VISION Workstation equipped with a C-18 column (5 cm; 10 ⁇ m) and a fraction collector and run at 35 ml/min with a gradient of buffer A (0.1% TFA, aq.) and buffer B (0.1% TFA, 90% MeCN, aq.). Fractions were analyzed by analytical HPLC and MS and relevant fractions were pooled and lyophilized. The final product was characterized by HPLC and MS. The synthesized compounds are shown in Table 1:
  • H-YSQGTFTSDYSKYLDSKAAHDFVEWLLRA-NH 2 (SEQ ID NO: 1) 2 H-Y-Aib-QGTFTSDYSKYLDS-K(Hexadecanoyl-isoGlu)- AAHDFVEWLLSA-NH 2 (SEQ ID NO: 2) 3 H-Y-Aib-QGTFTSDYSKYLDSKAAHDFVEWLLRA-NH 2 (SEQ ID NO: 3) 4 H-YSQGTFTSDYSKYLD-K(Hexadecanoyl-isoGlu)-KAAHDFVEWLLRA- NH 2 (SEQ ID NO: 4) 5 H-Y-Aib-QGTFTSDYSIYLDSKAAHDFVEWLLSA-NH 2 (SEQ ID NO: 5) 6 H-Y-Aib-QGTFTSDYSKYLDSKAARDFVEWLLSA-NH 2 (SEQ ID NO: 6) 7 H-Y-A
  • GIP-R Human GIP Receptor
  • GLP-1-R GLP-1 Receptor
  • GCG-R Glucagon Receptor
  • HEK293 cells expressing the human GIP R, GLP-1 R or GCG R (stable cell lines generated through transfection of the cDNA for human GIP R, GLP-1 R or GCG R and selection of stable clones) were seeded at 30,000 cells/well in 96-well microtiter plates coated with 0.01% poly-L-lysine, and grown for 1 day in culture in 200 ⁇ l growth medium (DMEM, 10% FCS, Penicillin (100 IU/ml), Streptomycin (100 ⁇ g/ml)).
  • DMEM 10% FCS
  • Penicillin 100 IU/ml
  • Streptomycin 100 ⁇ g/ml
  • Results were converted into cAMP concentrations using a cAMP standard curve prepared in KRBH buffer containing 0.1% (v/v) DMSO. The resulting cAMP curves were plotted as absolute cAMP concentrations (nM) over log (test compound concentration) and analyzed using the curve fitting program XLfit.
  • the exemplified compounds of the invention will have activities at the GCG-R that are close to that of native glucagon. At the same time, it is anticipated that they will exhibit strong GLP-1-R activation with EC 50 well below 1 nM. Likewise, it is anticipated that these peptides will also exhibit strong GIP-R activity with and EC 50 below or just above 1 nM.
  • mice Males with a body weight of approximately 25 g were given a single intravenous (i.v.) bolus of each peptide to be tested.
  • blood samples were drawn 0.08, 0.17, 0.5, 1, 4, 8, 16 and 24 hours post-dose. Blood samples were drawn by sublingual bleeding.
  • the dosing vehicle was a phosphate buffer containing mannitol (pH 7.5).
  • mice were drawn, i.e. 16 mice were included for each compound.
  • the mice were euthanized immediately after blood sampling by cervical dislocation.
  • Plasma samples were analyzed after solid phase extraction (SPE) or precipitation by liquid chromatography mass spectrometry (LC-MS/MS). Mean plasma concentrations were used for calculation of the pharmacokinetic parameters using the non-compartmental approach in Phoenix WinNonlin 6.3.
  • Plasma terminal elimination half-life (T1 ⁇ 2) was determined as In(2)/ ⁇ z where ⁇ z is the magnitude of the slope of the log linear regression of the log concentration versus time profile during the terminal phase. The results are summarized in Table 3

Abstract

The present invention relates to compounds which have agonist activity at the glucagon, GIP and GLP-1 receptors, and to their use in the treatment of metabolic disorders.

Description

    SEQUENCE LISTING
  • The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Jul. 29, 2021 is named 50412-096003_Sequence_Listing_07_29_21_ST25 and is 73,649 bytes in size.
  • FIELD OF THE INVENTION
  • The invention relates to compounds having agonist activity at the glucagon, GIP and GLP-1 receptors, and to their use in the treatment of metabolic disorders.
  • BACKGROUND OF THE INVENTION
  • Diabetes and obesity are increasing health problems globally and are associated with various other diseases, particularly cardiovascular diseases (CVD), obstructive sleep apnea, stroke, peripheral artery disease, microvascular complications and osteoarthritis. There are 246 million people worldwide with diabetes, and by 2025 it is estimated that 380 million will have diabetes. Many have additional cardiovascular risk factors including high/aberrant LDL and triglycerides and low HDL. Cardiovascular disease accounts for about 50% of the mortality in people with diabetes, and the morbidity and mortality rates relating to obesity and diabetes underscore the medical need for efficacious treatment options.
  • Pre-proglucagon is a 158 amino acid precursor polypeptide that is processed in different tissues to form a number of different proglucagon-derived peptides, including glucagon, glucagon-like peptide-1 (GLP-1 or GLP1), glucagon-like peptide-2 (GLP-2) and oxyntomodulin (OXM), which are involved in a wide variety of physiological functions, including glucose homeostasis, insulin secretion, gastric emptying, and intestinal growth, as well as the regulation of food intake. Glucagon is a 29-amino acid peptide that corresponds to amino acids 33 through 61 of pre-proglucagon, while GLP-1 is produced as a 37-amino acid peptide that corresponds to amino acids 72 through 108 of pre-proglucagon.
  • When blood glucose begins to fall, glucagon, a hormone produced by the pancreas, signals the liver to break down glycogen and release glucose, causing blood glucose levels to rise toward a normal level. In addition to controlling glucose homeostasis, glucagon reduces body weight probably through inhibition of food intake and stimulation of energy expenditure and/or lipolysis. GLP-1 has different biological activities compared to glucagon. Its actions include stimulation of insulin synthesis and secretion, inhibition of glucagon secretion, and inhibition of food intake. GLP-1 has been shown to reduce hyperglycemia (elevated glucose levels) in diabetic patients.
  • Exendin-4, a peptide from lizard venom that shares about 50% amino acid identity with GLP-1, activates the GLP-1 receptor and likewise has been shown to reduce hyperglycemia in diabetic patients.
  • Glucose-dependent insulinotropic polypeptide (GIP) is a 42-amino acid gastrointestinal regulatory peptide that, like GLP-1, stimulates insulin secretion from pancreatic β(beta) cells in the presence of elevated blood glucose levels. It is derived by proteolytic processing from a 133-amino acid precursor, preproGlP.
  • Interestingly, novel glucagon-GLP-1 dual acting receptor agonists are currently in pre-clinical development (see, e.g., WO2011/006497). In comparison to GLP-1 analogues, glucagon-GLP-1 dual agonists are associated with more profound and sustained body weight loss in animal models on top of the improvements in glycemic control. Thus, glucagon based drugs may have promise for the treatment of type 2 diabetes mellitus and/or obesity.
  • Incretins are gastrointestinal hormones that regulate blood glucose by enhancing glucose-stimulated insulin secretion (Drucker, D J and Nauck, M A, Lancet 368: 1696-705 (2006)). Two of the above mentioned peptides are known as incretins: GLP-1 and GIP. The discovery of the incretins has led to the development of two new classes of drugs for the treatment of diabetes mellitus. Thus, injectable GLP-1 receptor agonists, and small molecule compounds (oral DPP-4 inhibitors) that inhibit enzymatic inactivation of both endogenous GLP-1 and GIP, are now on the market (GLP-1 receptor agonists: Byetta™, Bydureon™ Lixisenatide™ and Liraglutide™, and DPP-4 inhibitors: Januvia™ Galvus™ Onglyza™ and Trajenta™). Apart from the acute effects of GLP-1 and GIP on insulin secretion, the two peptides also have long term effects. Evidence from several labs indicates that GLP-1 R agonists protect pancreatic β-cells by inhibiting apoptosis and enhancing proliferation. For instance, the study by Farilla et al. showed that GLP-1 had anti-apoptotic effects in human islets (Farilla, L, Endocrinology 144: 5149-58 (2003)). Such effects have not been reported for GIP until recently. In 2010, Weidenmaier et al. reported that a DPP-4 resistant GIP analogue has anti-apoptotic effects (Weidenmaier, S D, PLOS One 5(3): e9590 (2010)). Interestingly, in a mice model of diabetes and obesity the combination of the GLP-1 receptor agonist Liraglutide and an acylated GIP analogue show superior effects compared to treatment with Liraglutide or GIP analogue alone (Gault, V A, Clinical Science 121: 107-117 (2011)).
  • Chronic treatment with the GLP-1 receptor agonists causes significant weight loss in diabetic humans. Interestingly, extended use of DPP-4 inhibitors in similar patients does not consistently change body weight. Evidence suggests (Matthias Tschöp oral presentation at ADA (American Diabetes Association), 2011) that body weight loss associated with GLP-1 agonist treatment is enhanced when GLP-1 and GIP are co-administered. In rodents, co-administration of GLP-1 and GIP results in greater body weight loss than GLP-1 treatment alone (Finan, Sci Transl Med. 2013; 5(209):209ra151. Irwin N et al, 2009, Regul Pept; 153: 70-76. Gault et al, 2011, Clin Sci; 121:107-117). Thus, in addition to improving blood glucose, GIP may also enhance GLP-1-mediated body weight loss. In the same presentation it was also shown that combining glucagon, GLP-1 and GIP receptor agonism led to further body weight loss in DIO mice.
  • By combining glucagon, GLP-1 and GIP receptor agonism in novel inventive peptides it is anticipated that superior glycemic control and body weight loss can be achieved. Such peptides are likely to have strong incretin actions and improved β-cell preservation from the GLP-1 and GIP components, and have improved body weight loss from all three components by stimulating energy expenditure, lipolysis and reducing food intake.
  • SUMMARY OF THE INVENTION
  • Broadly, the present invention concerns Glucagon-GLP-1-GIP triple agonists (referred to in this specification as “triple agonists”) which comprise one or more substitutions as compared to wild-type glucagon and which may have the property of an altered, preferably increased GIP and GLP-1 receptor activity, e.g. as assessed in in vitro efficacy assays. In the present invention it has been found that Glucagon-GLP-1-GIP triple acting receptor agonists are superior to existing and marketed GLP-1 analogues because the triple agonists offer improved glycemic control, possible islet and β-cell preservation and enhanced body weight loss. The Glucagon-GLP-1-GIP triple agonists could be used as therapeutics for both type 2 diabetes mellitus, obesity and related disorders.
  • The invention provides a triple agonist having the general formula I:
  • (I)
    (SEQ ID NO: 52)
    R1-Tyr-X2-Gln-Gly-Thr-Phe-Thr-Ser-Asp-X10-Ser-X12-
    X13-Leu-X15-X16-X17-Ala-X19-X20-X21-Phe-X23-X24-
    Trp-Leu-X27-X28-X29-X30-Y1-R2
  • wherein
  • R1 is H— (i.e., hydrogen), C1-4 alkyl, acetyl, formyl, benzoyl, trifluoroacetyl or pGlu;
  • X2 is Aib, Gly, Ala, D-Ala, Ser, N-Me-Ser, Ac3c, Ac4c or Ac5c;
  • X10 is Tyr or Leu;
  • X12 is Lys, Ile or ψ;
  • X13 is Ala, Tyr or Aib;
  • X15 is Asp or Glu;
  • X16 is Ser, Glu, Lys or ψ;
  • X17 is Lys or ψ
  • X19 is Gln or Ala;
  • X20 is Lys, His, Arg or ψ;
  • X21 is Ala, Asp or Glu;
  • X23 is Val or Ile;
  • X24 is Asn, Glu or ψ;
  • X27 is Leu, Glu or Val;
  • X28 is Ala, Ser, Arg or ψ;
  • X29 is Aib, Ala, Gln or Lys;
  • X30 is Lys, Gly, or is absent;
  • Y1 is
    (SEQ ID NO: 53)
    Gly-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser,
    (SEQ ID NO: 54)
    Gly-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Ser,
    (SEQ ID NO: 55)
    Lys-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser,
    (SEQ ID NO: 56)
    Lys-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Ser,
    (SEQ ID NO: 57)
    Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser
    or
    (SEQ ID NO: 58)
    Pro-Ser-Ser-Gly-Ala-Pro-Pro-Ser,
    or
    is absent;
  • ψ is a residue of Lys, Arg, Orn or Cys in which the side chain is conjugated to a lipophilic substituent;
  • and
  • R2 is —NH2 or —OH;
  • or a pharmaceutically acceptable salt or solvate thereof.
  • In some embodiments, ψ is present at one of positions X12, X16 and X17.
  • In some embodiments, the compound contains only one residue ψ, which may be present at any one of positions X12, X16, X17, X20, X24 or X28. For example, it may be present at one of X12, X16 and X17.
  • In some embodiments, the compound may possess one or more of the following sets of residues:
  • K12 and Y13; I12 and Y13; K12 and A13; I12 and A13; or ψ12 and Y13;
  • D15 and S16; D15 and E16; E15 and K16; D15 and ψ16; E15 and S16; or E15 and ψ16;
  • A19, H20, and D21; A19, K20, and D21; A19, R20, and D21; Q19, K20, and E21; A19, K20, and E21; or Q19, R20, and A21; but especially A19, H20, and D21; A19, R20, and D21 or Q19, R20, and A21;
  • I23 and E24; V23 and E24; or V23 and N24;
  • L27, R28, and A29; L27, S28, and A29; L27, A28, and Q29; E27, S28, and A29; or V27, and A28, and Aib29;
  • E15 and K17;
  • E15 and ψ17;
  • E15 and ψ17 and Q19;
  • Q19 and E24;
  • E16 and ψ17 and Q19; and/or
  • K16 and ψ17 and Q19.
  • Any one of these sets of residues, or combinations thereof, may be combined with:
  • Aib2, Ser2 or Ac4c2, especially Aib2; and/or
  • Tyr10 or Leu10, especially Tyr10.
  • Positions 1 to 29 may have the sequence
  • (SEQ ID NO: 1)
    YSQGTFTSDYSKYLDSKAAHDFVEWLLRA;
    (SEQ ID NO: 59)
    YSQGTFTSDSKYLDΨKAAHDFVEWLLRA;
    (SEQ ID NO: 5)
    Y-Aib-QGTFTSDYSIYLDSKAAHDFVEWLLSA;
    (SEQ ID NO: 9)
    Y-Aib-QGTFTSDYSIYLDEKAAHDFVEWLLSA;
    (SEQ ID NO: 14)
    Y-Aib-QGTFTSDYSIYLEKKAAHDFVEWLLSA;
    (SEQ ID NO: 17)
    Y-Aib-QGTFTSDYSIYLESKAAHDFVEWLLSA;
    (SEQ ID NO: 18)
    Y-Aib-QGTFTSDYSIYLDKKAAHDFVEWLLSA;
    (SEQ ID NO: 60)
    Y-Aib-QGTFTSDYSIYLDEΨAAHDFVEWLLSA;
    (SEQ ID NO: 61)
    Y-Aib-QGTFTSDYSIYLDSΨAAHDFVEWLLSA;
    (SEQ ID NO: 62)
    Y-Aib-QGTFTSDYSKYLDSΨAAHDFVEWLLSA;
    (SEQ ID NO: 7)
    Y-Aib-QGTFTSDYSKALDSKAAHDFVEWLLSA;
    (SEQ ID NO: 8)
    Y-Aib-QGTFTSDYSKYLESKAAHDFVEWLLSA;
    (SEQ ID NO: 10)
    Y-Aib-QGTFTSDYSIYLDSKAAKDFVEWLLSA;
    (SEQ ID NO: 16)
    Y-Aib-QGTFTSDYSIYLDEKAAKDFVEWLLSA;
    (SEQ ID NO: 6)
    Y-Aib-QGTFTSDYSKYLDSKAARDFVEWLLSA;
    (SEQ ID NO: 15)
    Y-Aib-QGTFTSDYSIYLEKKAQKEFVEWLLSA;
    (SEQ ID NO: 21)
    Y-Aib-QGTFTSDYSKYLEKKAQKEFVEWLLSA;
    (SEQ ID NO: 19)
    Y-Aib-QGTFTSDYSIYLEKKAAKEFVEWLLSA;
    (SEQ ID NO: 20)
    Y-Aib-QGTFTSDYSKALDEKAAKEFVEWLLSA;
    (SEQ ID NO: 63)
    Y-Aib-QGTFTSDYSIYLEKΨAAKEFVEWLLSA;
    (SEQ ID NO: 64)
    Y-Aib-QGTFTSDYSIYLEΨKAAKEFVEWLLSA;
    (SEQ ID NO: 65)
    Y-Aib-QGTFTSDYSΨYLEKKAAKEFVEWLLSA;
    (SEQ ID NO: 11)
    Y-Aib-QGTFTSDYSIYLDSKAAHDFVNWLLSA;
    (SEQ ID NO: 110)
    Y-Aib-QGTFTSDLSIALEKΨAQRAFVEWLLAQ;
    (SEQ ID NO: 67)
    Y-Aib-QGTFTSDYSKYLDEΨAAKDFIEWLESA;
    (SEQ ID NO: 68)
    Y-Aib-QGTFTSDYSIYLDEΨAAKDFVEWLESA;
    (SEQ ID NO: 69)
    Y-Aib-QGTFTSDYSIYLDEΨAAKDFIEWLESA;
    (SEQ ID NO: 70)
    Y-Aib-QGTFTSDYSIYLDEΨAAKEFIEWLESA;
    (SEQ ID NO: 3)
    Y-Aib-QGTFTSDYSKYLDSKAAHDFVEWLLRA;
    (SEQ ID NO: 12)
    Y-Aib-QGTFTSDYSIYLDSKAAHDFVEWLLRA;
    (SEQ ID NO: 71)
    Y-Aib-QGTFTSDYSIYLDSΨAAHDFVEWLLRA;
    (SEQ ID NO: 72)
    Y-Aib-QGTFTSDYSIYLEKΨAQRAFVEWLLRA;
    (SEQ ID NO: 73)
    Y-Aib-QGTFTSDYSIALDKΨAQRAFVNWLVA-Aib;
    (SEQ ID NO: 74)
    Y-Ac4c-QGTFTSDYSIYLDEΨAAKEFIEWLESA;
    (SEQ ID NO: 75)
    Y-Ac4c-QGTFTSDYSIALEKΨAQRAFVEWLLAQ;
    or
    (SEQ ID NO: 76)
    Y-Ac4c-QGTFTSDYSIYLDKΨAQRAFVEWLLAQ.
  • Alternatively, positions 1 to 29 may differ at up to 4 positions, e.g. at 1, 2, 3 or 4 positions, from any of the specific sequences shown above, within the constraints of Formula I.
  • For example, positions 1 to 29 may differ at up to 4 positions, e.g. at 1, 2, 3 or 4 positions from one of the following sequences:
  • (SEQ ID NO: 63)
    Y-Aib-QGTFTSDYSIYLEKΨAAKEFVEWLLSA;
    (SEQ ID NO: 60)
    Y-Aib-QGTFTSDYSIYLDEΨAAHDFVEWLLSA;
    or
    (SEQ ID NO: 77)
    Y-Aib-QGTFTSDYSIALEKΨAQRAFVEWLLAQ.
  • Positions 1 to 29 may have the sequence:
  • (SEQ ID NO: 4)
    YSQGTFTSDYSKYLD-K(Hexadecanoyl-isoGlu)-KAAHDFVEWLLRA;
    (SEQ ID NO: 27)
    Y-Aib-QGTFTSDYSIYLDE-K(Hexadecanoyl-isoGlu)-AAHDFVEWLLSA;
    (SEQ ID NO: 85)
    Y-Aib-QGTFTSDYSIYLDS-K(Hexadecanoyl-isoGlu)-AAHDFVEWLLSA;
    (SEQ ID NO: 2)
    Y-Aib-QGTFTSDYSKYLDS-K(Hexadecanoyl-isoGlu)-AAHDFVEWLLSA;
    (SEQ ID NO: 22)
    Y-Aib-QGTFTSDYSIYLEK-K(Hexadecanoyl-isoGlu)-AAKEFVEWLLSA;
    (SEQ ID NO: 23)
    Y-Aib-QGTFTSDYSIYLE-K(Hexadecanoyl-isoGlu)-KAAKEFVEWLLSA;
    (SEQ ID NO: 24)
    Y-Aib-QGTFTSDYS-K(Hexadecanoyl-isoGlu)-YLEKKAAKEFVEWLLSA;
    (SEQ ID NO: 22)
    Y-Aib-QGTFTSDYSIYLEK-K(Hexadecanoyl-isoGlu)-AAKEFVEWLLSA;
    (SEQ ID NO: 86)
    Y-Aib-QGTFTSDYSIYLEK-K(Octadecanoyl-isoGlu-Peg3-Peg3)-AAKEFVEWLLSA;
    (SEQ ID NO: 87)
    Y-Aib-QGTFTSDYSIYLEK-K([17-carboxy-heptadecanoyl]-isoGlu-Peg3-Peg3)-
    AAKEFVEWLLSA;
    (SEQ ID NO: 88)
    Y-Aib-QGTFTSDYSIYLDK-K([17-carboxy-heptadecanoyl]-isoGlu-Peg3-Peg3)-
    AQRAFVEWLLAQ;
    (SEQ ID NO: 89)
    Y-Aib-QGTFTSDYSIYLDK-K([17-carboxy-heptadecanoyl]-isoGlu)-
    AQRAFVEWLLAQ;
    (SEQ ID NO: 90)
    Y-Aib-QGTFTSDYSIYLDK-K(Octadecanoyl-isoGlu-Peg3-Peg3)-AQRAFVEWLLAQ;
    (SEQ ID NO: 91)
    Y-Aib-QGTFTSDYSIYLD-K(eicosanoyl-isoGlu-Peg3-Peg3)-AQRAFVEWLLAQ;
    (SEQ ID NO: 92)
    Y-Aib-QGTFTSDYSIALEK-K([17-carboxy-heptadecanoyl]-isoGlu-Peg3-Peg3)-
    AQRAFVEWLLAQ;
    (SEQ ID NO: 93)
    Y-Aib-QGTFTSDYSIALEK-K(Octadecanoyl-isoGlu-Peg3-Peg3)-AQRAFVEWLLAQ;
    (SEQ ID NO: 94)
    Y-Aib-QGTFTSDYSIALE-K([19-carboxy-nonadecanoyl]-isoGlu-Peg3-Peg3)-
    AQRAFVEWLLAQ;
    (SEQ ID NO: 95)
    Y-Aib-QGTFTSDLSIALE-K(Octadecanoyl-isoGlu-Peg3-Peg3)-AQRAFVEWLLAQ;
    (SEQ ID NO: 25)
    Y-Aib-QGTFTSDYSKYLDE-K(Hexadecanoyl-isoGlu)-AAKDFIEWLESA;
    (SEQ ID NO: 29)
    Y-Aib-QGTFTSDYSIYLDE-K(Hexadecanoyl-isoGlu)-AAKDFVEWLESA;
    (SEQ ID NO: 30)
    Y-Aib-QGTFTSDYSIYLDE-K(Hexadecanoyl-isoGlu)-AAKDFIEWLESA;
    (SEQ ID NO: 38)
    Y-Aib-QGTFTSDYSIYLDE-K(Octadecanoyl-isoGlu-Peg3-Peg3)-AAKEFIEWLESA;
    (SEQ ID NO: 39)
    Y-Aib-QGTFTSDYSIYLDE-K([17-carboxy-heptadecanoyl]-isoGlu-Peg3-Peg3)-
    AAKEFIEWLESA;
    (SEQ ID NO: 26)
    Y-Aib-QGTFTSDYSIYLDS-K(Hexadecanoyl-isoGlu)-AAHDFVEWLLRA;
    (SEQ ID NO: 96)
    Y-Aib-QGTFTSDYSIYLE-K(Octadecanoyl-isoGlu-Peg3-Peg3)-AQRAFVEWLLRA;
    (SEQ ID NO: 97)
    Y-Aib-QGTFTSDYSIYLE-K([17-carboxy-heptadecanoyl]-isoGlu-Peg3-Peg3)-
    AQRAFVEWLLRA;
    (SEQ ID NO: 98)
    Y-Aib-QGTFTSDYSIALD-K(Octadecanoyl-isoGlu-Peg3-Peg3)-AQRAFVNWLVA-Aib;
    (SEQ ID NO: 99)
    Y-Aib-QGTFTSDYSIALD-K(Octadecanoyl-Dapa-Peg3-Peg3)-AQRAFVNWLVA-Aib;
    (SEQ ID NO: 100)
    Y-Aib-QGTFTSDYSIALDK-K([19-carboxy-nonadecanoyl]-isoGlu-Peg3-Peg3)-
    AQRAFVNWLVA-Aib;
    (SEQ ID NO: 40)
    Y-Ac4c-QGTFTSDYSIYLDE-K([19-carboxy-nonadecanoyl]-isoGlu-Peg3-Peg3)-
    AAKEFIEWLESA;
    (SEQ ID NO: 101)
    Y-Ac4c-QGTFTSDYSIALE-K([19-carboxy-nonadecanoyl]-isoGlu-Peg3-Peg3)-
    AQRAFVEWLLAQ;
    or
    (SEQ ID NO: 102)
    Y-Ac4c-QGTFTSDYSIYLDK-K(19-carboxy-heptadecanoyl-isoGlu-Peg3-Peg3)-
    AQRAFVEWLLAQ.
  • The peptide backbone of Formula I may have the sequence:
  • (SEQ ID NO: 1)
    YSQGTFTSDYSKYLDSKAAHDFVEWLLRA;
    (SEQ ID NO: 59)
    YSQGTFTSDYSKYLDΨKAAHDFVEWLLRA;
    (SEQ ID NO: 5)
    Y-Aib-QGTFTSDYSIYLDSKAAHDFVEWLLSA;
    (SEQ ID NO: 9)
    Y-Aib-QGTFTSDYSIYLDEKAAHDFVEWLLSA;
    (SEQ ID NO: 13)
    Y-Aib-QGTFTSDYSIYLDSKAAHDFVEWLLSAGPSSGAPPPS;
    (SEQ ID NO: 14)
    Y-Aib-QGTFTSDYSIYLEKKAAHDFVEWLLSA;
    (SEQ ID NO: 17)
    Y-Aib-QGTFTSDYSIYLESKAAHDFVEWLLSA;
    (SEQ ID NO: 18)
    Y-Aib-QGTFTSDYSIYLDKKAAHDFVEWLLSA;
    (SEQ ID NO: 60)
    Y-Aib-QGTFTSDYSIYLDEΨAAHDFVEWLLSA;
    (SEQ ID NO: 78)
    Y-Aib-QGTFTSDYSIYLDSΨAAHDFVEWLLSAGPSSGAPPPS;
    (SEQ ID NO: 62)
    Y-Aib-QGTFTSDYSKYLDSΨAAHDFVEWLLSA;
    (SEQ ID NO: 7)
    Y-Aib-QGTFTSDYSKALDSKAAHDFVEWLLSA;
    (SEQ ID NO: 8)
    Y-Aib-QGTFTSDYSKYLESKAAHDFVEWLLSA;
    (SEQ ID NO: 10)
    Y-Aib-QGTFTSDYSIYLDSKAAKDFVEWLLSA;
    (SEQ ID NO: 16)
    Y-Aib-QGTFTSDYSIYLDEKAAKDFVEWLLSA;
    (SEQ ID NO: 6)
    Y-Aib-QGTFTSDYSKYLDSKAARDFVEWLLSA;
    (SEQ ID NO: 15)
    Y-Aib-QGTFTSDYSIYLEKKAQKEFVEWLLSA;
    (SEQ ID NO: 21)
    Y-Aib-QGTFTSDYSKYLEKKAQKEFVEWLLSA;
    (SEQ ID NO: 19)
    Y-Aib-QGTFTSDYSIYLEKKAAKEFVEWLLSA;
    (SEQ ID NO: 20)
    Y-Aib-QGTFTSDYSKALDEKAAKEFVEWLLSA;
    (SEQ ID NO: 63)
    Y-Aib-QGTFTSDYSIYLEKΨAAKEFVEWLLSA;
    (SEQ ID NO: 64)
    Y-Aib-QGTFTSDYSIYLEΨKAAKEFVEWLLSA;
    (SEQ ID NO: 65)
    Y-Aib-QGTFTSDYSΨYLEKKAAKEFVEWLLSA;
    (SEQ ID NO: 79)
    Y-Aib-QGTFTSDYSIYLEKΨAAKEFVEWLLSAGPSSGAPPPS;
    (SEQ ID NO: 11)
    Y-Aib-QGTFTSDYSIYLDSKAAHDFVNWLLSA;
    (SEQ ID NO: 80)
    Y-Aib-QGTFTSDYSIYLDKΨAQRAFVEWLLAQGPSSGAPPPS;
    (SEQ ID NO: 81)
    Y-Aib-QGTFTSDYSIALEKΨAQRAFVEWLLAQK;
    (SEQ ID NO: 66)
    Y-Aib-QGTFTSDLSIALEKΨAQRAFVEWLLAQK;
    (SEQ ID NO: 67)
    Y-Aib-QGTFTSDYSKYLDEΨAAKDFIEWLESA;
    (SEQ ID NO: 68)
    Y-Aib-QGTFTSDYSIYLDEΨAAKDFVEWLESA;
    (SEQ ID NO: 69)
    Y-Aib-QGTFTSDYSIYLDEΨAAKDFIEWLESA;
    (SEQ ID NO: 70)
    Y-Aib-QGTFTSDYSIYLDEΨAAKEFIEWLESA;
    (SEQ ID NO: 3)
    Y-Aib-QGTFTSDYSKYLDSKAAHDFVEWLLRA;
    (SEQ ID NO: 12)
    Y-Aib-QGTFTSDYSIYLDSKAAHDFVEWLLRA;
    (SEQ ID NO: 71)
    Y-Aib-QGTFTSDYSIYLDSΨAAHDFVEWLLRA;
    (SEQ ID NO: 72)
    Y-Aib-QGTFTSDYSIYLEKΨAQRAFVEWLLRA;
    (SEQ ID NO: 82)
    Y-Aib-QGTFTSDYSIALDKΨAQRAFVNWLVA-Aib-KPSSGAPPPS;
    (SEQ ID NO: 74)
    Y-Ac4c-QGTFTSDYSIYLDEΨAAKEFIEWLESA;
    (SEQ ID NO: 83)
    Y-Ac4c-QGTFTSDYSIALEKΨAQRAFVEWLLAQK;
    [[Y]] or
    (SEQ ID NO: 84)
    Y-Ac4c-QGTFTSDYSIYLDKΨAQRAFVEWLLAQGPSSGAPPPS.
  • Alternatively, the peptide backbone sequence may differ at up to 5 positions from one of the sequences shown above, within the constraints of Formula I. For the avoidance of doubt, a sequence satisfying the definition of Y1 is regarded as a single position. Typically the compound differs from the reference sequence at only 4 positions in X1 to X29. Thus, if the compound differs from the reference sequence at 5 positions, one of those positions is generally X30 or Y1.
  • In particular, the peptide backbone sequence may differ at up to 5 positions from one of the sequences:
  • (SEQ ID NO: 79)
    Y-Aib-QGTFTSDYSIYLEKΨAAKEFVEWLLSAGPSSGAPPPS;
    (SEQ ID NO: 60)
    Y-Aib-QGTFTSDYSIYLDEΨAAHDFVEWLLSA;
    or
    (SEQ ID NO: 81)
    Y-Aib-QGTFTSDYSIALEKΨAQRAFVEWLLAQK.
  • The peptide backbone of Formula I may have the sequence:
  • (SEQ ID NO: 4)
    YSQGTFTSDYSKYLD-K(Hexadecanoyl-isoGlu)-
    KAAHDFVEWLLRA;
    (SEQ ID NO: 27)
    Y-Aib-QGTFTSDYSIYLDE-K(Hexadecanoyl-isoGlu)-
    AAHDFVEWLLSA;
    (SEQ ID NO: 28)
    Y-Aib-QGTFTSDYSIYLDS-K(Hexadecanoyl-isoGlu)-
    AAHDFVEWLLSAGPSSGAPPPS;
    (SEQ ID NO: 2)
    Y-Aib-QGTFTSDYSKYLDS-K(Hexadecanoyl-isoGlu)-
    AAHDFVEWLLSA;
    (SEQ ID NO: 22)
    Y-Aib-QGTFTSDYSIYLEK-K(Hexadecanoyl-isoGlu)-
    AAKEFVEWLLSA;
    (SEQ ID NO: 23)
    Y-Aib-QGTFTSDYSIYLE-K(Hexadecanoyl-isoGlu)-
    KAAKEFVEWLLSA;
    (SEQ ID NO: 24)
    Y-Aib-QGTFTSDYS-K(Hexadecanoyl-isoGlu)-
    YLEKKAAKEFVEWLLSA;
    (SEQ ID NO: 103)
    Y-Aib-QGTFTSDYSIYLEK-K(Hexadecanoyl-isoGlu)-
    AAKEFVEWLLSAGPSSGAPPPS;
    (SEQ ID NO: 36)
    Y-Aib-QGTFTSDYSIYLEK-K(Octadecanoyl-isoGlu-Peg3-
    Peg3)-AAKEFVEWLLSAGPSSGAPPPS;
    (SEQ ID NO: 37)
    Y-Aib-QGTFTSDYSIYLEK-K([17-carboxy-heptadecanoyl]-
    isoGlu-Peg3-Peg3)-AAKEFVEWLLSAGPSSGAPPPS;
    (SEQ ID NO: 32)
    Y-Aib-QGTFTSDYSIYLDK-K([17-carboxy-heptadecanoyl]-
    isoGlu-Peg3-Peg3)-AQRAFVEWLLAQGPSSGAPPPS;
    (SEQ ID NO: 33)
    Y-Aib-QGTFTSDYSIYLDK-K([17-carboxy-heptadecanoyl]-
    isoGlu)-AQRAFVEWLLAQGPSSGAPPPS;
    (SEQ ID NO: 34)
    Y-Aib-QGTFTSDYSIYLDK-K(Octadecanoyl-isoGlu-Peg3-
    Peg3)-AQRAFVEWLLAQGPSSGAPPPS;
    (SEQ ID NO: 104)
    Y-Aib-QGTFTSDYSIYLD-K(eicosanoyl-isoGlu-Peg3-Peg3)-
    AQRAFVEWLLAQGPSSGAPPPS;
    (SEQ ID NO: 44)
    Y-Aib-QGTFTSDYSIALEK-K([17-carboxy-heptadecanoyl]-
    isoGlu-Peg3-Peg3)-AQRAFVEWLLAQK;
    (SEQ ID NO: 45)
    Y-Aib-QGTFTSDYSIALEK-K(Octadecanoyl-isoGlu-Peg3-
    Peg3)-AQRAFVEWLLAQK;
    (SEQ ID NO: 105)
    Y-Aib-QGTFTSDYSIALE-K([19-carboxy-nonadecanoyl]-
    isoGlu-Peg3-Peg3)-AQRAFVEWLLAQK;
    (SEQ ID NO: 106)
    Y-Aib-QGTFTSDLSIALE-K(Octadecanoyl-isoGlu-Peg3-
    Peg3)-AQRAFVEWLLAQK;
    (SEQ ID NO: 25)
    Y-Aib-QGTFTSDYSKYLDE-K(Hexadecanoyl-isoGlu)-
    AAKDFIEWLESA;
    (SEQ ID NO: 29)
    Y-Aib-QGTFTSDYSIYLDE-K(Hexadecanoyl-isoGlu)-
    AAKDFVEWLESA;
    (SEQ ID NO: 30)
    Y-Aib-QGTFTSDYSIYLDE-K(Hexadecanoyl-isoGlu)-
    AAKDFIEWLESA;
    (SEQ ID NO: 38)
    Y-Aib-QGTFTSDYSIYLDE-K(Octadecanoyl-isoGlu-Peg3-
    Peg3)-AAKEFIEWLESA;
    (SEQ ID NO: 39)
    Y-Aib-QGTFTSDYSIYLDE-K([17-carboxy-heptadecanoyl]-
    isoGlu-Peg3-Peg3)-AAKEFIEWLESA;
    (SEQ ID NO: 26)
    Y-Aib-QGTFTSDYSIYLDS-K(Hexadecanoyl-isoGlu)-
    AAHDFVEWLLRA;
    (SEQ ID NO: 96)
    Y-Aib-QGTFTSDYSIYLE-K(Octadecanoyl-isoGlu-Peg3-
    Peg3)-AQRAFVEWLLRA;
    (SEQ ID NO: 97)
    Y-Aib-QGTFTSDYSIYLE-K([17-carboxy-heptadecanoyl]-
    isoGlu-Peg3-Peg3)-AQRAFVEWLLRA;
    (SEQ ID NO: 107)
    Y-Aib-QGTFTSDYSIALD-K(Octadecanoyl-isoGlu-Peg3-
    Peg3)-AQRAFVNWLVA-Aib-KPSSGAPPPS;
    (SEQ ID NO: 108)
    Y-Aib-QGTFTSDYSIALD-K(Octadecanoyl-Dapa-Peg3-Peg3)-
    AQRAFVNWLVA-Aib-KPSSGAPPPS;
    (SEQ ID NO: 43)
    Y-Aib-QGTFTSDYSIALDK-K([19-carboxy-nonadecanoyl]-
    isoGlu-Peg3-Peg3)-AQRAFVNWLVA-Aib-KPSSGAPPPS;
    (SEQ ID NO: 40)
    Y-Ac4c-QGTFTSDYSIYLDE-K([19-carboxy-nonadecanoyl]-
    isoGlu-Peg3-Peg3)-AAKEFIEWLESA;
    (SEQ ID NO: 109)
    Y-Ac4c-QGTFTSDYSIALE-K([19-carboxy-nonadecanoyl]-
    isoGlu-Peg3-Peg3)-AQRAFVEWLLAQK;
    or
    (SEQ ID NO: 51)
    Y-Ac4c-QGTFTSDYSIYLDK-K(19-carboxy-heptadecanoyl-
    isoGlu-Peg3-Peg3)-AQRAFVEWLLAQGPSSGAPPPS.
  • Certain of the Y1 groups, when present, may provide increased stability in vivo, e.g. in serum, and so may contribute to the half life of the GIP analogue. Without wishing to be bound by theory, it is believed that these groups may help to stabilize the three dimensional conformation of the molecule and/or provide resistance to proteolytic degradation.
  • For example, the Y1 sequences Gly-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser (SEQ ID NO:53), Gly-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Ser (SEQ ID NO: 54), Lys-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser (SEQ ID NO: 55), Lys-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Ser (SEQ ID NO: 56), Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser (SEQ ID NO: 57) and Pro-Ser-Ser-Gly-Ala-Pro-Pro-Ser (SEQ ID NO: 58) have homology with a C-terminal portion of the Exendin-4 molecule and appear to contribute to the stability of the molecule without concomitantly providing significant levels of GLP-1 agonist activity.
  • The invention further provides a nucleic acid encoding a peptide having the sequence X1-X30 of Formula I. Also provided is an expression construct (also known as an expression vector) comprising a nucleic acid of the invention in operable linkage with suitable regulatory elements to direct expression of the peptide, e.g. transcription and translation. The invention also provides a host cell comprising a nucleic acid or expression construct and capable of expressing, and optionally secreting, the peptide.
  • The peptide may itself be a compound of the invention, e.g. when the peptide contains only naturally occurring amino acids (i.e. proteinogenic amino acids), does not contain a residue ψ, and where R1 and R2 are H— and —OH respectively. Alternatively, the peptide may be a precursor of a compound of the invention.
  • The invention further provides a pharmaceutical composition comprising a triple agonist as described herein, or a pharmaceutically acceptable salt or solvate thereof, in admixture with a carrier, preferably a pharmaceutically acceptable carrier. The triple agonist may, for example, be a pharmaceutically acceptable acid addition salt.
  • The pharmaceutical composition may be formulated as a liquid suitable for administration by injection or infusion, or which is formulated to cause slow release of said triple agonist.
  • The invention further provides a therapeutic kit comprising a triple agonist as described herein, and a device comprising a triple agonist as described herein.
  • The invention further provides a triple agonist as described herein, or a pharmaceutically acceptable salt or solvate thereof, for use in a method of medical treatment, e.g. for use in the treatment and/or prevention of a metabolic disorder.
  • The invention further provides the use of a triple agonist as described herein, or a pharmaceutically acceptable salt or solvate thereof, in the preparation of a medicament for the treatment and/or prevention of a metabolic disorder.
  • The invention further provides a method of prevention and or/treatment of a metabolic disorder in a subject, comprising administering a triple agonist as described herein, or a pharmaceutically acceptable salt or solvate thereof, to the subject.
  • The metabolic disorder may be diabetes or a diabetes related disorder, or obesity or an obesity related disorder. The link between obesity and diabetes is well known, so these conditions are not necessarily separate or mutually exclusive.
  • Diabetes related disorders include insulin resistance, glucose intolerance, increased fasting glucose, pre-diabetes, type 1 diabetes, type 2 diabetes, gestational diabetes hypertension, dyslipidemia, bone related disorders and combinations thereof.
  • Diabetes related disorders also include atherosclerosis, arteriosclerosis, coronary heart disease, peripheral artery disease and stroke; or conditions associated with atherogenic dyslipidemia, blood fat disorders, elevated blood pressure, hypertension, a prothrombotic state and a proinflammatory state.
  • Bone related disorders include, but are not limited to osteoporosis and increased risk of bone fracture.
  • The blood fat disorder may be selected from high triglycerides, low HDL cholesterol, high LDL cholesterol, and plaque buildup in artery walls, or a combination thereof.
  • The prothrombotic state may be selected from high fibrinogen levels in the blood and high plasminogen activator inhibitor-1 levels in the blood.
  • The proinflammatory state may be an elevated C-reactive protein level in the blood.
  • Obesity related disorders include obesity linked inflammation, obesity linked gallbladder disease and obesity induced sleep apnea, or may be associated with a condition selected from atherogenic dyslipidemia, blood fat disorders, elevated blood pressure, hypertension, a prothrombotic state, and a proinflammatory state, or a combination thereof.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Unless otherwise defined herein, scientific and technical terms used in this application shall have the meanings that are commonly understood by those of ordinary skill in the art. Generally, nomenclature used in connection with, and techniques of, chemistry, molecular biology, cell and cancer biology, immunology, microbiology, pharmacology, and protein and nucleic acid chemistry, described herein, are those well known and commonly used in the art.
  • Definitions
  • Unless specified otherwise, the following definitions are provided for specific terms, which are used in the above written description.
  • Throughout this specification, the word “comprise” or variations such as “comprises” or “comprising” will be understood to imply the inclusion of a stated integer (or components) or group of integers (or components), but not the exclusion of any other integer (or components) or group of integers (or components).
  • The singular forms “a,” “an,” and “the” include the plurals unless the context clearly dictates otherwise.
  • The term “including” is used to mean “including but not limited to.” “Including” and “including but not limited to” are used interchangeably.
  • The terms “patient,” “subject,” and “individual” may be used interchangeably and refer to either a human or a non-human animal. These terms include mammals such as humans, primates, livestock animals (e.g., bovines, porcines), companion animals (e.g., canines, felines) and rodents (e.g., mice and rats).
  • The term “solvate” in the context of the present invention refers to a complex of defined stoichiometry formed between a solute (in casu, a peptide conjugate or pharmaceutically acceptable salt thereof according to the invention) and a solvent. The solvent in this connection may, for example, be water, ethanol or another pharmaceutically acceptable, typically small-molecular organic species, such as, but not limited to, acetic acid or lactic acid. When the solvent in question is water, such a solvate is normally referred to as a hydrate.
  • The term “agonist” as employed in the context of the invention refers to a substance (ligand) that activates the receptor type in question.
  • Throughout the description and claims the conventional one-letter and three-letter codes for natural (or “proteinogenic”) amino acids are used, as well as generally accepted three letter codes for other (non-natural or “non-proteinogenic”) α-amino acids, such as Aib (α-aminoisobutyric acid), Orn (ornithine) and D-Ala (D-alanine). All amino acid residues in peptides of the invention are preferably of the L-configuration except where explicitly stated.
  • Among sequences disclosed herein are sequences incorporating an “H—” moiety at the amino terminus (N-terminus) of the sequence, and either an “—OH” moiety or an “—NH2” moiety at the carboxy terminus (C-terminus) of the sequence. In such cases, and unless otherwise indicated, an “H—” moiety at the N-terminus of the sequence in question indicates a hydrogen atom (i.e. R1═H—), corresponding to the presence of a free primary or secondary amino group at the N-terminus, while an “—OH” or an “—NH2” moiety at the C-terminus of the sequence (i.e. R2═—OH or —NH2) indicates a carboxy (—COOH) group or an amido (—CONH2) group at the C-terminus, respectively.
  • Other R1 groups are possible at the N-terminus, including pyroglutamic acid (pGlu; (S)-(−)-2-pyrrolidone-5-carboxylic acid), C1-4 alkyl, acetyl, formyl, benzoyl and trifluoroacetyl.
  • Receptor Agonist Activity
  • As mentioned above, the compounds described herein are Glucagon-GIP-GLP 1 dual receptor agonists. That is to say, they have agonist activity at all three of the glucagon receptor, the GIP receptor and the GLP-1 receptor.
  • The term “agonist” as employed in the context of the invention refers to a substance (ligand) that is capable of binding to a particular receptor and activating signaling by that receptor. Thus a GIP receptor agonist is capable of binding to the GIP receptor (designated GIP-R) and activating signaling by that receptor, e.g. by generation of cAMP or inducing Ca2+ release.
  • Agonist activity at the GIP receptor may therefore be measured by assessing GIP receptor signalling, which may may, for example, be measured via cAMP production or Ca2+ release.
  • The cDNA sequence encoding the human GIP receptor has GenBank accession no. BC101673.1 (GI:75516688). The encoded amino acid sequence (including signal peptide) is:
  • (SEQ ID NO: 111)
      1 MTTSPILQLL LRLSLCGLLL QRAETGSKGQ TAGELYQRWE RYRRECQETL AAAEPPSGLA
     61 CNGSFDMYVC WDYAAPNATA RASCPWYLPW HHHVAAGFVL RQCGSDGQWG LWRDHTQCEN
    121 PEKNEAFLDQ RLILERLQVM YTVGYSLSLA TLLLALLILS LFRRLHCTRN YIHINLFTSF
    181 MLRAAAILSR DRLLPRPGPY LGDQALALWN QALAACRTAQ IVTQYCVGAN YTWLLVEGVY
    241 LHSLLVLVGG SEEGHFRYYL LLGWGAPALF VIPWVIVRYL YENTQCWERN EVKAIWWIIR
    301 TPILMTILIN FLIFIRILGI LLSKLRTRQM RCRDYRLRLA RSTLTLVPLL GVHEVVFAPV
    361 TEEQARGALR FAKLGFEIFL SSFQGFLVSV LYCFINKEVQ SEIRRGWHHC RLRRSLGEEQ
    421 RQLPERAFRA LPSGSGPGEV PTSRGLSSGT LPGPGNEASR ELESYC
    (GenBank AAI01674.1 GI: 75516689)”. This may be employed in any assays
    to determine GIP signalling.
  • Similarly the compounds have agonist activity at the GLP-1 receptor (GLP-1-R), i.e. they are capable of binding to the GLP-1 receptor and activating signaling by that receptor, e.g. by generation of cAMP or inducing Ca2+ release. Agonist activity at the GLP-1 receptor may therefore be measured by assessing GLP-1 receptor signalling, which may may, for example, be measured via cAMP production or Ca2+ release.
  • The GLP-1 receptor may have the sequence of the human glucagon-like peptide 1 receptor (GLP-1 R) having primary accession number P43220. The precursor protein (including signal peptide) has primary accession number NP_002053.3; GI:166795283 and has sequence:
  • (SEQ ID NO: 112)
      1 MAGAPGPLRL ALLLLGMVGR AGPRPQGATV SLWETVQKWR EYRRQCQRSL TEDPPPATDL
     61 FCNRTFDEYA CWPDGEPGSF VNVSCPWYLP WASSVPQGHV YRFCTAEGLW LQKDNSSLPW
    121 RDLSECEESK RGERSSPEEQ LLFLYIIYTV GYALSFSALV IASAILLGFR HLHCTRNYIH
    181 LNLFASFILR ALSVFIKDAA LKWMYSTAAQ QHQWDGLLSY QDSLSCRLVF LLMQYCVAAN
    241 YYWLLVEGVY LYTLLAFSVL SEQWIFRLYV SIGWGVPLLF VVPWGIVKYL YEDEGCWTRN
    301 SNMNYWLIIR LPILFAIGVN FLIFVRVICI VVSKLKANLM CKTDIKCRLA KSTLTLIPLL
    361 GTHEVIFAFV MDEHARGTLR FIKLFTELSF TSFQGLMVAI LYCFVNNEVQ LEFRKSWERW
    421 RLEHLHIQRD SSMKPLKCPT SSLSSGATAG SSMYTATCQA SCS.
  • Similarly the compounds have agonist activity at the glucagon receptor (Glu-R), i.e. they are capable of binding to the glucagon receptor and activating signaling by that receptor, e.g. by generation of cAMP or inducing Ca2+ release. Agonist activity at the glucagon receptor may therefore be measured by assessing glucagon receptor signalling, which may, for example, be measured via cAMP production or Ca2+ release.
  • The glucagon receptor may have the sequence of the human glucagon receptor (Glu-R) having primary accession number P47871. The precursor protein (including signal peptide) has primary accession number NP_000151.1; GI:4503947, and has the sequence:
  • (SEQ ID NO: 113)
      1 MPPCQPQRPL LLLLLLLACQ PQVPSAQVMD FLFEKWKLYG DQCHHNLSLL PPPTELVCNR
     61 TFDKYSCWPD TPANTTANIS CPWYLPWHHK VQHRFVFKRC GPDGQWVRGP RGQPWRDASQ
    121 CQMDGEEIEV QKEVAKMYSS FQVMYTVGYS LSLGALLLAL AILGGLSKLH CTRNAIHANL
    181 FASFVLKASS VLVIDGLLRT RYSQKIGDDL SVSTWLSDGA VAGCRVAAVF MQYGIVANYC
    241 WLLVEGLYLH NLLGLATLPE RSFFSLYLGI GWGAPMLFVV PWAVVKCLFE NVQCWTSNDN
    301 MGFWWILRFP VFLAILINFF IFVRIVQLLV AKLRARQMHH TDYKFRLAKS TLTLIPLLGV
    361 HEVVFAFVTD EHAQGTLRSA KLFFDLFLSS FQGLLVAVLY CFLNKEVQSE LRRRWHRWRL
    421 GKVLWEERNT SNHRASSSPG HGPPSKELQF GRGGGSQDSS AETPLAGGLP RLAESPF
  • In all cases, where sequences of precursor proteins are referred to, it should of course be understood that assays may make use of the mature protein, lacking the signal sequence.
  • The compounds of the present invention have at least one GIP, one glucagon, and one GLP-1 biological activity, in particular in treatment of metabolic diseases such as diabetes and obesity. This can be assessed, e.g., in in vivo assays, for example as described in the examples, in which the blood glucose level or another biological activity is determined after a test animal has been treated or exposed to a triple agonist. In particular, compounds of the invention may be capable of improving glycaemic control when adminstered to a diabetic subject. Additionally or alternatively, they may be capable of reducing body weight when administered to an overweight or obese subject. In either case, the effect may be superior to that obtained with an equivalent quantity (by mass, or molar ratio) of wild type human GIP or GLP-1 in comparable subjects when given according to a comparable dosing regime.
  • Activity in in vitro assays may also be used as a measure of the compounds' activity. Typically the compounds have activity at the glucagon, GLP-1 and GIP receptors (designated GCG-R, GLP-1-R and GIP-R respectively). EC50 values may be used as a numerical measure of agonist potency at a given receptor. An EC50 value is a measure of the concentration of a compound required to achieve half of that compound's maximal activity in a particular assay. Thus, for example, a compound having EC50[GLP-1R] lower than the EC50[GLP-1 R] of native GIP in a particular assay may be considered to have higher potency at the GLP-1R than GIP. In some embodiments of the present invention, the EC50 GLP-1-R and/or EC50 GIP-R and/or EC50 GCG-R is below 1.0 nM, below 0.9 nM, below 0.8 nM, below 0.7 nM, below 0.6 nM, below 0.5 nM, below 0.4 nM, below 0.3 nM, below 0.2 nM, below 0.1 nM, below 0.09 nM, below 0.08 nM, below 0.07 nM, below 0.06 nM, below 0.05 nM, below 0.04 nM, below 0.03 nM, below 0.02 nM, below 0.01 nM, below 0.009 nM, below 0.008 nM, below 0.007 nM, below 0.006 nM, or below 0.005 nM, e.g. when assessed using the assay described in Example 2.
  • Lipophilic Group
  • The compound of the invention may comprise a residue ψ, i.e. a residue selected from Lys, Arg, Orn and Cys in which the side chain is conjugated to a lipohilic substituent.
  • Without wishing to be bound by any particular theory, it is thought that the substituent binds plasma proteins (e.g. albumin) in the blood stream, thus shielding the compounds of the invention from enzymatic degradation and renal clearance and thereby enhancing the half-life of the compounds. It may also modulate the potency of the compound, e.g. with respect to the GIP receptor, the glucagon receptor and/or the GLP-1 receptor.
  • The substituent is conjugated to the functional group at the distal end of the side chain from the alpha-carbon. The normal ability of the Lys, Arg, Orn or Cys side chain to participate in interactions mediated by that functional group (e.g. intra- and inter-molecular interactions) may therefore be reduced or completely eliminated by the presence of the substituent. Thus, the overall properties of the compound may be relatively insensitive to changes in the actual amino acid present as residue ψ. Consequently, it is believed that any of the residues Lys, Arg, Orn and Cys may be present at any position where ψ is permitted. However, in certain embodiments, it may be advantageous that the amino acid component of ψ is Lys.
  • Thus, ψ is a residue of Lys, Arg, Orn or Cys in which the side chain is conjugated to a substituent having the formula —Z1 or —Z2—Z1.
  • —Z1 is a fatty chain having at a terminus a connection —X— to ψ or to Z2;
  • wherein
  • —X— is a bond, —CO—, —SO—, or —SO2—;
  • and, optionally, Z1 has a polar group at the end of the chain distal from connection —X—; said polar group comprising a carboxylic acid or a carboxylic acid bioisostere, a phosphonic acid, or a sulfonic acid group;
  • and wherein —Z2—, if present, is a spacer of formula:
  • Figure US20210363213A1-20211125-C00001
  • connecting Z1 to ψ;
  • wherein:
  • each Y is independently —NH, —NR, —S or —O, where R is alkyl, a protecting group or forms a linkage to another part of the spacer Z2;
  • each X is independently a bond, CO—, SO—, or SO2—;
  • with the proviso that when Y is —S, the X to which it is bound is a bond;
  • each V is independently a bivalent organic moiety linking Y and X;
  • and n is 1-10.
  • The group Z1
  • Z1 is a fatty chain having a connection to ψ or to Z2, referred to herein as —X—. —X— may be, for example, a bond, acyl (—CO—), sulfinyl (—SO—), or sulfonyl (—SO2—). When Z1 is bound directly to ψ, that is, when Z2 is not present, preferably —X— is acyl (—CO—), sulfinyl (—SO—), or sulfonyl (—SO2—). Most preferably, —X— is acyl (—CO—).
  • Z1 may further have a polar group, said polar group being located at the end of the chain distal from the connection —X—. In other words, the connection is located at the ω-position with respect to the polar group. The polar group may be bound directly to the terminus of the fatty chain, or may be bound via a linker.
  • Preferably, the polar group is an acidic or weakly acid group, for example a carboxylic acid or a carboxylic acid bioisostere, a phosphonate, or a sulfonate. The polar group may have a pKa of between −2 and 12 in water, more preferably between 1 and 7, more preferably between 3 and 6. Certain preferred polar groups have a pKa of between 4 and 5.
  • For example, and not by way of limitation, the polar group may comprise a carboxylic acid (—COOH) or a carboxylic acid bioisostere, a phosphonic acid (—P(O)(OH)2), or a sulfonic acid (—SO2OH) group.
  • Preferably the polar group, if present, comprises a carboxylic acid or carboxylic acid bioisostere. Suitable carboxylic acid bioisosteres are known in the art. Preferably the bioisostere has a proton having a pKa similar to the corresponding carboxylic acid. Examples of suitable bioisoteres may include, not by way of limitation, tetrazole, acylsulfomides, acylhydroxylamine, and squaric acid derivatives, as shown below (--- indicates the point of attachment):
  • Figure US20210363213A1-20211125-C00002
  • Fatty chain as used herein refers to a moiety comprising a chain of carbon atoms, the carbon atoms being predominantly substituted with hydrogen or hydrogen-like atoms, for example, a hydrocarbon chain. Such fatty chains are often referred to as lipophilic, although it will be appreciated that substitution may alter the lipophilic properties of the overall molecule.
  • The fatty chain may by aliphatic. It may be entirely saturated or may include one or more double or triple bonds. Each double bond, if present, may be in the E or Z configuration. The fatty chain may also have one or more cycloalkylene or heterocycloalkylene moieties in its length, and additionally or alternatively may have one or more arylene or heteroarylene moieties in its length. For example, the fatty chain may incorporate a phenylene or piperazinylene moiety in its length as, for example, shown below (wherein --- represents the points of attachment within the chain).
  • Figure US20210363213A1-20211125-C00003
  • The fatty chain may be derived from a fatty acid, for example, it may be derived from a medium-chain fatty acid (MCFA) with an aliphatic tail of 6-12 carbon atoms, a long-chain fatty acid (LCFA) with an aliphatic tail of 13-21 carbon atoms, or a very long-chain fatty acid (LCFA) with an aliphatic tail of 22 carbon atoms or more. Examples of linear saturated fatty acids from which suitable fatty chains may be derived include tridecylic (tridecanoic) acid, myristic (tetradecanoic) acid, pentadecylic (pentadecanoic) acid, palmitic (hexadecanoic) acid, and margaric (heptadecanoic) acid. Examples of linear unsaturated fatty acids from which suitable fatty chains may be derived include myristoleic acid, palmitoleic acid, sapienic acid and oleic acid.
  • The fatty chain may be connected to ψ or to Z2 by an amide linkage, a sulfinamide linkage, a sulfonamide linkage, or by an ester linkage, or by an ether, thioether or amine linkage. Accordingly, the fatty chain may have, a bond to ψ or to Z2 or an acyl (—CO—), sulfinyl (—SO—), or sulfonyl (—SO2—) group. Preferably, the fatty chain has a terminus having an acyl (—CO—) group and is connected to ψ or Z2 by an amide or ester linkage.
  • In some embodiments, Z1 is a group of formula:

  • A-B-Alk-X—
  • wherein
  • A is hydrogen or a carboxylic acid, a carboxylic acid bioisostere, a phosphonic acid, or a sulfonic acid group;
  • B is a bond or a linker;
  • X is a bond, acyl (—CO—), sulfinyl (—SO—), or sulfonyl (—SO2—); and
  • Alk is a fatty chain that may be optionally substituted with one or more substituents. The fatty chain is preferably 6 to 28 carbon atoms in length (e.g. a C6-28alkylene), more preferably, 12 to 26 carbons in length (e.g. a C12-26alkylene), more preferably, 16 to 22 carbons in length (e.g. C16-22alkylene), and may be saturated or unsaturated. Preferably, Alk is saturated, that is, preferably Alk is alkylene.
  • Optional substituents on the fatty chain may be independently selected from fluoro, C1-4alkyl, preferably methyl; trifluoromethyl, hydroxymethyl, amino, hydroxyl, C1-4alkoxy, preferably methoxy; oxo, and carboxyl, and may be independently located at any point along the chain. In some embodiments, each optional substituent is selected from fluoro, methyl, and hydroxyl. Where more than one substituent is present, substituents may be the same or different. Preferably, the number of substituents is 0 to 3; more preferably the fatty chain is unsubstituted.
  • B may be a bond or a linker. When B is a linker, it may be a cycloalkylene, heterocycloalkylene, C6arylene, or C5-6heteroarylene, or C6arylene-O— or C5-6heteroarylene-O—.
  • When B is phenylene it may, for example, be selected from 1,2-phenylene, 1,3-phenylene, 1,4-phenylene, preferably 1,4-phenylene (so that A-B— is a 4-benzoic acid substituent or 4-benzoic acid bioisostere). When B is phenylene-O—, it may, for example, be selected from 1,2-phenylene-O—, 1,3-phenylene-O—, 1,4-phenylene-O—, preferably 1,4-phenylene-O. Each phenylene of B may be optionally substituted with one or more substituents selected from fluoro, methyl, trifluoromethyl, amino, hydroxyl, and C1-4alkoxy, preferably methoxy. It will be appreciated that substituent identity and position may be selected to subtly alter the pKa of the polar group. Suitable inductively or mesomerically electron-withdrawing or donating groups and their positional effects are known in the art. In some embodiments, B may be C5-6heteroarylene, for example, pyridinylene or thiofuranylene, and may be optionally substituted as described.
  • For example, in some embodiments, A-B— may be selected from:
  • Figure US20210363213A1-20211125-C00004
  • Preferably, is H— or HOOC— and B is a bond.
  • It will be understood that when A is hydrogen, B is a bond and Alk is unsubstituted alkylene, A-B-Alk- is an alkyl chain of formula H3C—(CH2)n—.
  • In some embodiments, Z1 is an acyl group of formula:

  • A-B-Alk-(CO)—
  • or a sulfonyl group of formula:

  • A-B-Alk-(SO2)—.
  • Preferably, Z1 is an acyl group of formula:

  • A-B-alkylene-(CO)—
  • where A and B are as defined above.
  • In some embodiments, A is —COOH and B is a bond. Accordingly, certain preferred Z1 are derived from long-chain saturated α,ω-dicarboxylic acids of formula HOOC—(CH2)12-22—COOH, preferably, long-chain saturated α,ω-dicarboxylic acids having an even number of carbon atoms in the aliphatic chain. In some other embodiments, A is H and B is a bond. Accordingly, certain preferred Z1 are derived from long-chain saturated carboxylic acids of formula HOOC—(CH2)12-22—CH3, preferably, long-chain saturated carboxylic acids having an even number of carbon atoms in the aliphatic chain.
  • For example, and not by way of limitation, Z1 may be:
  • A-B-C16-20alkylene-(CO)— wherein A is H or —COOH and B is a bond, for example:
  • 17-carboxy-heptadecanoyl HOOC—(CH2)16—(CO)—;
  • 19-carboxy-nonadecanoyl HOOC—(CH2)13—(CO)—;
  • Octadecanoyl H3C—(CH2)16—(CO)—;
  • Eicosanoyl H3C—(CH2)18—(CO)—;
  • The carboxylic acid group, if present, may be replaced by a bioisotere as detailed herein.
  • The Group Z2
  • Z2 is an optional spacer that connects Z1 to the side chain of the amino acid component of ψ. At its most general, Z2, if present, is a spacer bound at one terminus by Y, which may be a nitrogen, oxygen or sulfur atom, and at the other terminus by X, which may be a bond or an acyl (—CO—), sulfinyl (—SO—), sulfonyl (—SO2—) or absent. Accordingly, Z2 may be a spacer of formula (--- indicate points of attachment):
  • Figure US20210363213A1-20211125-C00005
  • wherein:
  • Y may be —NH, —NR, —S or —O, where R may be alkyl, a protecting group or may form a linkage to another part of the spacer, with the remaining valency forming a linkage to Z1;
  • X may be a bond, CO—, SO—, or SO2—, with the remaining valency forming a linkage to the side chain of the amino acid component of Y;
  • V is a bivalent organic moiety linking Y and X;
  • and n may be 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10. Where n is 2 or more, each Y, V, and X is independent of every other Y, V, and X.
  • Accordingly, Z2 may be bound at each side by amide, sulfinamide, sulfonamide, or ester linkages or by amino, ether, or thioether linkages depending upon the nature of Y and X and the corresponding linking groups on Z1 and the side chain. Where n is 2 or greater, each V may also be bound to each adjacent V by linkages as described. Preferably, linkages are amides, esters or sulfonamides, most preferably amides. Accordingly, in some embodiments, each Y is —NH or —NR and each X is CO— or SO2—. Most preferably, —X— is acyl (—CO—).
  • In some embodiments, Z2 is a spacer of formula —SA—, —SB—, —SA—SB— or —SB—SA—, wherein SA and SB are as defined below.
  • In some embodiments, Z2 is selected from —SA— or —SB—SA—, that is, [side chain]—Z2Z1 is [side chain]—SA—Z1 or [side chain]—SB—SA—Z1.
  • The Group SA
  • SA may be a single amino acid residue or a residue of an amino acid derivative, especially an amino acid derivative residue having a sulfinyl or sulfonyl in place of the carboxy moiety at the C terminus. Additionally or alternatively, the single amino acid residue may have an oxygen or sulfur atom in place of the nitrogen atom at the N terminus. Preferably, SA is a single amino acid residue.
  • In some embodiments, the amino acid may be selected from γ-Glu, α-Glu, α-Asp, β-Asp, Ala, β-Ala (3-aminopropanoic acid), Dapa (2,3-diaminopropanoic acid), Dab (2,4-diaminobutanoic acid), and Gaba (4-aminobutanoic acid). It will be understood that where more than one carboxylic acid or amino moiety is present, connection may be at any moiety as appropropriate. Any carboxylic acid or amino resides not bound within the residue may be free, that is, present as a free carboxylic acid or primary amine, or may be derivatised. Suitable derivatisation is known in the art. For example, carboxylic acid moieties may be present in SA amino acid residues as esters, for example, as methyl esters. Amino moieties may be present as alkylated amines, for example, methylated, or may be protected as amide or carbamate moieties. Other suitable amino acids include β-Ala (3-aminopropanoic acid) and Gaba (4-aminobutanoic acid) and similar w amino acids.
  • It will be understood that amino acids may be D or L, or a racemic or enantioenriched mixture. In some embodiments, the amino acid is an L-amino acid. In some embodiments, the amino acid is a D-amino acid.
  • In some preferred embodiments, SA has a carboxylic acid substituent, with γ-Glu, α-Glu, α-Asp, and β-Asp, and sulfinyl and sulfonyl derivatives thereof, being preferred. Accordingly, in some embodiments, the amino acid residue is:
  • Figure US20210363213A1-20211125-C00006
  • where —X— is —CO—, —SO—, —SO2—, preferably —CO—, and a is 1 or 2, preferably 2. In some embodiments, the carboxylic acid is an ester, and the amino acid residue is:
  • Figure US20210363213A1-20211125-C00007
  • where —X— is —CO—, —SO—, —SO2—, preferably —CO—, and a is 1 or 2, preferably 2, and R is C1-4alkyl or C6aryl. Preferably R is C1-4alkyl, preferably methyl or ethyl, more preferably ethyl.
  • A preferred SA group bearing a carboxylic acid is γ-Glu.
  • Preferably, SA is selected from Dapa or γ-Glu. Most preferably, SA is γ-Glu.
  • The Group SB
  • SB may be a linker of general formula:
  • Figure US20210363213A1-20211125-C00008
  • wherein PU is a polymeric unit and n is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10. One terminus of the linker SB is an —NH, —NR, —S or —O, wherein R may be alkyl, a protecting group or may form a linkage to another part of the polymeric unit; while the other is a bond or CO—, SO— or SO2—. Accordingly, each polymeric unit PU may be bound at each side by amide, sulfinamide, sulfonamide, or ester linkages or by amino, ether, or thioether linkages depending upon the nature of Y and X and the corresponding linking groups on Z1, SA, and Lys.
  • In some embodiments, each PU may be independently a unit of formula:
  • Figure US20210363213A1-20211125-C00009
  • wherein:
  • Y may be —NH, —NR, —S or —O, wherein R may be alkyl, a protecting group or may form a linkage to another part of the spacer, with the remaining valency forming a linkage to Z1;
  • X may be a bond, CO—, SO—, or SO2—, with the remaining valency forming a linkage to the ψ side chain;
  • and V is a bivalent organic moiety linking Y and X.
  • In some embodiments, V is the α-carbon of a natural or unnatural amino acid, that is V is —CHRAA—, wherein RAA is an amino acid side chain; or V is an optionally substituted C1-6alkylene, or V is a chain comprising one or more units of ethylene glycol in series, also known as PEG chain, for example, —CH2CH2—(OCH2CH2)m—O—(CH2)p—, where m is 0, 1, 2, 3, 4, or 5, and p is 1, 2, 3, 4, or 5; when X is CO—, p is preferably 1, 3, 4, or 5. Optional alkylene substituents include fluoro, methyl, hydroxy, hydroxymethy, and amino.
  • Preferred PU units include:
  • (i). Single amino acid residues: PU i;
  • (ii). Dipeptide residues: PU ii; and
  • (iii). Amino-(PEG)m-carboxylic acid residues: PU iii,
  • and may be present in any combination or order. For example, SB may comprise one or more of each of PU i, PU ii, and PU ii in any order, or may comprise one or more units of PU i, PU ii, and PU i only, or one of more units selected from PU i and PU ii, PU i and PU iii, or PU ii and PU iii.
  • (1). PU i single amino acid residues
  • Each PU i may be independently selected from any natural or unnatural amino acid residue and, for example, may be selected from Gly, Pro, Ala, Val, Leu, lie, Met, Cys, Phe, Tyr, Trp, His, Lys, Arg, Gln, Asn, α-Glu, γ-Glu, Asp, Ser Thr, Dapa, Gaba, Aib, β-Ala, 5-aminopentanoyl, 6-aminohexanoyl, 7-aminoheptanoyl, 8-aminooctanoyl, 9-aminononanoyl, and 10-aminodecanoyl. Preferably, PU i amino acid residues are selected from Gly, Ser, Ala, Thr, and Cys, more preferably from Gly and Ser.
  • In some embodiments, SB is —(PU i)n—, wherein n is 1 to 8, more preferably 5 to 7, most preferably 6. In some preferred embodiments, SB is —(PU i)n—, n is 6 and each PU i is independently selected from Gly or Ser, with a preferred sequence being -Gly-Ser-Gly-Ser-Gly-Gly- (SEQ ID NO: 114).
  • (ii). PU ii dipeptide residues Each PU i may be independently selected from any dipeptide residue comprising two natural or unnatural amino acid residues bound by an amide linkage. Preferred PU ii dipeptide residues include Gly-Gly, Gly-Ser, Ser-Gly, Gly-Ala, Ala-Gly, and Ala-Ala, more preferably Gly-Ser and Gly-Gly.
  • In some embodiments, SB is —(PU ii)n—, wherein n is 2 to 4, more preferably 3, and each PU ii is independently selected from Gly-Ser and Gly-Gly. In some preferred embodiments SB is —(PU ii)n—, n is 3 and each PU i is independently selected from Gly-Ser and Gly-Gly, with a preferred sequence being -(Gly-Ser)-(Gly-Ser)-(Gly-Gly) (SEQ ID NO: 114).
  • Amino acids having stereogenic centres within PU i and PU ii may be racemic, enantioenriched, or enantiopure. In some embodiments, the or each amino acid is independently an L-amino acid.
  • In some embodiments, the or each amino acid is independently a D-amino acid.
  • (iii). PU iii amino-(PEG)m-carboxylic acid residues
  • Each PU iii may be independently a residue of general formula:
  • Figure US20210363213A1-20211125-C00010
  • wherein m is 0, 1, 2, 3, 4, or 5, preferably 1 or 2, and p is 1, 3, 4, or 5, preferably 1.
  • In some embodiments, m is 1 and p is 1, that is, PU iii is a residue of 8-amino-3,6-dioxaoctanoic acid (also known as {2-[2-aminoethoxy]ethoxy}acetic acid and H2N-PEG3-COOH). This residue is referred to herein as -PEG3-.
  • Other, longer, PEG chains are also known in the art. For example, 11-amino-3,6,9-trioxaundecanoic acid (also known as H2N-PEG4-COOH or -PEG4-).
  • In some embodiments, SB is —(PU iii)n—, wherein n is 1 to 3, more preferably 2.
  • Most preferably, SB is -PEG3-PEG3-.
  • Preferred Combinations
  • It will be understood that the above preferences may be independently combined to give preferred —Z1 and —Z2—Z1 moieties.
  • Some preferred —Z1 and —Z2—Z1 moieties are shown below (in each case, -- indicates the point of attachment to the side chain of the amino acid component of ψ:
  • Figure US20210363213A1-20211125-C00011
  • The skilled person will be well aware of suitable techniques for preparing the compounds employed in the context of the invention. For examples of suitable chemistry, see, e.g., WO98/08871, WO00/55184, WO00/55119, Madsen et al. (J. Med. Chem. 2007, 50, 6126-32), and Knudsen et al. 2000 (J. Med Chem. 43, 1664-1669).
  • Clinical Utility
  • The compounds of the invention may provide an attractive treatment option for metabolic diseases including obesity and diabetes mellitus (diabetes). Diabetes comprises a group of metabolic diseases characterized by hyperglycemia resulting from defects in insulin secretion, insulin action, or both. Acute signs of diabetes include excessive urine production, resulting compensatory thirst and increased fluid intake, blurred vision, unexplained weight loss, lethargy, and changes in energy metabolism. However, symptoms are often not severe or may be absent. The chronic hyperglycemia of diabetes is associated with long-term damage, dysfunction, and failure of various organs, notably the eyes, kidneys, nerves, heart and blood vessels. Diabetes is classified into type 1 diabetes, type 2 diabetes and gestational diabetes on the basis on pathogenetic characteristics. Type 1 diabetes accounts for 5-10% of all diabetes cases and is caused by auto-immune destruction of insulin-secreting pancreatic β-cells.
  • Type 2 diabetes accounts for 90-95% of diabetes cases and is a result of a complex set of metabolic disorders. However, symptoms are often not severe or may be absent. Type 2 diabetes is the consequence of endogenous insulin production becoming insufficient to maintain plasma glucose levels below diagnostic thresholds.
  • Gestational diabetes refers to any degree of glucose intolerance identified during pregnancy.
  • Pre-diabetes includes impaired fasting glucose and impaired glucose tolerance and refers to those states that occur when blood glucose levels are elevated but below the levels that are established for the clinical diagnosis for diabetes.
  • A large proportion of people with type 2 diabetes and pre-diabetes are at increased risk of morbidity and mortality due to the high prevalence of additional metabolic risk factors, including abdominal obesity (excessive fat tissue around the abdominal internal organs), atherogenic dyslipidemia (blood fat disorders including high triglycerides, low HDL cholesterol and/or high LDL cholesterol, which foster plaque buildup in artery walls), elevated blood pressure (hypertension) a prothrombotic state (e.g. high fibrinogen or plasminogen activator inhibitor- 1 in the blood), and/or a proinflammatory state (e.g., elevated C-reactive protein in the blood).
  • Conversely, obesity confers an increased risk of developing pre-diabetes, type 2 diabetes as well as, e.g., certain types of cancer, obstructive sleep apnea and gall-bladder disease.
  • Dyslipidemia is associated with increased risk of cardiovascular disease. High Density Lipoprotein (HDL) is of clinical importance since an inverse correlation exists between plasma HDL concentrations and risk of atherosclerotic disease. The majority of cholesterol stored in atherosclerotic plaques originates from LDL and hence an elevated concentration of Low Density Lipoproteins (LDL) is closely associated with atherosclerosis. The HDL/LDL ratio is a clinical risk indictor for atherosclerosis and coronary atherosclerosis in particular.
  • Compounds employed in the context of the invention act as glucagon-GIP-GLP1 triple agonists. The triple agonist may combine the effect of glucagon, e.g., on fat metabolism with the effect of GIP on improved glycemic control and the effect of GLP-1 e.g., on blood glucose levels and food intake. They may therefore act to accelerate elimination of excessive adipose tissue, induce sustainable weight loss, and improve glycemic control. Triple glucagon-GIP-GLP1 agonists may also act to reduce cardiovascular risk factors such as high cholesterol and such as high LDL-cholesterol.
  • The triple agonist compounds of the present invention may therefore be used (alone or in combination) as pharmaceutical agents for preventing weight gain, promoting weight loss, reducing excess body weight or treating obesity (e.g., by control of appetite, feeding, food intake, calorie intake, and/or energy expenditure and lipolysis), including morbid obesity, as well as associated diseases and health conditions including but not limited to obesity linked inflammation, obesity linked gallbladder disease and obesity induced sleep apnea. The compounds may also be used for treatment of insulin resistance, glucose intolerance, pre-diabetes, increased fasting glucose, type 2 diabetes, hypertension, dyslipidemia (or a combination of these metabolic risk factors), atherosclerosis, arteriosclerosis, coronary heart disease, peripheral artery disease and stroke. These are all conditions which may be associated with obesity. However, the effects of the compounds employed in the context of the invention on these conditions may be mediated in whole or in part via an effect on body weight, or may be independent thereof.
  • The triple agonist compounds may thus be used (alone or in combination) for the treatment and/or prevention of any of the diseases, disorders, or conditions described herein, including insulin resistance, glucose intolerance, increased fasting glucose, pre-diabetes, type 1 diabetes, type 2 diabetes, gestational diabetes hypertension, dyslipidemia, or a combination thereof. In certain embodiments, the diabetes related disorder is selected from atherosclerosis, arteriosclerosis, coronary heart disease, peripheral artery disease and stroke; or associated with a condition selected from atherogenic dyslipidemia, blood fat disorders, elevated blood pressure, hypertension, a prothrombotic state, and proinflammatory state, or a combination thereof. In certain embodiments, the blood fat disorder is selected from high triglycerides, low HDL cholesterol, high LDL cholesterol, plaque buildup in artery walls, or a combination thereof. In certain embodiments, the prothrombotic state is selected from high fibrinogen levels in the blood and high plasminogen activator inhibitor-1 levels in the blood. In certain embodiments, the proinflammatory state is an elevated C-reactive protein level in the blood. In certain embodiments, the obesity related disorder is selected from obesity linked inflammation, obesity linked gallbladder disease and obesity induced sleep apnea.
  • The triple agonist compounds may also be used for the treatment and/or prevention of any of the diseases, disorders, or conditions associated with diabetes related osteoporosis including increased risk of bone fractures. The observed increase in fracture risk is likely to be related to impaired bone quality rather than to bone mineral density. The related mechanisms, due at least in part to hyperglycemia, neuropathy, and higher incidence of hypovitaminosis D, are not yet fully understood.
  • The invention provides the use of a triple agonist compound as described, in the manufacture of a medicament for any of the clinical applications described in this specification. Reference to a compound for use in any such method should be construed accordingly.
  • In some embodiments, the invention also provides a therapeutic kit comprising a triple agonist of the invention, optionally in combination with a pharmaceutically acceptable carrier. In some embodiments, the invention provides a device comprising a triple agonist of the invention for delivery of the triple agonist to a subject.
  • Pharmaceutical Compositions
  • The triple agonist compounds of the present invention, or salts or solvates thereof, may be formulated as pharmaceutical compositions prepared for storage or administration, which typically comprise a therapeutically effective amount of a compound employed in the context of the invention, or a salt or solvate thereof, in a pharmaceutically acceptable carrier. In some embodiments, the pharmaceutical composition is formulated as a liquid suitable for administration by injection or infusion, or which is formulated to cause slow release of the triple agonist compound
  • The therapeutically effective amount of a compound of the present invention will depend, e.g., on the route of administration, the type of mammal being treated, and the physical characteristics of the specific mammal under consideration. These factors and their relationship to determining this amount are well known to skilled practitioners in the medical arts. This amount and the method of administration can be tailored to achieve optimal efficacy, and may depend on such factors as weight, diet, concurrent medication and other factors, well known to those skilled in the medical arts. The dosage sizes and dosing regimen most appropriate for human use may be guided by the results obtained by the present invention, and may be confirmed in properly designed clinical trials.
  • An effective dosage and treatment protocol may be determined by conventional means, starting with a low dose in laboratory animals and then increasing the dosage while monitoring the effects, and systematically varying the dosage regimen as well. Numerous factors may be taken into consideration by a clinician when determining an optimal dosage for a given subject. Such considerations are known to the skilled person. The term “pharmaceutically acceptable carrier” includes any of the standard pharmaceutical carriers. Pharmaceutically acceptable carriers for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remington's Pharmaceutical Sciences, Mack Publishing Co. (A. R. Gennaro edit. 1985). For example, sterile saline and phosphate- buffered saline at slightly acidic or physiological pH may be used. Suitable pH buffering agents may be, e.g., phosphate, citrate, acetate, lactate, maleate, tris/hydroxymethyl)aminomethane (TRIS), N-Tris(hydroxymethyl)methyl-3-aminopropanesulphonic acid (TAPS), ammonium bicarbonate, diethanolamine, histidine, which in certain embodiments is a preferred buffer, arginine, lysine, or acetate or mixtures thereof. The term further encompasses any agents listed in the US Pharmacopeia for use in animals, including humans.
  • The term “pharmaceutically acceptable salt” refers to a salt of the compound. Salts include pharmaceutically acceptable salts, such as, e.g., acid addition salts and basic salts. Examples of acid addition salts include hydrochloride salts, citrate salts and acetate salts. Examples of basic salts include salts where the cation is selected from alkali metals, such as sodium and potassium, alkaline earth metals such as calcium, and ammonium ions +N(R3)3(R4), where R3 and R4 independently designate optionally substituted C1-6-alkyl, optionally substituted C2-6-alkenyl, optionally substituted aryl, or optionally substituted heteroaryl. Other examples of pharmaceutically acceptable salts are described in “Remington's Pharmaceutical Sciences”, 17th edition. Ed. Alfonso R. Gennaro (Ed.), Mark Publishing Company, Easton, Pa., U.S.A., 1985 and more recent editions, and in the Encyclopaedia of Pharmaceutical Technology.
  • “Treatment” is an approach for obtaining beneficial or desired clinical results. For purposes of this invention, beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable. “Treatment” may also mean prolonging survival as compared to expected survival if not receiving treatment. “Treatment” is an intervention performed with the intention of preventing the development or altering the pathology of a disorder. Accordingly, “treatment” refers to both therapeutic treatment and prophylactic or preventative measures in certain embodiments. Those in need of treatment include those already with the disorder as well as those in which the disorder is to be prevented. By treatment is meant inhibiting or reducing an increase in pathology or symptoms (e.g. weight gain, hyperglycemia) when compared to the absence of treatment, and is not necessarily meant to imply complete cessation of the relevant condition.
  • The pharmaceutical compositions of the invention may be in unit dosage form. In such form, the composition is divided into unit doses containing appropriate quantities of the active component. The unit dosage form can be a packaged preparation, the package containing discrete quantities of the preparations, for example, packeted tablets, capsules, and powders in vials or ampoules. The unit dosage form can also be a capsule, cachet, or tablet itself, or it can be the appropriate number of any of these packaged forms. It may be provided in single dose injectable form, for example in the form of an injection pen. Compositions may be formulated for any suitable route and means of administration. Pharmaceutically acceptable carriers or diluents include those used in formulations suitable for oral, rectal, nasal or parenteral (including subcutaneous, intramuscular, intravenous, intradermal, and transdermal) administration. The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Subcutaneous or transdermal modes of administration may be particularly suitable for certain of the compounds described herein.
  • Combination Therapy
  • In certain embodiments, a compound of the invention may be administered as part of a combination therapy with at least one other agent for treatment of diabetes, obesity, dyslipidemia, or hypertension.
  • In such cases, the at least two active agents may be given together or separately, and as part of the same pharmaceutical formulation or as separate formulations. Thus, the triple agonist compound (or the salt or solvate thereof) may be used in combination with an antidiabetic agent including but not limited to metformin, a sulfonylurea, a glinide, a DPP-IV inhibitor, a glitazone, or insulin. In certain embodiments, the compound or salt or solvate thereof is used in combination with insulin, DPP-IV inhibitor, sulfonylurea or metformin, particularly sulfonylurea or metformin, for achieving adequate glycemic control. In certain preferred embodiments, the compound or salt or solvate thereof is used in combination with insulin or an insulin analogue for achieving adequate glycemic control. Examples of insulin analogues include but are not limited to LANTUS®, NOVORAPID®, HUMALOG®, NOVOMIX®, ACTRAPHANE HM®, LEVEMIR® and APIDRA®.
  • In certain embodiments, the triple agonist compound or salt or solvate thereof may further be used in combination with one or more of an anti-obesity agent, including but not limited to a glucagon-like peptide receptor 1 agonist, peptide YY or analogue thereof, cannabinoid receptor 1 antagonist, lipase inhibitor, melanocortin receptor 4 agonist, or melanin concentrating hormone receptor 1 antagonist.
  • In certain embodiments, the triple agonist compound or salt or solvate thereof may be used in combination with an anti-hypertension agent, including but not limited to an angiotensin-converting enzyme inhibitor, angiotensin II receptor blocker, diuretics, beta-blocker, or calcium channel blocker.
  • In certain embodiments, the triple agonist compound or salt thereof may be used in combination with an anti-dyslipidemia agent, including but not limited to a statin, a fibrate, a niacin and/or a cholesterol absorption inhibitor.
  • Nucleic Acids, Vectors, and Host Cells
  • The invention provides a nucleic acid encoding a peptide having the sequence X1-X30 of Formula I. Also provided is an expression construct (also known as an expression vector) comprising a nucleic acid of the invention in operable linkage with suitable regulatory elements to direct expression of the peptide, e.g. transcription and translation. The invention also provides a host cell comprising a nucleic acid or expression construct and capable of expressing, and optionally secreting, the peptide.
  • In some embodiments, the invention provides a method of producing a compound of the invention, the method comprising culturing the host cells described above under conditions suitable for expressing the compound and purifying the compound thus produced.
  • The invention also provides a nucleic acid molecule, an expression vector, or a host cell, as described above, for use in a method of medica treatment, and in particular for treatment of the metabolic disorders discussed elsewhere in this specification.
  • Synthesis of Compounds of the Invention
  • A nucleic acid molecule may encode a compound of the invention, a peptide having the amino acid sequence X1-X30 of Formula I, or a peptide which is a precursor of a compound of the invention.
  • Typically, such nucleic acid sequences will be provided as expression constructs wherein the encoding nucleic acid is in functional linkage with appropriate control sequences to direct its expression. The expression construct may be provided in the context of a host cell capable of expressing (and optionally also secreting) the =precursor, or in a cell-free expression system.
  • The invention provides a method of producing a triple agonist of the invention, the method comprising expressing an amino acid precursor of the triple agonist and modifying the precursor to provide the triple agonist. The modification may comprise chemical modification of a Lys, Arg or Cys residue present at a position ψ to introduce the lipophilic moiety, modification of the N- or C-terminus, and/or modification of any other amino acid side chains in the molecule (e.g. to introduce a non-naturally occurring amino acid residue).
  • The compounds of the invention may also be manufactured by standard peptide synthetic methods, e.g. by standard solid-phase or liquid-phase methodology, either stepwise or by fragment assembly, and isolating and purifying the final peptide compound product, or by any combinations of recombinant and synthetic methods.
  • It may be preferable to synthesize the peptide compounds of the invention by means of solid-phase or liquid-phase peptide synthesis. In this context, reference may be made to WO 98/11125 or, inter alia, Fields, G. B. et al., “Principles and Practice of Solid-Phase Peptide Synthesis”; in: Synthetic Peptides, Gregory A. Grant (ed.), Oxford University Press (2nd edition, 2002) and the synthesis examples herein.
  • EXAMPLES
  • The following examples demonstrate certain embodiments of the present invention. However, it is to be understood that these examples neither purport nor are they intended to be wholly definitive as to conditions and scope of this invention. The examples were carried out using standard techniques, which are well known and routine to those of skill in the art, except where otherwise described in detail. The following examples are presented for illustrative purposes only, and should not be construed in any way as limiting the scope of this invention.
  • All publications, patents, and patent applications referred to herein are herein incorporated by reference in their entirety to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety.
  • Example 1
  • The methods used in the instant invention are described below, except where expressly indicated otherwise.
  • General Synthesis of Acylated Triple Agonists Solid phase peptide synthesis was performed on a CEM Liberty Peptide Synthesizer using standard Fmoc chemistry. TentaGel S Ram resin (1 g; 0.25 mmol/g) was swelled in NMP (10 ml) prior to use and transferred between tube and reaction vessel using DCM and NMP.
  • Coupling
  • An Fmoc-amino acid in NMP/DMF/DCM (1:1:1; 0.2 M; 5 ml) was added to the resin in a CEM Discover microwave unit together with COMU/NMP (0.5 M; 2 ml) and DIPEA/DMF (2.0 M; 1 ml). The coupling mixture was heated to 75° C. for 5 min while nitrogen was bubbled through the mixture. The resin was then washed with DMF (4×10 ml).
  • Deprotection
  • Piperidine/NMP (20%; 10 ml) was added to the resin for initial deprotection and the mixture was heated by microwaves (30 sec; 40° C.). The reaction vessel was drained and a second portion of piperidine/DMF (20%; 10 ml) was added and heated (75° C.; 3 min.) again. The resin was then washed with NMP (6×10 ml).
  • Side Chain Acylation
  • Fmoc-Lys(ivDde)-OH or alternatively another amino acid with an orthogonal side chain protective group was introduced at the position of the acylation. The N-terminal of the peptide backbone was then Boc-protected using Boc2O or alternatively by using a Boc-protected amino acid in the last coupling. While the peptide was still attached to the resin, the orthogonal side chain protective group was selectively cleaved using freshly prepared hydrazine hydrate (2-4%) in NMP for 2×15 min. The unprotected lysine side chain was first coupled with Fmoc-Glu-OtBu or another spacer amino acid, which was deprotected with piperidine and acylated with a lipophilic moiety using the peptide coupling methodology as described above. Abbreviations employed are as follows:
  • COMU: 1-[(1-(cyano-2-ethoxy-2-oxoethylideneaminooxy)-dimethylamino-morpholinomethylene)]methanaminium hexafluorophosphate
  • ivDde: 1-(4,4-dimethyl-2,6-dioxocyclohexylidene)3-methyl-butyl
  • Dde: 1-(4,4-dimethyl-2,6-dioxocyclohexylidene)-ethyl
  • DCM: dichloromethane
  • DMF: N,N-dimethylformamide
  • DIPEA: diisopropylethylamine
  • EtOH: ethanol
  • Et2O: diethyl ether
  • HATU: N-[(dimethylamino)-1H-1,2,3-triazol[4,5-b]pyridine-1-ylmethylene]-N-methylmethanaminium hexafluorophosphate N-oxide
  • MeCN: acetonitrile
  • NMP: N-methylpyrrolidone
  • TFA: trifluoroacetic acid
  • TIS: triisopropylsilane
  • Cleavage
  • The resin was washed with EtOH (3×10 ml) and Et2O (3×10 ml) and dried to constant weight at room temperature (r.t.). The crude peptide was cleaved from the resin by treatment with TFA/TIS/water (95/2.5/2.5; 40 ml, 2 h; r.t.). Most of the TFA was removed at reduced pressure and the crude peptide was precipitated and washed three times with diethylether and dried to constant weight at room temperature.
  • HPLC Purification of the Crude Peptide
  • The crude peptide was purified to greater than 90% by preparative reverse phase HPLC using a PerSeptive Biosystems VISION Workstation equipped with a C-18 column (5 cm; 10 μm) and a fraction collector and run at 35 ml/min with a gradient of buffer A (0.1% TFA, aq.) and buffer B (0.1% TFA, 90% MeCN, aq.). Fractions were analyzed by analytical HPLC and MS and relevant fractions were pooled and lyophilized. The final product was characterized by HPLC and MS. The synthesized compounds are shown in Table 1:
  • TABLE 1
    Compound
    No Sequence
     1 H-YSQGTFTSDYSKYLDSKAAHDFVEWLLRA-NH2 (SEQ ID NO: 1)
     2 H-Y-Aib-QGTFTSDYSKYLDS-K(Hexadecanoyl-isoGlu)-
    AAHDFVEWLLSA-NH2 (SEQ ID NO: 2)
     3 H-Y-Aib-QGTFTSDYSKYLDSKAAHDFVEWLLRA-NH2 (SEQ ID NO: 3)
     4 H-YSQGTFTSDYSKYLD-K(Hexadecanoyl-isoGlu)-KAAHDFVEWLLRA-
    NH2 (SEQ ID NO: 4)
     5 H-Y-Aib-QGTFTSDYSIYLDSKAAHDFVEWLLSA-NH2 (SEQ ID NO: 5)
     6 H-Y-Aib-QGTFTSDYSKYLDSKAARDFVEWLLSA-NH2 (SEQ ID NO: 6)
     7 H-Y-Aib-QGTFTSDYSKALDSKAAHDFVEWLLSA-NH2 (SEQ ID NO: 7)
     8 H-Y-Aib-QGTFTSDYSKYLESKAAHDFVEWLLSA-NH2 (SEQ ID NO: 8)
     9 H-Y-Aib-QGTFTSDYSIYLDEKAAHDFVEWLLSA-NH2 (SEQ ID NO: 9)
    10 H-Y-Aib-QGTFTSDYSIYLDSKAAKDFVEWLLSA-NH2 (SEQ ID NO: 10)
    11 H-Y-Aib-QGTFTSDYSIYLDSKAAHDFVNWLLSA-NH2 (SEQ ID NO: 11)
    12 H-Y-Aib-QGTFTSDYSIYLDSKAAHDFVEWLLRA-NH2 (SEQ ID NO: 12)
    13 H-Y-Aib-QGTFTSDYSIYLDSKAAHDFVEWLLSAGPSSGAPPPS-NH2
    (SEQ ID NO: 13)
    14 H-Y-Aib-QGTFTSDYSIYLE-KKAAHDFVEWLLSA-NH2 (SEQ ID NO: 14)
    15 H-Y-Aib-QGTFTSDYSIYLE-KKAAHDFVEWLLSA-NH2 (SEQ ID NO: 14)
    16 H-Y-Aib-QGTFTSDYSIYLEKKAQKEFVEWLLSA-NH2 (SEQ ID NO: 15)
    17 H-Y-Aib-QGTFTSDYSIYLDEKAAKDFVEWLLSA-NH2 (SEQ ID NO: 16)
    18 H-Y-Aib-QGTFTSDYSIYLESKAAHDFVEWLLSA-NH2 (SEQ ID NO: 17)
    19 H-Y-Aib-QGTFTSDYSIYLDKKAAHDFVEWLLSA-NH2 (SEQ ID NO: 18)
    20 H-Y-Aib-QGTFTSDYSIYLEKKAAKEFVEWLLSA-NH2 (SEQ ID NO: 19)
    21 H-Y-Aib-QGTFTSDYSKALDEKAAKEFVEWLLSA-NH2 (SEQ ID NO: 20)
    22 H-Y-Aib-QGTFTSDYSKYLEKKAQKEFVEWLLSA-NH2 (SEQ ID NO: 21)
    23 H-Y-Aib-QGTFTSDYSIYLEK-K(Hexadecanoyl-isoGlu)-
    AAKEFVEWLLSA-NH2 (SEQ ID NO: 22)
    24 H-Y-Aib-QGTFTSDYSIYLE-K(Hexadecanoyl-isoGlu)-
    KAAKEFVEWLLSA-NH2 (SEQ ID NO: 23)
    25 H-Y-Aib-QGTFTSDYS-K(Hexadecanoyl-isoGlu)-
    YLEKKAAKEFVEWLLSA-NH2 (SEQ ID NO: 24)
    26 H-Y-Aib-QGTFTSDYSKYLDE-K(Hexadecanoyl-isoGlu)-
    AAKDFIEWLESA-NH2 (SEQ ID NO: 25)
    27 H-Y-Aib-QGTFTSDYSIYLDS-K(Hexadecanoyl-isoGlu)-
    AAHDFVEWLLRA-NH2 (SEQ ID NO: 26)
    28 H-Y-Aib-QGTFTSDYSIYLDE-K(Hexadecanoyl-isoGlu)-
    AAHDFVEWLLSA-NH2 (SEQ ID NO: 27)
    29 H-Y-Aib-QGTFTSDYSIYLDS-K(Hexadecanoyl-isoGlu)-
    AAHDFVEWLLSAGPSSGAPPPS-NH2 (SEQ ID NO: 28)
    30 H-Y-Aib-QGTFTSDYSIYLDE-K(Hexadecanoyl-isoGlu)-
    AAKDFVEWLESA-NH2 (SEQ ID NO: 29)
    31 H-Y-Aib-QGTFTSDYSIYLDE-K(Hexadecanoyl-isoGlu)-
    AAKDFIEWLESA-NH2 (SEQ ID NO: 30)
    32 H-Y-Aib-QGTFTSDYSIYLEK-K(isoGlu-Hexadecanoyl)-
    AAKEFVEWLLSAGPSSGAPPPS-NH2 (SEQ ID NO: 31)
    33 H-Y-Aib-QGTFTSDYSIYLDK-K([17-carboxy-heptadecanoyl]-isoGlu-
    Peg3-Peg3)-AQRAFVEWLLAQGPSSGAPPPS-NH2 (SEQ ID NO: 32)
    34 H-Y-Aib-QGTFTSDYSIYLDK-K([17-carboxy-heptadecanoyl]-isoGlu)-
    AQRAFVEWLLAQGPSSGAPPPS-NH2 (SEQ ID NO: 33)
    35 H-Y-Aib-QGTFTSDYSIYLDK-K(Octadecanoyl-isoGlu-Peg3-Peg3)-
    AQRAFVEWLLAQGPSSGAPPPS-NH2 (SEQ ID NO: 34)
    36 H-Y-Aib-QGTFTSDYSIYLDK-K(eicosanoyl-isoGlu-Peg3-Peg3)-
    AQRAFVEWLLAQGPSSGAPPPS-NH2 (SEQ ID NO: 35)
    37 H-Y-Aib-QGTFTSDYSIYLEK-K(Octadecanoyl-isoGlu-Peg3-Peg3)-
    AAKEFVEWLLSAGPSSGAPPPS-NH2 (SEQ ID NO: 36)
    38 H-Y-Aib-QGTFTSDYSIYLEK-K([17-carboxy-heptadecanoyl]-isoGlu-
    Peg3-Peg3)-AAKEFVEVVLLSAGPSSGAPPPS-NH2 (SEQ ID NO: 37)
    39 H-Y-Aib-QGTFTSDYSIYLDE-K(Octadecanoyl-isoGlu-Peg3-Peg3)-
    AAKEFIEWLESA-NH2 (SEQ ID NO: 38)
    40 H-Y-Aib-QGTFTSDYSIYLDE-K([17-carboxy-heptadecanoyl]-isoGlu-
    Peg3-Peg3)-AAKEFIEWLESA-NH2 (SEQ ID NO: 39)
    41 H-Y-Ac4c-QGTFTSDYSIYLDE-K([19-carboxy-nonadecanoyl]-isoGlu-
    Peg3-Peg3)-AAKEFIEWLESA-NH2 (SEQ ID NO: 40)
    42 H-Y-Aib-QGTFTSDYSIALDK-K(Octadecanoyl-isoGlu-Peg3-Peg3)-
    AQRAFVNWLVA-Aib-KPSSGAPPPS-NH2 (SEQ ID NO: 41)
    43 H-Y-Aib-QGTFTSDYSIALDK-K(Octadecanoyl-Dapa-Peg3-Peg3)-
    AQRAFVNWLVA-Aib-KPSSGAPPPS-NH2 (SEQ ID NO: 42)
    44 H-Y-Aib-QGTFTSDYSIALDK-K([19-carboxy-nonadecanoyl]-isoGlu-
    Peg3-Peg3)-AQRAFVNWLVA-Aib-KPSSGAPPPS-NH2 (SEQ ID NO: 43)
    45 H-Y-Aib-QGTFTSDYSIALEK-K([17-carboxy-heptadecanoyl]-isoGlu-
    Peg3-Peg3)-AQRAFVEWLLAQK-NH2 (SEQ ID NO: 44)
    46 H-Y-Aib-QGTFTSDYSIALEK-K(Octadecanoyl-isoGlu-Peg3-Peg3)-
    AQRAFVEWLLAQK-NH2 (SEQ ID NO: 45)
    47 H-Y-Aib-QGTFTSDYSIALEK-K[19-carboxy-nonadecanoyl]-isoGlu-Peg3-
    Peg3)-AQRAFVEWLLAQK-NH2 (SEQ ID NO: 46)
    48 H-Y-Aib-QGTFTSDLSIALEK-K(Octadecanoyl-isoGlu-Peg3-Peg3)-
    AQRAFVEWLLAQK-NH2 (SEQ ID NO: 47)
    49 H-Y-Ac4c-QGTFTSDYSIALEK-K([19-carboxy-nonadecanoyl]-isoGlu-
    Peg3-Peg3)-AQRAFVEWLLAQK-NH2 (SEQ ID NO: 48)
    50 H-Y-Aib-QGTFTSDYSIYLEK-K(Octadecanoyl-isoGlu-Peg3-Peg3)-
    AQRAFVEWLLRA-NH2 (SEQ ID NO: 49)
    51 H-Y-Aib-QGTFTSDYSIYLEK-K([17-carboxy-heptadecanoyl]-isoGlu-
    Peg3-Peg3)-AQRAFVEWLLRA-NH2 (SEQ ID NO: 50)
    52 H-Y-Ac4c-QGTFTSDYSIYLDK-K(19-carboxy-heptadecanoyl]-isoGlu-
    Peg3-Peg3)-AQRAFVEWLLAQGPSSGAPPPS-NH2 (SEQ ID NO: 51)
  • Example 2
  • Human GIP Receptor (GIP-R), GLP-1 Receptor (GLP-1-R) and Glucagon Receptor (GCG-R) Activity Assay
  • In vitro effects of peptide conjugates of the invention were assessed by measuring the induction of cAMP following stimulation of the respective receptor by glucagon, GIP, GLP1 or analogues of these as outlined in the invention, using the AlphaSceen® cAMP kit from Perkin-Elmer according to instructions. Briefly, HEK293 cells expressing the human GIP R, GLP-1 R or GCG R (stable cell lines generated through transfection of the cDNA for human GIP R, GLP-1 R or GCG R and selection of stable clones) were seeded at 30,000 cells/well in 96-well microtiter plates coated with 0.01% poly-L-lysine, and grown for 1 day in culture in 200 μl growth medium (DMEM, 10% FCS, Penicillin (100 IU/ml), Streptomycin (100 μg/ml)). On the day of analysis, growth medium was removed and the cells were washed once with 150 μl Tyrode's buffer (Tyrode's Salts (9.6 g/I), 10 mM HEPES, pH 7.4). Cells were then incubated in 100 μl Assay buffer (0.1% W/V Alkali-treated Casein and 100 μM IBMX in Tyrode's Buffer) containing increasing concentrations of control and test compounds for 15 min at 370° C. The Assay buffer was removed and cells are lysed in 80 μl Lysis buffer (0.1% w/v BSA, 5 mM HEPES, 0.3% v/v Tween-20) per well. From each well 10 μl of lysed cells were transferred to a 384-well plate and mixed with 15 μl bead-mix (1 Unit/15 μl anti-cAMP Acceptor Beads, 1 Unit/15 μl Donor Beads, and 1 Unit/15 μl Biotinylated cAMP in Assay Buffer). The plates were mixed and incubated in the dark for an hour at room temperature before measuring using an Envision™ plate reader (Perkin-Elmer).
  • Results were converted into cAMP concentrations using a cAMP standard curve prepared in KRBH buffer containing 0.1% (v/v) DMSO. The resulting cAMP curves were plotted as absolute cAMP concentrations (nM) over log (test compound concentration) and analyzed using the curve fitting program XLfit.
  • Parameters calculated to describe both the potency as well as the agonistic activity of each test compound on the receptors were:
  • EC50, a concentration resulting in a half-maximal elevation of cAMP levels, reflecting the potency of the test compound. The results are summarized in Table 2 and Table 2b. The most comprehensive data are summarized in table 2b.
  • TABLE 2
    Average EC50 values on the GIP-R, GLP1-R and GCG-R
    respectively as compared to control peptides.
    GIP R GLP1 R GCG R
    Compound (EC50 in nM) (EC50 in nM) (EC50 in nM)
    hGIP 0.0038
    Exendin-4 0.0043
    glucagon 0.010*
     1 0.38 0.033 0.013
     2 0.35 0.089 0.046
     3 0.13 0.015 0.022
     5 0.17 0.0092 0.065
     6 0.34 0.0095 0.018
     7 0.42 0.031 0.086
     8 0.20 0.015 0.042
     9 0.056 0.0055 0.097
    10 0.24 0.012 0.10
    11 0.28 0.020 0.11
    12 0.084 0.012 0.076
    13 0.083 0.0099 0.24
    14 0.060 0.013 0.10
    15 0.025 0.016 0.058
    17 0.091 0.011 0.16
    18 0.07 0.021 0.15
    19 0.032 0.013 0.024
    20 0.047 0.0094 0.057
    21 0.18 0.014 0.10
    22 0.18 0.028 0.76
    23 0.13 0.11 0.14
    24 0.10 0.091 0.28
    25 0.25 0.28 0.30
    26 0.070 0.030 0.040
    27 0.12 0.16 0.070
    28 0.054 0.058 0.16
    29 0.060 0.050 0.15
    30 0.15 0.022 0.024
    31 0.087 0.013 0.011
    32 0.044 0.015 0.15
  • TABLE 2b
    Average EC050 values on the GIP-R, GLP1-R and GCG-R
    respectively as compared to control peptides.
    GIP R GLP1 R GCG R
    Compound (EC50 in nM) (EC50 in nM) (EC50 in nM)
    hGIP 0.003 >10 >10
    Exendin-4 >10 0.004 >10
    glucagon >10 0.48 0.009
     1 0.38 0.033 0.013
     2 0.35 0.089 0.046
     3 0.13 0.015 0.022
     5 0.17 0.0092 0.065
     6 0.32 0.0095 0.018
     7 0.5 0.031 0.086
     8 0.2 0.015 0.042
     9 0.056 0.0055 0.097
    10 0.2 0.012 0.1
    11 0.25 0.02 0.11
    12 0.084 0.012 0.076
    13 0.083 0.0099 0.24
    14 0.06 0.013 0.1
    15 0.022 0.014 0.058
    17 0.091 0.013 0.16
    18 0.07 0.021 0.15
    19 0.032 0.013 0.024
    20 0.047 0.0093 0.057
    21 0.18 0.015 0.1
    22 0.18 0.03 0.76
    23 0.13 0.11 0.14
    24 0.097 0.11 0.28
    25 0.19 0.28 0.3
    26 0.07 0.03 0.04
    27 0.099 0.16 0.07
    28 0.054 0.058 0.16
    29 0.064 0.05 0.15
    30 0.15 0.022 0.024
    31 0.087 0.013 0.011
    32 0.04 0.015 0.15
    33 0.0099 0.0093 0.58
    34 0.0073 0.0066 0.11
    35 0.01 0.012 0.011
    37 0.018 NT 0.014
    38 0.064 0.015 0.27
    39 0.015 0.015 0.013
    42 0.0098 0.015 0.032
    43 0.0098 0.014 0.036
    46 0.011 0.016 0.13
    48 0.013 0.013 0.19
    51 0.033 0.026 1.4
    52 0.016 0.0096 1.8
    NT: Not tested
  • It is anticipated that the exemplified compounds of the invention will have activities at the GCG-R that are close to that of native glucagon. At the same time, it is anticipated that they will exhibit strong GLP-1-R activation with EC50 well below 1 nM. Likewise, it is anticipated that these peptides will also exhibit strong GIP-R activity with and EC50 below or just above 1 nM.
  • Example 3
  • Pharmacokinetics of Selected Compounds in Mice
  • Method
  • C57BL/6J mice (males with a body weight of approximately 25 g) were given a single intravenous (i.v.) bolus of each peptide to be tested.
  • Following administration of the selected compounds (100 or 200 nmol/kg), blood samples were drawn 0.08, 0.17, 0.5, 1, 4, 8, 16 and 24 hours post-dose. Blood samples were drawn by sublingual bleeding. The dosing vehicle was a phosphate buffer containing mannitol (pH 7.5).
  • At each sampling time point, samples from two mice were drawn, i.e. 16 mice were included for each compound. The mice were euthanized immediately after blood sampling by cervical dislocation. Plasma samples were analyzed after solid phase extraction (SPE) or precipitation by liquid chromatography mass spectrometry (LC-MS/MS). Mean plasma concentrations were used for calculation of the pharmacokinetic parameters using the non-compartmental approach in Phoenix WinNonlin 6.3. Plasma terminal elimination half-life (T½) was determined as In(2)/λz where λz is the magnitude of the slope of the log linear regression of the log concentration versus time profile during the terminal phase. The results are summarized in Table 3
  • TABLE 3
    Terminal elimination half-life (h) in mice following i.v.
    administration of selected compounds.
    Compound T½ (h.)
    28 7.8
    32 4.1

Claims (36)

1. A glucagon-GLP-1-GIP triple agonist compound having the general formula I:
(I) R1-Tyr-X2-Gln-Gly-Thr-Phe-Thr-Ser-Asp-X10-Ser-X12- X13-Leu-X15-X16-X17-Ala-X19-X20-X21-Phe-X23-X24- Trp-Leu-X27-X28-X29-X30-Y1-R2
wherein
R1 is H—, C1-4 alkyl, acetyl, formyl, benzoyl, trifluoroacetyl or pGlu;
X2 is Aib, Gly, Ala, D-Ala, Ser, N-Me-Ser, Ac3c, Ac4c or Ac5c;
X10 is Tyr or Leu;
X12 is Lys, Ile or ψ;
X13 is Ala, Tyr or Aib;
X15 is Asp or Glu;
X16 is Ser, Glu, Lys or ψ;
X17 is Lys or ψ;
X19 is Gln or Ala;
X20 is Lys, His, Arg or ψ;
X21 is Ala, Asp or Glu;
X23 is Val or lie;
X24 is Asn, Glu or ψ;
X27 is Leu, Glu or Val;
X28 is Ala, Ser, Arg or ψ;
X29 is Aib, Ala, Gln or Lys;
X30 is Lys, Gly, or is absent;
Y1 is Gly-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser, Gly- Pro-Ser-Ser-Gly-Ala-Pro-Pro-Ser, Lys-Pro-Ser-Ser- Gly-Ala-Pro-Pro-Pro-Ser, Lys-Pro-Ser-Ser-Gly-Ala- Pro-Pro-Ser, Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser or Pro-Ser-Ser-Gly-Ala-Pro-Pro-Ser, or is absent;
ψ is a residue of Lys, Arg, Orn or Cys in which the side chain is conjugated to a lipophilic substituent;
and
R2 is —NH2 or —OH;
or a pharmaceutically acceptable salt or solvate thereof.
2. A compound according to claim 1 wherein W is present at one of positions X12, X16 or X17.
3. A compound according to claim 1 or claim 2 wherein the compound contains only one residue ψ.
4. A compound according to any one of claims 1 to 3 wherein the compound possesses one or more of the following sets of residues:
K12 and K12 and Y13; I12 and Y13; K12 and A13; I12 and A13; or ψ12 and Y13;
D15 and S16; D15 and E16; E15 and K16; D15 and ψ16; E15 and S16; or E15 and ψ16;
A19, H20, and D21; A19, K20, and D21; A19, R20, and D21; Q19, K20, and E21; A19, K20, and E21; or Q19, R20, and A21;
I23 and E24; V23 and E24; or V23 and N24;
L27, R28, and A29; L27, S28, and A29; L27, A28, and Q29; E27, S28, and A29; or V27, A28, and Aib29;
E15 and K17;
E15 and ψ17;
E15, ψ17, and Q19;
Q19 and E24;
E16, ψ17, and Q19; or
K16, ψ17, and Q19.
5. A compound according to claim 4 further comprising:
Aib2, Ser2 or Ac4c2; and/or
Tyr10; or
Leu10.
6. A compound according to any one of the preceding claims wherein positions 1 to 29 have the sequence:
YSQGTFTSDYSKYLDSKAAHDFVEWLLRA; YSQGTFTSDYSKYLDΨKAAHDFVEWLLRA; Y-Aib-QGTFTSDYSIYLDSKAAHDFVEWLLSA; Y-Aib-QGTFTSDYSIYLDEKAAHDFVEWLLSA; Y-Aib-QGTFTSDYSIYLEKKAAHDFVEWLLSA; Y-Aib-QGTFTSDYSIYLESKAAHDFVEWLLSA; Y-Aib-QGTFTSDYSIYLDKKAAHDFVEWLLSA; Y-Aib-QGTFTSDYSIYLDEΨAAHDFVEWLLSA; Y-Aib-QGTFTSDYSIYLDSΨAAHDFVEWLLSA; Y-Aib-QGTFTSDYSKYLDSΨAAHDFVEWLLSA; Y-Aib-QGTFTSDYSKALDSKAAHDFVEWLLSA; Y-Aib-QGTFTSDYSKYLESKAAHDFVEWLLSA; Y-Aib-QGTFTSDYSIYLDSKAAKDFVEWLLSA; Y-Aib-QGTFTSDYSIYLDEKAAKDFVEWLLSA; Y-Aib-QGTFTSDYSKYLDSKAARDFVEWLLSA; Y-Aib-QGTFTSDYSIYLEKKAQKEFVEWLLSA; Y-Aib-QGTFTSDYSKYLEKKAQKEFVEWLLSA; Y-Aib-QGTFTSDYSIYLEKKAAKEFVEWLLSA; Y-Aib-QGTFTSDYSKALDEKAAKEFVEWLLSA; Y-Aib-QGTFTSDYSIYLEKΨAAKEFVEWLLSA; Y-Aib-QGTFTSDYSIYLEΨKAAKEFVEWLLSA; Y-Aib-QGTFTSDYSΨYLEKKAAKEFVEWLLSA; Y-Aib-QGTFTSDYSIYLDSKAAHDFVNWLLSA; Y-Aib-QGTFTSDLSIALEKΨAQRAFVEWLLAQK; Y-Aib-QGTFTSDYSKYLDEΨAAKDFIEWLESA; Y-Aib-QGTFTSDYSIYLDEΨAAKDFVEWLESA; Y-Aib-QGTFTSDYSIYLDEΨAAKDFIEWLESA; Y-Aib-QGTFTSDYSIYLDEΨAAKEFIEWLESA; Y-Aib-QGTFTSDYSKYLDSKAAHDFVEWLLRA; Y-Aib-QGTFTSDYSIYLDSKAAHDFVEWLLRA; Y-Aib-QGTFTSDYSIYLDSΨAAHDFVEWLLRA; Y-Aib-QGTFTSDYSIYLEKΨAQRAFVEWLLRA; Y-Aib-QGTFTSDYSIALDKΨAQRAFVNWLVA-Aib; Y-Ac4c-QGTFTSDYSIYLDEΨAAKEFIEWLESA; Y-Ac4c-QGTFTSDYSIALEKΨAQRAFVEWLLAQ; or Y-Ac4c-QGTFTSDYSIYLDKΨAQRAFVEWLLAQ.
7. A compound according to claim 1 which differs at up to 4 positions from any of the sequences listed in claim 6.
8. A compound according to claim 7 which differs at up to 4 positions from one of the sequences:
Y-Aib-QGTFTSDYSIYLEKΨAAKEFVEWLLSA; Y-Aib-QGTFTSDYSIYLDEΨAAHDFVEWLLSA; and Y-Aib-QGTFTSDYSIALEKΨAQRAFVEWLLAQ.
9. A compound according to claim 1 wherein the peptide backbone of Formula I has the sequence:
YSQGTFTSDYSKYLDSKAAHDFVEWLLRA; YSQGTFTSDYSKYLDΨKAAHDFVEWLLRA; Y-Aib-QGTFTSDYSIYLDSKAAHDFVEWLLSA; Y-Aib-QGTFTSDYSIYLDEKAAHDFVEWLLSA; Y-Aib-QGTFTSDYSIYLDSKAAHDFVEWLLSAGPSSGAPPPS; Y-Aib-QGTFTSDYSIYLEKKAAHDFVEWLLSA; Y-Aib-QGTFTSDYSIYLESKAAHDFVEWLLSA; Y-Aib-QGTFTSDYSIYLDKKAAHDFVEWLLSA; Y-Aib-QGTFTSDYSIYLDEΨAAHDFVEWLLSA; Y-Aib-QGTFTSDYSIYLDSΨAAHDFVEWLLSAGPSSGAPPPS; Y-Aib-QGTFTSDYSKYLDSΨAAHDFVEWLLSA; Y-Aib-QGTFTSDYSKALDSKAAHDFVEWLLSA; Y-Aib-QGTFTSDYSKYLESKAAHDFVEWLLSA; Y-Aib-QGTFTSDYSIYLDSKAAKDFVEWLLSA; Y-Aib-QGTFTSDYSIYLDEKAAKDFVEWLLSA; Y-Aib-QGTFTSDYSKYLDSKAARDFVEWLLSA; Y-Aib-QGTFTSDYSIYLEKKAQKEFVEWLLSA; Y-Aib-QGTFTSDYSKYLEKKAQKEFVEWLLSA; Y-Aib-QGTFTSDYSIYLEKKAAKEFVEWLLSA; Y-Aib-QGTFTSDYSKALDEKAAKEFVEWLLSA; Y-Aib-QGTFTSDYSIYLEKΨAAKEFVEWLLSA; Y-Aib-QGTFTSDYSIYLEΨKAAKEFVEWLLSA; Y-Aib-QGTFTSDYSΨYLEKKAAKEFVEWLLSA; Y-Aib-QGTFTSDYSIYLEKΨAAKEFVEWLLSAGPSSGAPPPS; Y-Aib-QGTFTSDYSIYLDSKAAHDFVNWLLSA; Y-Aib-QGTFTSDYSIYLDKΨAQRAFVEWLLAQGPSSGAPPPS; Y-Aib-QGTFTSDYSIALEKΨAQRAFVEWLLAQK; Y-Aib-QGTFTSDLSIALEKΨAQRAFVEWLLAQK; Y-Aib-QGTFTSDYSKYLDEΨAAKDFIEWLESA; Y-Aib-QGTFTSDYSIYLDEΨAAKDFVEWLESA; Y-Aib-QGTFTSDYSIYLDEΨAAKDFIEWLESA; Y-Aib-QGTFTSDYSIYLDEΨAAKEFIEWLESA ; Y-Aib-QGTFTSDYSKYLDSKAAHDFVEWLLRA; Y-Aib-QGTFTSDYSIYLDSKAAHDFVEWLLRA; Y-Aib-QGTFTSDYSIYLDSΨAAHDFVEWLLRA; Y-Aib-QGTFTSDYSIYLEKΨAQRAFVEWLLRA; Y-Aib-QGTFTSDYSIALDKΨAQRAFVNWLVA-Aib-KPSSGAPPPS; Y-Ac4c-QGTFTSDYSIYLDEΨAAKEFIEWLESA; Y-Ac4c-QGTFTSDYSIALEKΨAQRAFVEWLLAQK; or Y-Ac4c-QGTFTSDYSIYLDKΨAQRAFVEWLLAQGPSSGAPPPS.
10. A compound according to claim 1 wherein the peptide backbone of Formula I has a sequence which differs at up to 5 positions from one of the sequences shown in in claim 10.
11. A compound according to claim 10 which differs at up to 5 positions from one of the sequences:
Y-Aib-QGTFTSDYSIYLEKΨAAKEFVEWLLSAGPSSGAPPPS; Y-Aib-QGTFTSDYSIYLDEΨAAHDFVEWLLSA; or Y-Aib-QGTFTSDYSIALEKΨAQRAFVEWLLAQK.
12. A compound according to any one of the preceding claims wherein W is a residue of Lys in which the side chain is conjugated to a lipophilic substituent.
13. A compound according to any one of the preceding claims wherein the lipophilic substituent has the formula —Z1 or —Z2—Z1 wherein:
—Z1 is a fatty chain having at a terminus a connection —X— to ψ or to Z2;
wherein
—X— is a bond, —CO—, —SO—, or —SO2—;
and, optionally, Z1 has a polar group at the end of the chain distal from connection —X—, said polar group comprising a carboxylic acid or a carboxylic acid bioisostere, a phosphonic acid, or a sulfonic acid group;
and wherein —Z2—, if present, is a spacer of formula:
Figure US20210363213A1-20211125-C00012
connecting Z1 to ψ;
wherein:
each Y is independently —NH, —NR, —S or —O, where R is alkyl, a protecting group or forms a linkage to another part of the spacer Z2;
each X is independently a bond, CO—, SO—, or SO2—;
with the proviso that when Y is —S, the X to which it is bound is a bond;
each V is independently a bivalent organic moiety linking Y and X;
and n is 1-10.
14. A compound according to claim 13 wherein the lipophilic substituent has the formula —Z2—Z1.
15. A compound according to claim 12 or claim 14 wherein Z1 is A-B-alkylene-CO— wherein A is H or —COOH and B is a bond.
16. A compound according to claim 15 wherein Z1 is A-B—C16-20alkylene-(CO)—.
17. A compound according to any one of claims 13 to 16 wherein Z1 is:
hexadecanoyl; 17-carboxy-heptadecanoyl [HOOC—(CH2)16—(CO)—];
19-carboxy-nonadecanoyl [HOOC—(CH2)18—(CO)—];
Octadecanoyl [H3C—(CH2)16—(CO)—]; or
Eicosanoyl [H3C—(CH2)18—(CO)—].
18. A compound according to any one of claims 13 to 17 wherein Z2 comprises a residue selected from isoGlu or Dapa.
19. A compound according to claim 13 wherein —Z1 or —Z2—Z1 is:
Figure US20210363213A1-20211125-C00013
20. A compound according to claim 1 wherein positions 1 to 29 have the sequence:
YSQGTFTSDYSKYLD-K(Hexadecanoyl-isoGlu)- KAAHDFVEWLLRA; Y-Aib-QGTFTSDYSIYLDE-K(Hexadecanoyl-isoGlu)- AAHDFVEWLLSA; Y-Aib-QGTFTSDYSIYLDS-K(Hexadecanoyl-isoGlu)- AAHDFVEWLLSA; Y-Aib-QGTFTSDYSKYLDS-K(Hexadecanoyl-isoGlu)- AAHDFVEWLLSA; Y-Aib-QGTFTSDYSIYLEK-K(Hexadecanoyl-isoGlu)- AAKEFVEWLLSA; Y-Aib-QGTFTSDYSIYLE-K(Hexadecanoyl-isoGlu)- KAAKEFVEWLLSA; Y-Aib-QGTFTSDYS-K(Hexadecanoyl-isoGlu)- YLEKKAAKEFVEWLLSA; Y-Aib-QGTFTSDYSIYLEK-K(Hexadecanoyl-isoGlu)- AAKEFVEWLLSA; Y-Aib-QGTFTSDYSIYLEK-K(Octadecanoyl-isoGlu-Peg3- Peg3)-AAKEFVEWLLSA; Y-Aib-QGTFTSDYSIYLEK-K([17-carboxy-heptadecanoyl]- isoGlu-Peg3-Peg3)-AAKEFVEWLLSA; Y-Aib-QGTFTSDYSIYLDK-K([17-carboxy-heptadecanoyl]- isoGlu-Peg3-Peg3)-AQRAFVEWLLAQ; Y-Aib-QGTFTSDYSIYLDK-K([17-carboxy-heptadecanoyl]- isoGlu)-AQRAFVEWLLAQ; Y-Aib-QGTFTSDYSIYLDK-K(Octadecanoyl-isoGlu-Peg3- Peg3)-AQRAFVEWLLAQ; Y-Aib-QGTFTSDYSIYLD-K(eicosanoyl-isoGlu-Peg3-Peg3)- AQRAFVEWLLAQ; Y-Aib-QGTFTSDYSIALEK-K([17-carboxy-heptadecanoyl]- isoGlu-Peg3-Peg3)-AQRAFVEWLLAQ; Y-Aib-QGTFTSDYSIALEK-K(Octadecanoyl-isoGlu-Peg3- Peg3)-AQRAFVEWLLAQ; Y-Aib-QGTFTSDYSIALE-K([19-carboxy-nonadecanoyl]- isoGlu-Peg3-Peg3)-AQRAFVEWLLAQ; Y-Aib-QGTFTSDLSIALE-K(Octadecanoyl-isoGlu-Peg3- Peg3)-AQRAFVEWLLAQ; Y-Aib-QGTFTSDYSKYLDE-K(Hexadecanoyl-isoGlu)- AAKDFIEWLESA; Y-Aib-QGTFTSDYSIYLDE-K(Hexadecanoyl-isoGlu)- AAKDFVEWLESA; Y-Aib-QGTFTSDYSIYLDE-K(Hexadecanoyl-isoGlu)- AAKDFIEWLESA; Y-Aib-QGTFTSDYSIYLDE-K(Octadecanoyl-isoGlu-Peg3- Peg3)-AAKEFIEWLESA; Y-Aib-QGTFTSDYSIYLDE-K([17-carboxy-heptadecanoyl]- isoGlu-Peg3-Peg3)-AAKEFIEWLESA; Y-Aib-QGTFTSDYSIYLDS-K(Hexadecanoyl-isoGlu)- AAHDFVEWLLRA; Y-Aib-QGTFTSDYSIYLE-K(Octadecanoyl-isoGlu-Peg3- Peg3)-AQRAFVEWLLRA; Y-Aib-QGTFTSDYSIYLE-K([17-carboxy-heptadecanoyl]- isoGlu-Peg3-Peg3)-AQRAFVEWLLRA; Y-Aib-QGTFTSDYSIALD-K(Octadecanoyl-isoGlu-Peg3- Peg3)-AQRAFVNWLVA-Aib; Y-Aib-QGTFTSDYSIALD-K(Octadecanoyl-Dapa-Peg3-Peg3)- AQRAFVNWLVA-Aib; Y-Aib-QGTFTSDYSIALDK-K([19-carboxy-nonadecanoyl]- isoGlu-Peg3-Peg3)-AQRAFVNWLVA-Aib; Y-Ac4c-QGTFTSDYSIYLDE-K([19-carboxy-nonadecanoyl]- isoGlu-Peg3-Peg3)-AAKEFIEWLESA; Y-Ac4c-QGTFTSDYSIALE-K([19-carboxy-nonadecanoyl]- isoGlu-Peg3-Peg3)-AQRAFVEWLLAQ; or Y-Ac4c-QGTFTSDYSIYLDK-K(19-carboxy-heptadecanoyl- isoGlu-Peg3-Peg3)-AQRAFVEWLLAQ.
21. A compound according to claim 1 wherein the peptide backbone of Formula I has the sequence:
YSQGTFTSDYSKYLD-K(Hexadecanoyl-isoGlu)- KAAHDFVEWLLRA; Y-Aib-QGTFTSDYSIYLDE-K(Hexadecanoyl-isoGlu)- AAHDFVEWLLSA; Y-Aib-QGTFTSDYSIYLDS-K(Hexadecanoyl-isoGlu)- AAHDFVEWLLSAGPSSGAPPPS; Y-Aib-QGTFTSDYSKYLDS-K(Hexadecanoyl-isoGlu)- AAHDFVEWLLSA; Y-Aib-QGTFTSDYSIYLEK-K(Hexadecanoyl-isoGlu)- AAKEFVEWLLSA; Y-Aib-QGTFTSDYSIYLE-K(Hexadecanoyl-isoGlu)- KAAKEFVEWLLSA; Y-Aib-QGTFTSDYS-K(Hexadecanoyl-isoGlu)- YLEKKAAKEFVEWLLSA; Y-Aib-QGTFTSDYSIYLEK-K(Hexadecanoyl-isoGlu)- AAKEFVEWLLSAGPSSGAPPPS; Y-Aib-QGTFTSDYSIYLEK-K(Octadecanoyl-isoGlu-Peg3- Peg3)-AAKEFVEWLLSAGPSSGAPPPS; Y-Aib-QGTFTSDYSIYLEK-K([17-carboxy-heptadecanoyl]- isoGlu-Peg3-Peg3)-AAKEFVEWLLSAGPSSGAPPPS; Y-Aib-QGTFTSDYSIYLDK-K([17-carboxy-heptadecanoyl]- isoGlu-Peg3-Peg3)-AQRAFVEWLLAQGPSSGAPPPS; Y-Aib-QGTFTSDYSIYLDK-K([17-carboxy-heptadecanoyl]- isoGlu)-AQRAFVEWLLAQGPSSGAPPPS; Y-Aib-QGTFTSDYSIYLDK-K(Octadecanoyl-isoGlu-Peg3- Peg3)-AQRAFVEWLLAQGPSSGAPPPS; Y-Aib-QGTFTSDYSIYLD-K(eicosanoyl-isoGlu-Peg3- Peg3)-AQRAFVEWLLAQGPSSGAPPPS; Y-Aib-QGTFTSDYSIALEK-K([17-carboxy-heptadecanoyl]- isoGlu-Peg3-Peg3)-AQRAFVEWLLAQK; Y-Aib-QGTFTSDYSIALEK-K(Octadecanoyl-isoGlu-Peg3- Peg3)-AQRAFVEWLLAQK; Y-Aib-QGTFTSDYSIALE-K([19-carboxy-nonadecanoyl]- isoGlu-Peg3-Peg3)-AQRAFVEWLLAQK; Y-Aib-QGTFTSDLSIALE-K(Octadecanoyl-isoGlu-Peg3- Peg3)-AQRAFVEWLLAQK; Y-Aib-QGTFTSDYSKYLDE-K(Hexadecanoyl-isoGlu)- AAKDFIEWLESA; Y-Aib-QGTFTSDYSIYLDE-K(Hexadecanoyl-isoGlu)- AAKDFVEWLESA; Y-Aib-QGTFTSDYSIYLDE-K(Hexadecanoyl-isoGlu)- AAKDFIEWLESA; Y-Aib-QGTFTSDYSIYLDE-K(Octadecanoyl-isoGlu-Peg3- Peg3)-AAKEFIEWLESA; Y-Aib-QGTFTSDYSIYLDE-K([17-carboxy-heptadecanoyl]- isoGlu-Peg3-Peg3)-AAKEFIEWLESA; Y-Aib-QGTFTSDYSIYLDS-K(Hexadecanoyl-isoGlu)- AAHDFVEWLLRA; Y-Aib-QGTFTSDYSIYLE-K(Octadecanoyl-isoGlu-Peg3- Peg3)-AQRAFVEWLLRA; Y-Aib-QGTFTSDYSIYLE-K([17-carboxy-heptadecanoyl]- isoGlu-Peg3-Peg3)-AQRAFVEWLLRA; Y-Aib-QGTFTSDYSIALD-K(Octadecanoyl-isoGlu-Peg3- Peg3)-AQRAFVNWLVA-Aib-KPSSGAPPPS; Y-Aib-QGTFTSDYSIALD-K(Octadecanoyl-Dapa-Peg3-Peg3)- AQRAFVNWLVA-Aib-KPSSGAPPPS; Y-Aib-QGTFTSDYSIALDK-K([19-carboxy-nonadecanoyl]- isoGlu-Peg3-Peg3)-AQRAFVNWLVA-Aib-KPSSGAPPPS; Y-Ac4c-QGTFTSDYSIYLDE-K([19-carboxy-nonadecanoyl]- isoGlu-Peg3-Peg3)-AAKEFIEWLESA; Y-Ac4c-QGTFTSDYSIALE-K([19-carboxy-nonadecanoyl]- isoGlu-Peg3-Peg3)-AQRAFVEWLLAQK; or Y-Ac4c-QGTFTSDYSIYLDK-K(19-carboxy-heptadecanoyl- isoGlu-Peg3-Peg3)-AQRAFVEWLLAQGPSSGAPPPS
22. A nucleic acid encoding a peptide having the sequence of Formula I.
23. An expression construct comprising a nucleic acid according to claim 22.
24. A host cell comprising a nucleic acid according to 22 or an expression construct according to claim 23 and capable of expressing, and optionally secreting, the peptide.
25. A pharmaceutical composition comprising a triple agonist compound according to any one of claims 1 to 21, or a pharmaceutically acceptable salt or solvate thereof, in admixture with a carrier.
26. A pharmaceutical composition according to claim 25 formulated as a liquid suitable for administration by injection or infusion, or formulated to cause slow release of said triple agonist compound.
27. A triple agonist compound according to any one of claims 1 to 21, or a pharmaceutically acceptable salt or solvate thereof, for use in a method of medical treatment.
28. A triple agonist compound according to any one of claims 1 to 21, or a pharmaceutically acceptable salt or solvate thereof, for use in a method of treatment and/or prevention of a metabolic disorder.
29. A triple agonist compound for use according to claim 28 wherein the metabolic disorder is diabetes or a diabetes related disorder, or obesity or an obesity related disorder.
30. A triple agonist compound for use according to claim 29 wherein the diabetes related disorder is insulin resistance, glucose intolerance, increased fasting glucose, pre-diabetes, type 1 diabetes, type 2 diabetes, gestational diabetes hypertension, dyslipidemia, bone related disorder or a combination thereof.
31. A GIP analogue or pharmaceutically acceptable salt or solvate thereof for use according to claim 30 wherein the diabetes-related disorder is osteoporosis, including increased risk of bone fracture.
32. A triple agonist compound for use according to claim 29 wherein the diabetes related disorder is atherosclerosis, arteriosclerosis, coronary heart disease, peripheral artery disease, stroke; or is a condition associated with atherogenic dyslipidemia, a blood fat disorder, elevated blood pressure, hypertension, a prothrombotic state, or a proinflammatory state.
33. A triple agonist compound for use according to claim 32 wherein the blood fat disorder is high triglycerides, low HDL cholesterol, high LDL cholesterol, plaque buildup in artery walls, or a combination thereof.
34. A triple agonist compound for use according to claim 32 wherein the prothrombotic state comprises high fibrinogen levels in the blood or high plasminogen activator inhibitor-1 levels in the blood.
35. A triple agonist compound for use according to claim 32 wherein the proinflammatory state comprises an elevated C-reactive protein level in the blood.
36. A triple agonist compound for use according to claim 32 wherein the obesity related disorder is obesity linked inflammation, obesity linked gallbladder disease or obesity induced sleep apnea, or may be associated with a condition selected from atherogenic dyslipidemia, blood fat disorders, elevated blood pressure, hypertension, a prothrombotic state, and a proinflammatory state, or a combination thereof.
US17/388,328 2013-11-06 2021-07-29 Glucagon-glp-1-gip triple agonist compounds Abandoned US20210363213A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/388,328 US20210363213A1 (en) 2013-11-06 2021-07-29 Glucagon-glp-1-gip triple agonist compounds

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201361900933P 2013-11-06 2013-11-06
PCT/EP2014/073971 WO2015067716A1 (en) 2013-11-06 2014-11-06 Glucagon-glp-1-gip triple agonist compounds
US201615034784A 2016-05-05 2016-05-05
US16/151,603 US11111285B2 (en) 2013-11-06 2018-10-04 Glucagon-GLP-1-GIP triple agonist compounds
US17/388,328 US20210363213A1 (en) 2013-11-06 2021-07-29 Glucagon-glp-1-gip triple agonist compounds

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US16/151,603 Continuation US11111285B2 (en) 2013-11-06 2018-10-04 Glucagon-GLP-1-GIP triple agonist compounds

Publications (1)

Publication Number Publication Date
US20210363213A1 true US20210363213A1 (en) 2021-11-25

Family

ID=51999398

Family Applications (3)

Application Number Title Priority Date Filing Date
US15/034,784 Active US10131702B2 (en) 2013-11-06 2014-11-06 Glucagon-GLP-1-GIP triple agonist compounds
US16/151,603 Active US11111285B2 (en) 2013-11-06 2018-10-04 Glucagon-GLP-1-GIP triple agonist compounds
US17/388,328 Abandoned US20210363213A1 (en) 2013-11-06 2021-07-29 Glucagon-glp-1-gip triple agonist compounds

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US15/034,784 Active US10131702B2 (en) 2013-11-06 2014-11-06 Glucagon-GLP-1-GIP triple agonist compounds
US16/151,603 Active US11111285B2 (en) 2013-11-06 2018-10-04 Glucagon-GLP-1-GIP triple agonist compounds

Country Status (12)

Country Link
US (3) US10131702B2 (en)
EP (1) EP3066117B1 (en)
JP (2) JP2017503474A (en)
KR (1) KR102310392B1 (en)
CN (1) CN105829339B (en)
AU (1) AU2014345570B2 (en)
BR (1) BR112016009995B1 (en)
CA (1) CA2929107C (en)
EA (1) EA035688B1 (en)
MX (1) MX369770B (en)
TR (1) TR201902516T4 (en)
WO (1) WO2015067716A1 (en)

Families Citing this family (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
UA116217C2 (en) 2012-10-09 2018-02-26 Санофі Exendin-4 derivatives as dual glp1/glucagon agonists
AU2013366691A1 (en) 2012-12-21 2015-07-09 Sanofi Exendin-4 derivatives
SI3004155T1 (en) 2013-05-28 2022-02-28 Takeda Pharmaceutical Company Limited Peptide compound
EP3065767B1 (en) 2013-11-06 2020-12-30 Zealand Pharma A/S Gip-glp-1 dual agonist compounds and methods
MX369770B (en) * 2013-11-06 2019-11-21 Zealand Pharma As Glucagon-glp-1-gip triple agonist compounds.
TW201609799A (en) 2013-12-13 2016-03-16 賽諾菲公司 Dual GLP-1/GIP receptor agonists
TW201609796A (en) 2013-12-13 2016-03-16 賽諾菲公司 Non-acylated EXENDIN-4 peptide analogues
TW201609797A (en) 2013-12-13 2016-03-16 賽諾菲公司 Dual GLP-1/glucagon receptor agonists
EP3080150B1 (en) 2013-12-13 2018-08-01 Sanofi Exendin-4 peptide analogues as dual glp-1/gip receptor agonists
TW201625668A (en) 2014-04-07 2016-07-16 賽諾菲公司 Exendin-4 derivatives as peptidic dual GLP-1/glucagon receptor agonists
TW201625669A (en) 2014-04-07 2016-07-16 賽諾菲公司 Peptidic dual GLP-1/glucagon receptor agonists derived from Exendin-4
TW201625670A (en) 2014-04-07 2016-07-16 賽諾菲公司 Dual GLP-1/glucagon receptor agonists derived from EXENDIN-4
US9932381B2 (en) 2014-06-18 2018-04-03 Sanofi Exendin-4 derivatives as selective glucagon receptor agonists
CA2965732A1 (en) 2014-10-29 2016-05-06 Zealand Pharma A/S Gip agonist compounds and methods
AR105319A1 (en) 2015-06-05 2017-09-27 Sanofi Sa PROPHARMS THAT INCLUDE A DUAL AGONIST GLU-1 / GLUCAGON CONJUGATE HIALURONIC ACID CONNECTOR
WO2016198624A1 (en) 2015-06-12 2016-12-15 Sanofi Exendin-4 derivatives as trigonal glp-1/glucagon/gip receptor agonists
TW201706291A (en) 2015-07-10 2017-02-16 賽諾菲公司 New EXENDIN-4 derivatives as selective peptidic dual GLP-1/glucagon receptor agonists
TWI622596B (en) 2015-10-26 2018-05-01 美國禮來大藥廠 Glucagon receptor agonists
JP6995042B2 (en) * 2016-05-24 2022-02-04 武田薬品工業株式会社 Peptide compound
AR110300A1 (en) * 2016-12-02 2019-03-13 Sanofi Sa COMPOUNDS AS TRIGONAL PEPTIDE AGONISTS OF GLP1 / GLUCAGÓN / GIP RECEPTORS
AR110299A1 (en) 2016-12-02 2019-03-13 Sanofi Sa CONJUGATES UNDERSTANDING A DUAL GLP-1 / GLUCAGON AGONIST, A CONNECTOR AND Hyaluronic Acid
AR110301A1 (en) 2016-12-02 2019-03-13 Sanofi Sa COMPOUNDS AS GLP1 / GLUCAGÓN / GIP RECEPTING AGENISTS
EP3551202B1 (en) 2016-12-06 2024-01-24 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods of enhancing the potency of incretin-based drugs in subjects in need thereof
JP6563614B1 (en) * 2016-12-09 2019-08-21 ジーランド・ファーマ・ア/エス Acylated GLP-1 / GLP-2 dual agonist
JOP20180028A1 (en) 2017-03-31 2019-01-30 Takeda Pharmaceuticals Co Peptide compound
AR113486A1 (en) * 2017-12-21 2020-05-06 Lilly Co Eli INCRETINE ANALOGUES AND ITS USES
TWI810937B (en) * 2017-12-21 2023-08-01 美商美國禮來大藥廠 Incretin analogs and uses thereof
US11560402B2 (en) 2018-04-10 2023-01-24 Sanofi-Aventis Deutschland Gmbh Method for cleavage of solid phase-bound peptides from the solid phase
ES2928207T3 (en) 2018-04-10 2022-11-16 Sanofi Aventis Deutschland Synthesis of lixisenatide with hooding
CA3097939A1 (en) 2018-05-04 2019-11-07 Novo Nordisk A/S Gip derivatives and uses thereof
UY38249A (en) 2018-05-30 2019-12-31 Sanofi Sa CONJUGATED PRODUCTS INCLUDING A TRIPLE GLP-1 / GLUCAGON / GIP RECEPTOR AGONIST, A CONNECTOR AND HYALURONIC ACID
JP2022515229A (en) * 2018-12-21 2022-02-17 ハンミ ファーマシューティカル カンパニー リミテッド A pharmaceutical composition comprising insulin and a triple active agent having activity on all of glucagon, GLP-1 and GIP receptors.
JP2022516439A (en) 2018-12-21 2022-02-28 ジエンス ヘンルイ メデイシンカンパニー リミテッド Bispecific protein
PE20221049A1 (en) 2019-08-19 2022-06-30 Lilly Co Eli METHODS FOR PREPARING INCRETIN ANALOGS
CN110684082B (en) * 2019-10-08 2021-12-10 江苏诺泰澳赛诺生物制药股份有限公司 GIP and GLP-1 dual-agonist polypeptide compound, pharmaceutically acceptable salt and application
PE20221168A1 (en) 2019-11-11 2022-07-25 Boehringer Ingelheim Int NPY2 RECEPTOR AGONISTS
AR120714A1 (en) 2019-12-18 2022-03-09 Lilly Co Eli INCRETIN ANALOGS AND THEIR USES
CN111040022B (en) * 2019-12-23 2021-12-14 万新医药科技(苏州)有限公司 Triplex agonists directed to glucagon-like peptide-1 receptor, glucagon receptor, and pepstatin receptor
CR20230074A (en) 2020-08-07 2023-04-19 Boehringer Ingelheim Int Soluble npy2 receptor agonists
JP2023537895A (en) * 2020-08-14 2023-09-06 ハンミ ファーマシューティカル カンパニー リミテッド Pharmaceutical composition containing triple active long-acting conjugate as an active ingredient
CA3188884A1 (en) * 2020-08-14 2022-02-17 Seungjae Baek Hypotensive pharmaceutical composition comprising triple activator having activity for all of glucagon, glp-1, and gip receptors
WO2022065898A1 (en) * 2020-09-25 2022-03-31 한미약품 주식회사 Pharmaceutical composition for preventing or treating bone diseases, comprising triple agonist or conjugate thereof having activity with respect to all of glucagon, glp-1 and glp receptors
CN116583533A (en) * 2020-10-16 2023-08-11 韩美药品株式会社 GIP derivative, long-acting conjugate thereof and pharmaceutical composition comprising the same
KR20220050821A (en) * 2020-10-16 2022-04-25 한미약품 주식회사 Pharmaceutical composition for preventing or treating vasculitis comprising glucagon/GLP-1/GIP triple agonist or long acting conjugate thereof
JP2023546088A (en) * 2020-10-16 2023-11-01 ハンミ ファーマシューティカルズ カンパニー リミテッド A pharmaceutical composition for the prevention or treatment of lupus-related diseases comprising a glucagon/GLP-1/GIP triple agonist or a long-acting conjugate thereof
JP2023550594A (en) 2020-10-30 2023-12-04 ノヴォ ノルディスク アー/エス GLP-1, GIP, and glucagon receptor triple agonist
EP4249505A1 (en) 2020-12-23 2023-09-27 Zhejiang Doer Biologics Co., Ltd. Long-acting glucagon derivative
KR20220092442A (en) * 2020-12-24 2022-07-01 한미약품 주식회사 Novel trigonal glucagon/GLP-1/GIP receptor agonist and use thereof
MX2023008330A (en) 2021-01-20 2024-01-18 Viking Therapeutics Inc Compositions and methods for the treatment of metabolic and liver disorders.
CN117062618A (en) 2021-05-26 2023-11-14 联邦生物科技(珠海横琴)有限公司 Multiple agonists and uses thereof
CN115572326A (en) * 2021-06-21 2023-01-06 广东东阳光药业有限公司 Triplex agonists of GLP-1, GCG and GIP receptors
WO2023228156A1 (en) * 2022-05-27 2023-11-30 D&D Pharmatech Inc. Peptide compositions and methods of use threof
WO2024067662A1 (en) * 2022-09-28 2024-04-04 广东东阳光药业股份有限公司 Glp-1/gcg/gip triple-receptor agonist and use thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10093713B2 (en) * 2013-11-06 2018-10-09 Zealand Pharma A/S GIP-GLP-1 dual agonist compounds and methods
US11111285B2 (en) * 2013-11-06 2021-09-07 Zealand Pharma A/S Glucagon-GLP-1-GIP triple agonist compounds

Family Cites Families (170)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4288627A (en) 1980-02-12 1981-09-08 Phillips Petroleum Company Oxidation of thiols employing cobalt molybdate/triethylamine catalyst
NZ202757A (en) 1981-12-23 1985-11-08 Novo Industri As Peptides and medicaments
US5118666A (en) 1986-05-05 1992-06-02 The General Hospital Corporation Insulinotropic hormone
US5120712A (en) 1986-05-05 1992-06-09 The General Hospital Corporation Insulinotropic hormone
US5614492A (en) 1986-05-05 1997-03-25 The General Hospital Corporation Insulinotropic hormone GLP-1 (7-36) and uses thereof
CA2073856C (en) 1990-01-24 2002-12-03 Douglas I. Buckley Glp-1 analogs useful for diabetes treatment
US5545618A (en) 1990-01-24 1996-08-13 Buckley; Douglas I. GLP-1 analogs useful for diabetes treatment
BR9106435A (en) 1990-05-09 1993-05-04 Novo Nordisk As CELLULLASE PREPARATION, ENZYME DEMONSTRATING ANDDOGLUCANASE ACTIVITY, ENDOGLUCANASE ENZYME, DNA CONSTRUCTION, CELL EXPRESSION VECTOR, PROCESS FOR PRODUCING AN ENDOGLUCANASE ENZYME, ADDITIVE DETERGENT COMPOSITION, AND PROCESS TO REDUCE THE RATE AT WHICH CELLULOSE CONTAINING TISSUES BECOME ROUGH, PROVIDE COLOR LIGHTENING OF TISSUES CONTAINING COLORED CELLULOSE, PROVIDES A LOCAL COLOR VARIATION OF TISSUES CONTAINING COLORED, AND IMPROVES PULP DRAINAGE PROPERTIES
DK36392D0 (en) 1992-03-19 1992-03-19 Novo Nordisk As USE OF CHEMICAL COMPOUND
US5846747A (en) 1992-03-25 1998-12-08 Novo Nordisk A/S Method for detecting glucagon-like peptide-1 antagonists and agonists
DK39892D0 (en) 1992-03-25 1992-03-25 Bernard Thorens PEPTIDE
US5424286A (en) 1993-05-24 1995-06-13 Eng; John Exendin-3 and exendin-4 polypeptides, and pharmaceutical compositions comprising same
WO1995005848A1 (en) 1993-08-24 1995-03-02 Novo Nordisk A/S Protracted glp-1
KR100429966B1 (en) 1993-09-07 2004-05-04 아밀린 파마슈티칼스, 인크. Composition For Reugulating Gastrointestinal Motility
US5705483A (en) 1993-12-09 1998-01-06 Eli Lilly And Company Glucagon-like insulinotropic peptides, compositions and methods
US5512549A (en) 1994-10-18 1996-04-30 Eli Lilly And Company Glucagon-like insulinotropic peptide analogs, compositions, and methods of use
US5523449A (en) 1995-05-17 1996-06-04 Bayer Corporation Process for preparing phosphorodichlorido-dithioates by reacting alkylmercaptans with phosphorus trichloride in the presence of sulfur
ATE316100T1 (en) 1996-06-05 2006-02-15 Roche Diagnostics Gmbh EXENDIN ANALOGAS, METHOD FOR THE PRODUCTION THEREOF AND MEDICINAL PRODUCTS CONTAINING SAME
US6110703A (en) 1996-07-05 2000-08-29 Novo Nordisk A/S Method for the production of polypeptides
CA2262647C (en) 1996-08-08 2007-12-04 Amylin Pharmaceuticals, Inc. Methods for regulating gastrointestinal motility
DE69737479T4 (en) 1996-08-30 2010-05-06 Novo Nordisk A/S GLP-1 DERIVATIVES
US6458924B2 (en) 1996-08-30 2002-10-01 Novo Nordisk A/S Derivatives of GLP-1 analogs
US6268343B1 (en) 1996-08-30 2001-07-31 Novo Nordisk A/S Derivatives of GLP-1 analogs
US6384016B1 (en) 1998-03-13 2002-05-07 Novo Nordisk A/S Stabilized aqueous peptide solutions
US7235627B2 (en) 1996-08-30 2007-06-26 Novo Nordisk A/S Derivatives of GLP-1 analogs
US6006753A (en) 1996-08-30 1999-12-28 Eli Lilly And Company Use of GLP-1 or analogs to abolish catabolic changes after surgery
US6277819B1 (en) 1996-08-30 2001-08-21 Eli Lilly And Company Use of GLP-1 or analogs in treatment of myocardial infarction
DE69732640T2 (en) 1996-09-09 2006-01-12 Zealand Pharma A/S SOLID PHASE PEPTIDE SYNTHESIS
JP2001505872A (en) 1996-09-09 2001-05-08 ジーランド ファーマシューティカルズ アクティーゼルスカブ Peptide prodrug containing α-hydroxy acid linker
UA65549C2 (en) 1996-11-05 2004-04-15 Елі Ліллі Енд Компані Use of glucagon-like peptides such as glp-1, glp-1 analog, or glp-1 derivative in methods and compositions for reducing body weight
WO1998022577A1 (en) 1996-11-15 1998-05-28 Maria Grazia Masucci Fusion proteins having increased half-lives
ES2425559T5 (en) 1997-01-07 2018-02-02 Amylin Pharmaceuticals, Llc Pharmaceutical compositions comprising exendins and their agonists
US6410511B2 (en) 1997-01-08 2002-06-25 Amylin Pharmaceuticals, Inc. Formulations for amylin agonist peptides
US6136784A (en) 1997-01-08 2000-10-24 Amylin Pharmaceuticals, Inc. Amylin agonist pharmaceutical compositions containing insulin
WO1998035033A1 (en) 1997-02-05 1998-08-13 1149336 Ontario Inc. Polynucleotides encoding proexendin, and methods and uses thereof
US5846937A (en) 1997-03-03 1998-12-08 1149336 Ontario Inc. Method of using exendin and GLP-1 to affect the central nervous system
CA2289094A1 (en) 1997-05-07 1998-11-12 Max-Planck-Gesellschaft zur Forderung der Wissenschaften E.V., Berlin New cysteine derivatives, processes for their production, and pharmaceuticals containing them
PT1019077E (en) 1997-08-08 2008-02-21 Amylin Pharmaceuticals Inc Novel exendin agonist compounds
US7157555B1 (en) 1997-08-08 2007-01-02 Amylin Pharmaceuticals, Inc. Exendin agonist compounds
US7223725B1 (en) 1997-11-14 2007-05-29 Amylin Pharmaceuticals, Inc. Exendin agonist compounds
DK1032587T4 (en) 1997-11-14 2013-04-08 Amylin Pharmaceuticals Llc New exendin agonist compounds
US7220721B1 (en) 1997-11-14 2007-05-22 Amylin Pharmaceuticals, Inc. Exendin agonist peptides
JP2003522721A (en) 1997-11-14 2003-07-29 アミリン・ファーマシューティカルズ,インコーポレイテッド New exendin agonist compounds
JP2001525371A (en) 1997-12-05 2001-12-11 イーライ・リリー・アンド・カンパニー GLP-1 preparation
US6703359B1 (en) 1998-02-13 2004-03-09 Amylin Pharmaceuticals, Inc. Inotropic and diuretic effects of exendin and GLP-1
ATE366115T1 (en) 1998-02-13 2007-07-15 Amylin Pharmaceuticals Inc INOTROPIC AND DIURETIC EFFECTS OF EXENDIN AND GLP-1
AU3247799A (en) 1998-02-27 1999-09-15 Novo Nordisk A/S Glp-1 derivatives of glp-1 and exendin with protracted profile of action
ATE466028T1 (en) 1998-02-27 2010-05-15 Novo Nordisk As N-TERMINALLY MODIFIED GLP-1 DERIVATIVES
JP4394279B2 (en) 1998-03-09 2010-01-06 ジーランド ファーマ アクティーゼルスカブ Pharmacologically active peptide conjugates with reduced propensity to enzymatic hydrolysis
WO1999049788A1 (en) 1998-03-30 1999-10-07 Focus Surgery, Inc. Ablation system
SE9802080D0 (en) 1998-06-11 1998-06-11 Hellstroem Pharmaceutical composition for the treatment of functional dyspepsia and / or irritable bowel syndrome and new use of substances therein
CA2339326A1 (en) 1998-08-10 2000-02-24 Josephine Egan Differentiation of non-insulin producing cells into insulin producing cells by glp-1 or exendin-4 and uses thereof
WO2000020592A1 (en) 1998-10-07 2000-04-13 Medical College Of Georgia Research Institute, Inc. Glucose-dependent insulinotropic peptide for use as an osteotropic hormone
US6284725B1 (en) 1998-10-08 2001-09-04 Bionebraska, Inc. Metabolic intervention with GLP-1 to improve the function of ischemic and reperfused tissue
EP2322545A1 (en) 1998-12-07 2011-05-18 Ipsen Pharma Analogues of GLP-1
US7399489B2 (en) 1999-01-14 2008-07-15 Amylin Pharmaceuticals, Inc. Exendin analog formulations
CA2356706C (en) 1999-01-14 2014-09-30 Amylin Pharmaceuticals, Inc. Novel exendin agonist formulations and methods of administration thereof
ATE460942T1 (en) 1999-01-14 2010-04-15 Amylin Pharmaceuticals Inc EXENDINE FOR GLUCAGON SUPPRESSION
US6451987B1 (en) 1999-03-15 2002-09-17 Novo Nordisk A/S Ion exchange chromatography of proteins and peptides
US6451974B1 (en) 1999-03-17 2002-09-17 Novo Nordisk A/S Method of acylating peptides and novel acylating agents
EP1956000B1 (en) 1999-03-17 2016-10-05 Novo Nordisk A/S Acylating agents useful for acylating peptides
US6271241B1 (en) 1999-04-02 2001-08-07 Neurogen Corporation Cycloalkyl and aryl fused aminoalkyl-imidazole derivatives: modulators and GLP-1 receptors
US6924264B1 (en) 1999-04-30 2005-08-02 Amylin Pharmaceuticals, Inc. Modified exendins and exendin agonists
CA2372214A1 (en) 1999-04-30 2000-11-09 Amylin Pharmaceuticals, Inc. Modified exendins and exendin agonists
SI1180121T1 (en) 1999-05-17 2004-04-30 Conjuchem, Inc. Long lasting insulinotropic peptides
US7601691B2 (en) 1999-05-17 2009-10-13 Conjuchem Biotechnologies Inc. Anti-obesity agents
US6506724B1 (en) 1999-06-01 2003-01-14 Amylin Pharmaceuticals, Inc. Use of exendins and agonists thereof for the treatment of gestational diabetes mellitus
US6344180B1 (en) 1999-06-15 2002-02-05 Bionebraska, Inc. GLP-1 as a diagnostic test to determine β-cell function and the presence of the condition of IGT and type II diabetes
EP1076066A1 (en) 1999-07-12 2001-02-14 Zealand Pharmaceuticals A/S Peptides for lowering blood glucose levels
US6528486B1 (en) 1999-07-12 2003-03-04 Zealand Pharma A/S Peptide agonists of GLP-1 activity
US6586438B2 (en) 1999-11-03 2003-07-01 Bristol-Myers Squibb Co. Antidiabetic formulation and method
US6894024B2 (en) 2000-10-20 2005-05-17 Amylin Pharmaceuticals, Inc. Treatment of hibernating myocardium and diabetic cardiomyopathy with a GLP-1 peptide
GB0121709D0 (en) 2001-09-07 2001-10-31 Imp College Innovations Ltd Food inhibition agent
WO2003053460A1 (en) 2001-12-19 2003-07-03 Eli Lilly And Company Crystalline compositions for controlling blood glucose
EP1545460A4 (en) 2001-12-20 2005-11-16 Lilly Co Eli Insulin molecule having protracted time action
US20030232761A1 (en) 2002-03-28 2003-12-18 Hinke Simon A. Novel analogues of glucose-dependent insulinotropic polypeptide
EP1837031B1 (en) 2002-06-07 2009-10-14 Waratah Pharmaceuticals, Inc. Compositions and methods for treating diabetes
AU2003243929B2 (en) 2002-07-04 2009-06-04 Zp Holding Spv K/S GLP-1 and methods for treating diabetes
KR20050083713A (en) 2002-10-02 2005-08-26 질랜드 파마 에이/에스 Stabilized exendin-4 compounds
US7192922B2 (en) 2002-11-19 2007-03-20 Allegheny-Singer Research Institute Method of treating left ventricular dysfunction
GB0300571D0 (en) 2003-01-10 2003-02-12 Imp College Innovations Ltd Modification of feeding behaviour
CA2518776A1 (en) 2003-04-29 2004-11-11 Eli Lilly And Company Insulin analogs having protracted time action
AU2004235872A1 (en) 2003-05-09 2004-11-18 Novo Nordisk A/S Peptides for use in treating obesity
US7623530B2 (en) 2003-11-20 2009-11-24 Nokia Corporation Indication of service flow termination by network control to policy decision function
RU2006131046A (en) 2004-01-30 2008-03-10 Уэрейта Фармасьютикалз, Инк. (Ca) JOINT USE OF GLP-1 AGONIST AND GASTRIN COMPOUNDS
US8076288B2 (en) 2004-02-11 2011-12-13 Amylin Pharmaceuticals, Inc. Hybrid polypeptides having glucose lowering activity
CA2849552A1 (en) 2004-02-11 2005-08-25 Amylin Pharmaceuticals, Llc Hybrid polypeptides with selectable properties
WO2006051110A2 (en) 2004-11-12 2006-05-18 Novo Nordisk A/S Stable formulations of insulinoptropic peptides
TWI362392B (en) 2005-03-18 2012-04-21 Novo Nordisk As Acylated glp-1 compounds
JP2008539735A (en) 2005-05-06 2008-11-20 バイエル・フアーマシユーチカルズ・コーポレーシヨン Glucagon-like peptide 1 (GLP-1) receptor antagonists and methods for their pharmacological use
JP2008543816A (en) 2005-06-13 2008-12-04 インペリアル イノベーションズ リミテッド Novel compounds and their effects on eating behavior
US20090202497A1 (en) 2005-08-23 2009-08-13 The General Hospital Corporation Use of glp-1, glp-1 derivatives or glp-1 fragments for skin regeneration, stimulation of hair growth, or treatment of diabetes
CN101534846B (en) 2005-11-07 2014-11-05 印第安纳大学研究及科技有限公司 Glucagon analogs exhibiting physiological solubility and stability
WO2007081824A2 (en) 2006-01-06 2007-07-19 Case Western Reserve University Fibrillation resistant proteins
WO2007095737A1 (en) 2006-02-21 2007-08-30 Waratah Pharmaceuticals Inc. Combination therapy for the treatment of diabetes comprising an exendin agonist and a gastrin compound
WO2007100535A2 (en) 2006-02-22 2007-09-07 Merck & Co., Inc. Oxyntomodulin derivatives
KR101528939B1 (en) 2006-07-18 2015-06-15 사노피 Antagonist antibody against epha2 for the treatment of cancer
ITMI20061607A1 (en) 2006-08-09 2008-02-10 Maria Vincenza Carriero PEPTIDES WITH PHARMACOLOGICAL ACTIVITY
ES2554773T3 (en) 2006-10-04 2015-12-23 Case Western Reserve University Insulin and fibrillation resistant insulin analogues
JP5819586B2 (en) 2006-11-08 2015-11-24 ジーランド ファーマ アクティーゼルスカブ Selective glucagon-like peptide-2 (GLP-2) analogs
WO2008071010A1 (en) 2006-12-12 2008-06-19 Waratah Pharmaceuticals Inc. Combination treatments with selected growth/hormone regulatory factors for diabetes and related diseases
TWI428346B (en) 2006-12-13 2014-03-01 Imp Innovations Ltd Novel compounds and their effects on feeding behaviour
US8669228B2 (en) 2007-01-05 2014-03-11 Indiana University Research And Technology Corporation Glucagon analogs exhibiting enhanced solubility in physiological pH buffers
AU2008216265B2 (en) 2007-02-15 2014-04-03 Indiana University Research And Technology Corporation Glucagon/GLP-1 receptor co-agonists
EP2025684A1 (en) 2007-08-15 2009-02-18 Zealand Pharma A/S Glucagon analogues
JP5385266B2 (en) 2007-06-15 2014-01-08 ジーランド ファーマ アクティーゼルスカブ Glucagon analog
FR2917552B1 (en) 2007-06-15 2009-08-28 Sagem Defense Securite METHOD FOR REGULATING THE TRANSMISSION GEIGE WITHIN A RECEPTION TERMINAL
EP2679597A1 (en) 2007-09-05 2014-01-01 Novo Nordisk A/S Glucagon-like peptide-1 derivatives and their pharmaceutical use
AU2008326324B9 (en) 2007-11-20 2012-11-15 Ambrx, Inc. Modified insulin polypeptides and their uses
GB2455553B (en) 2007-12-14 2012-10-24 Nuaire Ltd Motor mounting assembly for an axial fan
BRPI0907119A2 (en) 2008-01-09 2015-07-14 Sanofi Aventis Deutschland Insulin derivatives having an extremely delayed time action profile
DE102008003566A1 (en) 2008-01-09 2009-07-16 Sanofi-Aventis Deutschland Gmbh New insulin analogs useful for treating diabetes
DE102008003568A1 (en) 2008-01-09 2009-07-16 Sanofi-Aventis Deutschland Gmbh New insulin analogs useful for treating diabetes
MY152979A (en) 2008-01-09 2014-12-15 Sanofi Aventis Deutschland Novel insulin derivatives having an extremely delayed time-action profile
WO2009129250A2 (en) 2008-04-14 2009-10-22 Case Western Reserve University Meal-time insulin analogues of enhanced stability
RU2010147076A (en) 2008-04-22 2012-05-27 Кейз Вестерн Ризев Юнивесити (Us) ANSULIN ANALOGUES SPECIFIC TO ISOFORM
TWI451876B (en) 2008-06-13 2014-09-11 Lilly Co Eli Pegylated insulin lispro compounds
PE20100255A1 (en) 2008-06-17 2010-04-25 Univ Indiana Res & Tech Corp GLUCAGON / GLP-1 RECEPTOR CO-AGONISTS
TWI474835B (en) * 2008-06-17 2015-03-01 Univ Indiana Res & Tech Corp Gip-based mixed agonists for treatment of metabolic disorders and obesity
CN102088989B (en) 2008-06-17 2014-11-26 印第安纳大学研究及科技有限公司 Glucagon analogs exhibiting enhanced solubility and stability physiological pH buffers
PL219335B1 (en) 2008-07-04 2015-04-30 Inst Biotechnologii I Antybiotyków New slow-release insulin analogues
MX2011001181A (en) 2008-07-31 2011-04-05 Univ Case Western Reserve Halogen-stabilized insulin.
CN102171244B (en) 2008-08-07 2015-05-13 益普生制药股份有限公司 Analogues of glucose-dependent insulinotropic polypeptide
CN102149411A (en) 2008-09-12 2011-08-10 诺沃—诺迪斯克有限公司 Method of acylating a peptide or protein
MX2011006320A (en) 2008-12-15 2011-09-22 Zealand Pharma As Glucagon analogues.
UA104605C2 (en) 2008-12-15 2014-02-25 Зіленд Фарма А/С Glucagon analogues
DK2370462T3 (en) 2008-12-15 2014-09-08 Zealand Pharma As Glucagon-ANALOGS
ES2477880T3 (en) 2008-12-15 2014-07-18 Zealand Pharma A/S Glucagon analogues
JP5789515B2 (en) 2008-12-19 2015-10-07 インディアナ ユニバーシティー リサーチ アンド テクノロジー コーポレーションIndiana University Research And Technology Corporation Insulin analogue
WO2010080609A1 (en) 2008-12-19 2010-07-15 Indiana University Research And Technology Corporation Amide-based insulin prodrugs
WO2010096052A1 (en) 2009-02-19 2010-08-26 Merck Sharp & Dohme Corp. Oxyntomodulin analogs
WO2010107487A2 (en) 2009-03-18 2010-09-23 Wu Nian Lipid-drug conjugates for drug delivery
CN101519446A (en) 2009-03-31 2009-09-02 上海一就生物医药有限公司 Method for preparing recombinant human insulin and analogs of recombinant human insulin
AU2010260058B2 (en) 2009-06-16 2015-09-24 Indiana University Research And Technology Corporation GIP receptor-active glucagon compounds
DK2454282T3 (en) 2009-07-13 2015-05-04 Zealand Pharma As acetylated glucagonanaloger
DK2513140T3 (en) 2009-12-16 2016-01-18 Novo Nordisk As Double-acylated GLP-1 derivatives
US20110312881A1 (en) 2009-12-21 2011-12-22 Amunix, Inc. Bifunctional polypeptide compositions and methods for treatment of metabolic and cardiovascular diseases
MX342409B (en) * 2010-01-20 2016-09-28 Zealand Pharma As Treatment of cardiac conditions.
CA2788304A1 (en) 2010-01-27 2011-08-04 Indiana University Research And Technology Corporation Glucagon antagonist - gip agonist conjugates and compositions for the treatment of metabolic disorders and obesity
AR080592A1 (en) 2010-03-26 2012-04-18 Lilly Co Eli PEPTIDE WITH ACTIVITY FOR GIP-R AND GLP-1-R, FAMILY FORMULATION THAT UNDERSTANDS IT, ITS USE TO PREPARE A USEFUL MEDICINAL PRODUCT FOR THE TREATMENT OF MELLITUS DIABETES AND TO INDICATE WEIGHT LOSS
EP2552951A1 (en) 2010-03-26 2013-02-06 Novo Nordisk A/S Novel glucagon analogues
EP2552952A1 (en) 2010-03-26 2013-02-06 Novo Nordisk A/S Novel glucagon analogues
SG184988A1 (en) 2010-04-27 2012-11-29 Zealand Pharma As Peptide conjugates of glp-1 receptor agonists and gastrin and their use
AU2011247824B2 (en) 2010-04-27 2014-02-13 Betta Pharmaceuticals Co., Ltd Glucagon-like peptide-1 analogue and use thereof
UY33462A (en) 2010-06-23 2012-01-31 Zealand Pharma As GLUCAGON ANALOGS
WO2011160633A1 (en) 2010-06-24 2011-12-29 Zealand Pharma A/S Glucagon analogues
EP2637699B1 (en) 2010-11-09 2018-05-16 Novo Nordisk A/S Double-acylated glp-1 derivatives with a linker
EP2665487A1 (en) 2011-01-20 2013-11-27 Zealand Pharma A/S Combination of acylated glucagon analogues with insulin analogues
AU2012234276A1 (en) 2011-03-28 2013-08-29 Novo Nordisk A/S Novel glucagon analogues
WO2012138941A1 (en) 2011-04-05 2012-10-11 Longevity Biotech, Inc. Compositions comprising glucagon analogs and methods of making and using the same
MX355361B (en) 2011-04-12 2018-04-17 Novo Nordisk As Double-acylated glp-1 derivatives.
WO2012150503A2 (en) * 2011-05-03 2012-11-08 Zealand Pharma A/S Glu-glp-1 dual agonist signaling-selective compounds
WO2012153196A2 (en) 2011-05-10 2012-11-15 Zealand Pharma A/S Glu-glp-1 dual agonist signaling-selective compounds
JP5914641B2 (en) 2011-06-10 2016-05-11 ベイジン・ハンミ・ファーマシューティカル・カンパニー・リミテッドBeijing Hanmi Pharmaceutical Co., Ltd. Glucose-dependent insulinotropic polypeptide analogs, pharmaceutical compositions and uses thereof
JP6352806B2 (en) 2011-09-23 2018-07-04 ノヴォ ノルディスク アー/エス New glucagon analogues
BR112014010780A2 (en) 2011-11-03 2017-04-25 Zealand Pharma As glp-1-gastrin receptor agonist peptide conjugates
PE20142113A1 (en) 2011-12-23 2014-12-03 Zealand Pharma As GLUCAGON ANALOGS
KR20150006052A (en) 2012-05-03 2015-01-15 질랜드 파마 에이/에스 Glucagon-like-peptide-2 (glp-2) analogues
MX356641B (en) 2012-05-03 2018-06-07 Zealand Pharma As Gip-glp-1 dual agonist compounds and methods.
AR091476A1 (en) 2012-06-21 2015-02-04 Univ Indiana Res & Tech Corp REGION FUSION POLYPEPTIDES Fc POLYPEPTIDE BINDING RECEIVER OF INCRETINA AND CONJUGADOS WITH EFECTIVE FUNCTION Fc ALTERED
NZ704043A (en) 2012-07-23 2017-07-28 Zealand Pharma As Glucagon analogues
TWI608013B (en) 2012-09-17 2017-12-11 西蘭製藥公司 Glucagon analogues
AU2013366691A1 (en) 2012-12-21 2015-07-09 Sanofi Exendin-4 derivatives
WO2014140222A1 (en) * 2013-03-14 2014-09-18 Medimmune Limited Pegylated glucagon and glp-1 co-agonists for the treatment of obesity
US9988429B2 (en) 2013-10-17 2018-06-05 Zealand Pharma A/S Glucagon analogues
PL3057984T3 (en) 2013-10-17 2018-12-31 Zealand Pharma A/S Acylated glucagon analogues
TW201609796A (en) 2013-12-13 2016-03-16 賽諾菲公司 Non-acylated EXENDIN-4 peptide analogues
CN106029088A (en) 2014-02-18 2016-10-12 诺和诺德股份有限公司 Stable glucagon analogues and use for treatment of hypoglycaemia
CA2965732A1 (en) 2014-10-29 2016-05-06 Zealand Pharma A/S Gip agonist compounds and methods
JP6989385B2 (en) 2015-04-16 2022-01-05 ジーランド ファーマ アクティーゼルスカブ Acylated glucagon analog

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10093713B2 (en) * 2013-11-06 2018-10-09 Zealand Pharma A/S GIP-GLP-1 dual agonist compounds and methods
US11008375B2 (en) * 2013-11-06 2021-05-18 Zealand Pharma A/S GIP-GLP-1 dual agonist compounds and methods
US11111285B2 (en) * 2013-11-06 2021-09-07 Zealand Pharma A/S Glucagon-GLP-1-GIP triple agonist compounds

Also Published As

Publication number Publication date
MX2016005560A (en) 2016-11-15
EA201690629A1 (en) 2016-10-31
AU2014345570B2 (en) 2019-01-24
US10131702B2 (en) 2018-11-20
CA2929107C (en) 2023-09-26
AU2014345570A1 (en) 2016-05-12
MX369770B (en) 2019-11-21
KR102310392B1 (en) 2021-10-13
TR201902516T4 (en) 2019-03-21
US11111285B2 (en) 2021-09-07
CA2929107A1 (en) 2015-05-14
KR20160074008A (en) 2016-06-27
CN105829339A (en) 2016-08-03
EA035688B1 (en) 2020-07-27
BR112016009995A2 (en) 2017-12-05
EP3066117B1 (en) 2019-01-02
WO2015067716A1 (en) 2015-05-14
EP3066117A1 (en) 2016-09-14
US20160257729A1 (en) 2016-09-08
CN105829339B (en) 2021-03-12
JP2020045362A (en) 2020-03-26
JP2017503474A (en) 2017-02-02
US20190270789A1 (en) 2019-09-05
BR112016009995B1 (en) 2023-04-18

Similar Documents

Publication Publication Date Title
US11111285B2 (en) Glucagon-GLP-1-GIP triple agonist compounds
US11008375B2 (en) GIP-GLP-1 dual agonist compounds and methods
US11814417B2 (en) GIP agonist compounds and methods
AU2014345569A1 (en) GIP-GLP-1 dual agonist compounds and methods

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION UNDERGOING PREEXAM PROCESSING

AS Assignment

Owner name: ZOOLANDER SA LLC, NEW YORK

Free format text: PATENT SECURITY AGREEMENT;ASSIGNOR:ZEALAND PHARMA A/S;REEL/FRAME:058593/0261

Effective date: 20211213

AS Assignment

Owner name: ZEALAND PHARMA A/S, DENMARK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:JUST, RASMUS;RIBER, DITTE;SHELTON, ANNE PERNILLE TOFTENG;AND OTHERS;SIGNING DATES FROM 20150211 TO 20160216;REEL/FRAME:059037/0522

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

AS Assignment

Owner name: ZEALAND PHARMA A/S, DENMARK

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:ZOOLANDER SA LLC;REEL/FRAME:063624/0547

Effective date: 20230509

AS Assignment

Owner name: ZEALAND PHARMA A/S, DENMARK

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:ZOOLANDER SA LLC;REEL/FRAME:063672/0342

Effective date: 20230509

STCB Information on status: application discontinuation

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