CN116970063A - GLP-1/GIP receptor co-agonists, pharmaceutical compositions comprising same and uses thereof - Google Patents

GLP-1/GIP receptor co-agonists, pharmaceutical compositions comprising same and uses thereof Download PDF

Info

Publication number
CN116970063A
CN116970063A CN202211568720.9A CN202211568720A CN116970063A CN 116970063 A CN116970063 A CN 116970063A CN 202211568720 A CN202211568720 A CN 202211568720A CN 116970063 A CN116970063 A CN 116970063A
Authority
CN
China
Prior art keywords
ser
ala
pro
ile
lys
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.)
Pending
Application number
CN202211568720.9A
Other languages
Chinese (zh)
Inventor
吕佩
田长麟
郑勇
吴文奎
王辉
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.)
Suzhou Xingzhou Biotechnology Co ltd
Original Assignee
Suzhou Xingzhou Biotechnology Co ltd
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 Suzhou Xingzhou Biotechnology Co ltd filed Critical Suzhou Xingzhou Biotechnology Co ltd
Publication of CN116970063A publication Critical patent/CN116970063A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/26Glucagons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/107General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides
    • 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
    • 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

Abstract

The present invention provides a GLP-1/GIP receptor co-agonist, and also provides pharmaceutical compositions comprising these compounds, their use, and methods of treating and/or preventing metabolic diseases or disorders.

Description

GLP-1/GIP receptor co-agonists, pharmaceutical compositions comprising same and uses thereof
Technical Field
The present disclosure relates to GLP-1/GIP receptor co-agonists, pharmaceutical compositions comprising the same, and uses and methods for treating and/or preventing metabolic diseases or disorders.
Background
Glucagon-like peptide GLP-1 (a polypeptide hormone secreted by the intestinal tract after food stimulation, GLP-1 stimulates insulin secretion in a glucose dependent manner and reduces glucagon secretion. GLP-1 receptors are widely distributed throughout many organs or tissues of the body, including the central nervous system, gastrointestinal tract, cardiovascular system, liver, adipose tissue, muscle, etc., in addition to the pancreas. GLP-1 receptor agonists exert hypoglycemic effects through a variety of mechanisms that slow down gastric emptying, central appetite suppression, and reduce food intake. However, natural GLP-1 is easily degraded by dipeptidyl peptidase in vivo to lose activity, and the half-life in vivo is only 1-2min, so that the clinical application of the natural GLP-1 is greatly limited.
Glucose-dependent insulinotropic hormone GIP (gluocose-dependent insulinotropic) is currently thought to be secreted mainly by enteroendocrine K cells in the duodenum and upper jejunum. Similar to GLP-1, GIP can stimulate insulin secretion. GIP receptor GIPR is widely distributed in organisms and expressed in pancreas, stomach, small intestine, adipose tissue, heart and brain tissue. In addition, activation of the GIP-GIPR pathway also exerts a weight-loss effect. However, GIP has a short in vivo bioactive half-life of less than 2min in mice and 7min and 5min in normal and type II diabetics, respectively.
Glucagon (GCG) is a hormone produced in the α cells of the pancreas, and acts on the liver in a stress state such as cold and hunger of the body to decompose glycogen in the liver, thereby increasing blood sugar. In addition to its glycemic effect, GCG has effects of promoting lipolysis, fat oxidation, fever, etc. in vivo (see diabetes, 2017, 60, 1851-1861), and long-term administration can exhibit a weight-loss effect by increasing energy metabolism, but GCG has not been widely used for its beneficial effects on energy metabolism due to its inherent glycemic effect.
Drug developers developed a range of GLP-1 receptor agonists and GIP receptor agonists. GLP-1 receptor agonists and GIP receptor agonists exert the same biological effects as native GLP-1 and GIP, and are also prevented from being degraded and deactivated, thereby prolonging the duration of action.
However, there remains a need for alternative GLP-1 receptor agonists, in particular co-agonists having co-agonism for the GLP-1 receptor and the GIP receptor, in particular co-agonists having co-agonism for the GLP-1 receptor, the GIP receptor and the GCG receptor. In particular, it is desirable that the agonist has a good blood sugar-reducing effect, especially, a combination of a blood sugar-reducing effect and a weight-reducing effect. It is also desirable that the agonist have high plasma stability and thus pharmacokinetic profiles that support once a week subcutaneous administration in humans.
Disclosure of Invention
In order to solve the above technical problems, the inventors have conducted intensive studies and have proposed a technical solution of the present disclosure. In one aspect, the present disclosure provides a compound of formula I:
L 1 -NH-(CH 2 ) n -C(O)-L 2 -NH 2 i is a kind of
Wherein, the liquid crystal display device comprises a liquid crystal display device,
L 1 peptide analogs of the GIP (1-28) peptide, said L 1 Is a peptide consisting of 28 amino acids, and the L 1 Amino acid sequence of (a) and SEQ ID NO:1 has an identity of at least 39%,
L 2 is a polypeptide having a sequence represented by SEQ ID NO:2, a peptide of an amino acid sequence consisting of,
n is any integer from 2 to 6, and
the compounds have GLP-1 receptor agonist activity, or GIP receptor agonist activity, or both.
By peptide chain engineering of GIP (1-28) peptides, in particular by use of-NH- (CH) 2 ) n -C (O) -units (n is an integer from 2 to 6) replace the common-NH- (CH) in peptide chains 2 ) -C (O) -units, the resulting compounds of formula I unexpectedly retain high activity at the GLP-1 receptor, even with co-agonism at the GLP-1 receptor and the GIP receptor, providing a hypoglycemic and weight-reducing effect.
In another aspect, the present disclosure provides a compound of formula II:
Tyr 1 -X 2 -Glu 3 -Gly 4 -X 5 -X 6 -Thr 7 -Ser 8 -Asp 9 -X 10 -Ser 11 -X 12 -X 13 -Leu 14 -Asp 15 -X 16 -Ile 17 -X 18 -Gln 19 -Lys 20 -X 21 -Phe 22 -X 23 -X 24 -X 25 -Leu 26 -Ile 27 -Ala 28 -NH-(CH 2 ) 4 -C(O)-Pro 31 -Ser 32 -Ser 33 -Gly 34 -Ala 35 -Pro 36 -Pro 37 -Pro 38 -Ser 39 -NH 2 the compound of the formula II is shown in the specification,
wherein X is 2 、X 5 、X 6 、X 10 、X 12 、X 13 、X 16 、X 18 、X 21 、X 23 、X 24 And X 25 Each independently selected from natural amino acids or non-natural amino acid residues.
By peptide chain engineering of GIP (1-28) peptides, in particular by use of-NH- (CH) 2 ) 4 -C (O) -unit substitution of the-NH-CH common in peptide chains 2 -C (O) -units, the resulting compounds of formula II unexpectedly retain co-agonism at the GLP-1 receptor and the GIP receptor, providing a hypoglycemic and weight-reducing effect. At the same time, the compounds of formula II have a long half-life supporting subcutaneous injections once a week for administration to humans as a drug generation The kinetics are superior to those of known drugs administered subcutaneously once daily, thereby improving patient compliance.
Furthermore, the compounds according to the present disclosure have less and controllable adverse gastrointestinal irritation, and thus can improve therapeutic effects by increasing the administered dose.
In another aspect, the present disclosure provides a pharmaceutical composition comprising:
a compound according to the present disclosure or a pharmaceutically acceptable salt thereof, and
a pharmaceutically acceptable carrier, diluent or excipient.
In another aspect, the present disclosure provides the use of a pharmaceutical composition according to the present disclosure or a compound according to the present disclosure or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment and/or prevention of a metabolic disease or disorder.
In yet another aspect, the present disclosure provides a method of treating and/or preventing a metabolic disease or disorder comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition according to the present disclosure or a compound according to the present disclosure or a pharmaceutically acceptable salt thereof.
Drawings
FIG. 1 is a mass spectrum of compound NBB-T007 according to the present disclosure.
FIG. 2 is a high performance liquid chromatogram of compound NBB-T007 according to the present disclosure.
Fig. 3 is a graph of potency versus concentration of the control cord Ma Lutai acting on a target.
FIG. 4 is a graph of potency-concentration change of compound NBB-T007-1 against target according to the present disclosure.
FIG. 5 is a graph of potency-concentration change of a compound NBB-T007-2 against a target according to the present disclosure.
FIG. 6 is a graph of potency-concentration change of compound NBB-T007-3 against target according to the present disclosure.
FIG. 7 is a graph of potency-concentration change of compound NBB-T007-4 against target according to the present disclosure.
FIG. 8 is a graph of potency-concentration change of compound NBB-T007-5 against target according to the present disclosure.
FIG. 9 is a graph of potency-concentration change of compound NBB-T007-6 against target according to the present disclosure.
FIG. 10 is a graph of potency-concentration change of compound NBB-T007-7 against target according to the present disclosure.
FIG. 11 is a graph of potency-concentration change of compound NBB-T007-8 against target according to the present disclosure.
FIG. 12 is a graph of potency-concentration change of compound NBB-T007-9 against target according to the present disclosure.
FIG. 13 is a graph of potency-concentration change of compound NBB-T007-10 against target according to the present disclosure.
FIG. 14 is a graph of potency-concentration change of compound NBB-T007-11 against target according to the present disclosure.
FIG. 15 is a graph of potency-concentration change of compound NBB-T007-12 against target according to the present disclosure.
FIG. 16 is a graph of potency-concentration change of compound NBB-T007-14 against target according to the present disclosure.
FIG. 17 is a graph of potency-concentration change of compound NBB-T007-15 against target according to the present disclosure.
Fig. 18 is a graph of potency-concentration change of compound NBB-T007 on a target according to the present disclosure.
FIG. 19 is a graph of potency-concentration change of compound NBB-T007-17 against target according to the present disclosure.
FIG. 20 is a graph of potency-concentration change of compound NBB-T007-18 against target according to the present disclosure.
FIG. 21 is a graph of potency-concentration change of compound NBB-T007-19 against target according to the present disclosure.
FIG. 22 is a graph of potency-concentration change of compound NBB-T007-21 against target according to the present disclosure.
FIG. 23 is a graph of potency-concentration change of compound NBB-T007-22 against target according to the present disclosure.
FIG. 24 is a graph of potency-concentration change of compound NBB-T007-23 against target according to the present disclosure.
FIG. 25 is a graph of potency-concentration change of compound NBB-T007-24 against target according to the present disclosure.
Fig. 26 is a graph of potency-concentration change of compound NBB-EX4 against a target according to the present disclosure.
FIG. 27 is a graph of blood glucose versus time for test db/db mice.
FIG. 28 is a graph of body weight versus time for db/db mice tested.
FIG. 29 is a graph of percent blood glucose reduction versus time for the db/db mice tested.
FIG. 30 is a graph of blood glucose versus time for normal mice tested.
FIG. 31 is a graph showing the weight-time change in normal mice tested.
Figure 32 is a concentration-time profile in plasma of compound NBB-T007 according to the present disclosure injected subcutaneously into SD rats tested.
Fig. 33 is a concentration-time profile of control cord Ma Lutai in plasma injected subcutaneously into SD rats.
FIG. 34 is a concentration-time profile of NBB-T007-10 in plasma according to the present disclosure injected subcutaneously in SD rats tested.
FIG. 35 is a concentration-time profile of NBB-T007-12 in plasma according to the present disclosure injected subcutaneously in SD rats tested.
Fig. 36 is a weight-time profile of the DIO mice tested.
Fig. 37 is a graph of blood glucose versus time for the DIO mice tested.
Fig. 38 is a graph of blood glucose versus time for the DIO mice tested.
FIG. 39 (a) is a bar graph of serum insulin content change in the DIO mice tested.
FIG. 39 (b) is a bar graph of serum biochemical marker changes (UREA, TG, CHO, HDL, LDL and CREA) in the DIO mice tested.
FIG. 39 (c) is a bar graph of serum biochemical marker changes (ALT, AST, ALB and TBIL) in the DIO mice tested.
FIG. 40 (a) is a bar graph of body fat rate change in the DIO mice tested.
Fig. 40 (b) is a bar graph of triglyceride content change in the tested DIO mice.
Detailed Description
[ definition ]
As used herein, "analog" means a compound, such as a natural or synthetic peptide or polypeptide, that activates a target receptor and triggers at least one in vivo or in vitro effect of an agonist on the receptor.
As used herein, a sequence or structural formula of a compound contains standard single-letter or three-letter codes for the natural amino acids (also referred to as protein amino acids or encoded amino acids) of 20 constituent proteins. The amino and carboxyl groups of the remaining 19 protein amino acids, except proline (Pro), are both attached to an alpha carbon atom, also known as alpha-amino acids. For example, alpha-Ala represents (alpha-) alanine, having the structure CH 3 CH(NH 2 ) COOH, wherein the amino group is attached to the alpha carbon atom. beta-Ala represents beta-alanine, and has the structural formula of NH 2 CH 2 CH 2 COOH, wherein the amino group is attached to the β carbon atom.
As used herein, each amino acid residue may be in either the L-or D-configuration independently of the other, or may have pendant substituents on carbon atoms independently of the other.
As used herein, the sequence or structural formula of a compound may also contain unnatural amino acid residues. For example, aib represents a 2-aminoisobutyric acid residue; homoPhe represents a homophenylalanine residue; cpa-Ala represents a p-chlorophenyl alanine residue; fpa5-Ala represents a pentafluorophenylalanine residue; na1 represents a 1-theaylalanine residue.
In the context of this document, unless clearly contradicted by context, a compound of formula I or formula II is synonymous with "polypeptide".
In the context of this document, unless indicated otherwise or clearly contradicted, the numbering of the amino acid positions is from the peptide chain of the compound or the leftmost N-terminus of the structural formula. For example, using the compound of formula II, the N-terminal amino acid is tyrosine (Tyr) at position 1 and the C-terminal amino acid is serine (Ser) at position 39. The chemical formula IIIn the compound, -NH- (CH) 2 ) 4 the-C (O) -unit is not a residue of the natural α -amino acid, nor is it a residue of 2 amino acids, but is only a residue of 1 amino acid. However, for convenience, the form-NH- (CH) will be described herein 2 ) 4 -C (O) -unit numbering is the amino acids at position 29 and 30 occupying 2 positions.
As used herein, "individual in need thereof means a mammal, preferably a human, but also non-human animals, including non-human primates (e.g., monkeys, cynomolgus monkeys, etc.), pets (e.g., cats, dogs, etc.), livestock (e.g., cows, sheep, pigs, horses, etc.), and rodents (e.g., rats, mice, guinea pigs, etc.) having a condition, disease, disorder or symptom that requires treatment or prevention.
As used herein, "effective amount" means the amount, concentration, or dose of one or more compounds of the present disclosure, or a pharmaceutically acceptable salt thereof, that provides a desired effect (i.e., can result in a clinically measurable difference in the condition of the individual, such as, for example, a decrease in blood glucose, and/or a decrease in weight or fat) in the individual being diagnosed or treated after single or multiple doses are administered to the individual in need thereof. The effective amount can be readily determined by one skilled in the art by using known techniques and by observing results obtained in similar circumstances. In determining an effective amount for an individual, a number of factors are considered, including, but not limited to, the species, size, age, general health of the mammal, the particular disease or disorder involved, the severity of the disease or disorder, the response of the individual, the particular compound administered, the mode of administration, the bioavailability characteristics of the formulation administered, the selected dosage regimen, use with drug therapy, and other relevant circumstances.
As used herein, the term "treating" means attenuating, inhibiting, reversing, slowing or stopping the progression or severity of an existing condition, disease, disorder or symptom.
As used herein, the term "C 12 -C 24 Aliphatic diacid "means a linear or branched dicarboxylic acid having 12 to 24 carbon atoms. In one embodiment, C is suitable for the present disclosure 12 -C 24 The aliphatic diacid may be a saturated diacid or an unsaturated diacid, preferably a saturated diacid. C suitable for the compounds of the present disclosure 12 -C 24 Fatty acids include, but are not limited to, dodecanedioic acid (C 12 Diacid), tridecanedioic acid (C) 13 Diacid), tetradecanedioic acid (C) 14 Diacid), pentadecanedioic acid (C) 15 Diacid), hexadecanediacid (C) 16 Diacid), heptadecanediacid (C) 17 Diacid), octadecanedioic acid (C) 18 Diacid), nonadecanoic acid (C) 19 Diacid), eicosanedioic acid (C) 20 Diacid), heneicosanedioic acid (C) 21 Diacid), behenic acid (C) 22 Diacid), tricosadiacid (C) 23 Diacid), tetracosanedioic acid (C) 24 Diacid), and branched and/or substituted derivatives thereof.
As used herein, the term "plasma half-life" or "half-life" refers to the time required for half of the relevant compound to clear from plasma.
As used herein, "in vitro activity" refers to an indication of the ability of a peptide to activate GLP-1 receptors, GIP receptors and/or GCG receptors in a cell-based assay. In vitro activity is expressed as "half maximal Effective Concentration (EC) 50 ) ", which is the effective concentration of a compound that results in 50% activity in a single dose response experiment. As used herein, "EC 50 By "is meant the effective concentration of the compound that results in 50% activation/stimulation of the assay endpoint, such as a dose-response curve (e.g., cAMP).
In the present context, cord Ma Lutai (Semaglutide) refers to a chemically synthesized GLP-1 analogue having the structure shown below:
in the context of the present invention, tirzeppa (TZP) is a GLP-1/GIP receptor co-agonist.
[ GLP-1/GIP receptor Co-agonists ]
The present disclosure provides a compound of formula I:
L 1 -NH-(CH 2 ) n -C(O)-L 2 -NH 2 i is a kind of
Wherein, the liquid crystal display device comprises a liquid crystal display device,
L 1 peptide analogs of the GIP (1-28) peptide, said L 1 Is a peptide consisting of 28 amino acids, and the L 1 Amino acid sequence of (a) and SEQ ID NO:1 has an identity of at least 39%,
L 2 is a polypeptide having a sequence represented by SEQ ID NO:2, a peptide of an amino acid sequence consisting of,
n is any integer from 2 to 6, and
the compounds have GLP-1 receptor agonist activity, or GIP receptor agonist activity, or both.
sEQID NO:1 is GIP (1-28) sequence: YAEGTFISDYSIAMDKIHQQDFVNWLLA it is Tyr-Ala-Glu-Gly-Thr-Phe-Ile-Ser-Asp-Tyr-Ser-Ile-Ala-Met-Asp-Lys-Ile-His-Gln-Gln-Asp-Phe-Val-Asn-Trp-Leu-Leu-Ala.
SEQ ID NO:2 is PSSGAPPPS, namely Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser.
“L 1 Amino acid sequence of (a) and SEQ ID NO:1 has an identity of at least 39% means L 1 And SEQ ID NO:1, in the 28 amino acids from position 1 to position 28, the same amino acid is present at a total of at least 11 positions, for example at a total of 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27 or 28 positions.
In some examples, L 1 Amino acid sequence of (a) and SEQ ID NO:1, have identical amino acids at a total of 11, 18, 19 or 20 positions.
In some examples, L 1 Amino acid sequence of (a) and SEQ ID NO:1 has at least 39% identity, for example 39%, 43%, 46%, 50%, 54%, 57%, 61%, 64%, 68%, 71%, 75%, 79%, 82%, 86%, 89%, 93%, 96% or 100% identity.
In some examples, L 1 Amino acid sequence of (2)SEQ ID NO:1 has 39%, 64%, 68% or 71% identity.
In some examples, n is 2, 3, 4, 5, or 6, preferably 4.
By peptide chain engineering of GIP (1-28) peptides, in particular by use of-NH- (CH) 2 ) n -C (O) -units (n is an integer from 2 to 6) replace the common-NH- (CH) in peptide chains 2 ) -C (O) -units, the resulting compounds of formula I can surprisingly retain agonism to the GIP receptor, even with co-agonism to the GLP-1 receptor and the GIP receptor, providing a hypoglycemic and weight-reducing effect.
In some examples, the polypeptide that hybridizes to SEQ ID NO:1 compared with L 1 Comprising Y1H substitutions.
In some examples, the polypeptide that hybridizes to SEQ ID NO:1 compared with L 1 Comprising a substitution selected from the group consisting of A2G, A2 (Aib) and A2 (beta-Ala).
In some examples, the polypeptide that hybridizes to SEQ ID NO:1 compared with L 1 Comprising a T5S substitution.
In some examples, the polypeptide that hybridizes to SEQ ID NO:1 compared with L 1 Comprising a substitution selected from F6 (homo-Phe) and F6 (Cpa-Ala).
In some examples, the polypeptide that hybridizes to SEQ ID NO:1 compared with L 1 Comprising an I7T substitution.
In some examples, the polypeptide that hybridizes to SEQ ID NO:1 compared with L 1 Comprising a substitution selected from Y10L and Y10 (Fpa 5-Ala).
In some examples, the polypeptide that hybridizes to SEQ ID NO:1 compared with L 1 Comprising an I12K substitution.
In some examples, the polypeptide that hybridizes to SEQ ID NO:1 compared with L 1 Comprising a substitution selected from a13Q, A (Aib) and a 13Y.
In some examples, the polypeptide that hybridizes to SEQ ID NO:1 compared with L 1 Comprising a substitution selected from M14L and M14 (. Alpha. -meL).
In some examples, the polypeptide that hybridizes to SEQ ID NO:1 compared with L 1 Comprising a D15E substitution.
In some examples, the polypeptide that hybridizes to SEQ ID NO:1 compared with L 1 Comprising a substitution selected from the group consisting of K16E and K16 (Ala).
In some examples, the polypeptide that hybridizes to SEQ ID NO:1 compared with L 1 Comprising an I17E substitution.
In some examples, the polypeptide that hybridizes to SEQ ID NO:1 compared with L 1 Comprising a substitution selected from H18A and H18 (Aib).
In some examples, the polypeptide that hybridizes to SEQ ID NO:1 compared with L 1 Comprising a Q19V substitution.
In some examples, the polypeptide that hybridizes to SEQ ID NO:1 compared with L 1 Comprising a substitution selected from Q20R and Q20K.
In some examples, the polypeptide that hybridizes to SEQ ID NO:1 compared with L 1 Comprising a substitution selected from the group consisting of D21L, D21A, D E and D21 (Abu).
In some examples, the polypeptide that hybridizes to SEQ ID NO:1 compared with L 1 Comprising a substitution selected from V23I and V23L.
In some examples, the polypeptide that hybridizes to SEQ ID NO:1 compared with L 1 Comprising a substitution selected from the group consisting of N24E and N24Q.
In some examples, the polypeptide that hybridizes to SEQ ID NO:1 compared with L 1 Comprising a substitution selected from the group consisting of W25 (Na 1) and W25 (2-me-Trp).
In some examples, the polypeptide that hybridizes to SEQ ID NO:1 compared with L 1 Comprising a substitution selected from the group consisting of L27K and L27I.
In some examples, the polypeptide that hybridizes to SEQ ID NO:1 compared with L 1 Comprising a28N substitution.
The present disclosure also provides a compound of formula XXI below or a pharmaceutically acceptable salt thereof:
His-Gly-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Leu-Ser-Lys-Gln-Met-Glu-Glu-Glu-Ala-Val-Arg-Leu-Phe-Ile-Glu-Trp-Leu-Lys-Asn-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2 formula XXI.
By peptide chain engineering of GIP (1-28) peptides, in particular by use of-NH- (CH) 2 ) 4 -C (O) -unit substitution of the-NH-CH common in peptide chains 2 -C (O) -units, the resulting compounds of formula XXI unexpectedly retain agonism at the GLP-1 receptor, providing hypoglycemic and weight-reducing effects.
In some examples, in formula I, the amino group at position 16 or 20When the acid is lysine (Lys), C is bonded directly or via a linker 12 -C 24 Chemical modification of the aliphatic diacid conjugated to the epsilon-amino group of the lysine (Lys) side chain, the linker being selected from (AEEA) 2 -(γ-Glu)a、AEEA-Ahx-(γ-Glu) a 、(Ahx) 2 -(γ-Glu) a And (beta-Ala) 2 -(γ-Glu) a Wherein a is 1 or 2;
preferably via (AEEA) 2 - (gamma-Glu) conjugation of eicosanedioic acid to the epsilon-amino group of lysine (Lys) side chain at position 16 or 20 for chemical modification; or alternatively
Via (Ahx) 2 - (gamma-Glu), AEEA-Ahx- (gamma-Glu) or (. Beta. -Ala) 2 - (gamma-Glu) conjugated eicosanedioic acid to the epsilon-amino group of lysine (Lys) side chain at position 20.
In some examples, in formula I, the histidine (His) or tyrosine (Tyr) at position 1 is amidated, e.g., by-C m H 2m+1 -C (O) -is attached to the amino group of histidine (His) or tyrosine (Tyr), and m is an integer from 1 to 20;
preferably, the amino group of tyrosine 1 (Tyr) is bound to CH by 3 C (O) or with C 19 H 39 -C (O) -linked amidation.
In some examples, in formula I, the α -carbon atoms of any two amino acids may be linked into a ring via a direct bond or via a linker selected from alkyl or alkenyl groups containing 2 to 20 carbon atoms;
preferably, when the amino acids at position 13 and 16 are both alanine, the alpha-carbon atom of alanine (Ala) at position 13 is attached to the alpha-carbon atom of alanine (Ala) at position 16 via an alkenyl group containing 10 carbon atoms.
The present disclosure also provides a compound of formula II:
Tyr 1 -X 2 -Glu 3 -Gly 4 -X 5 -X 6 -Thr 7 -Ser 8 -Asp 9 -X 10 -Ser 11 -X 12 -X 13 -Leu 14 -Asp 15 -X 16 -Ile 17 -X 18 -Gln 19 -Lys 20 -X 21 -Phe 22 -X 23 -X 24 -X 25 -Leu 26 -Ile 27 -Ala 28 -NH-(CH 2 ) 4 -C(O)-Pro 31 -Ser 32 -Ser 33 -Gly 34 -Ala 35 -Pro 36 -Pro 37 -Pro 38 -Ser 39 -NH 2
the compound of the formula II is shown in the specification,
wherein X is 2 、X 5 、X 6 、X 10 、X 12 、X 13 、X 16 、X 18 、X 21 、X 23 、X 24 And X 25 Each independently selected from natural amino acids or non-natural amino acid residues.
By peptide chain engineering of GIP (1-28) peptides, in particular by use of-NH- (CH) 2 ) 4 -C (O) -unit substitution of the-NH-CH common in peptide chains 2 -C (O) -units, the resulting compounds of formula II unexpectedly retain co-agonism at the GLP-1 receptor and the GIP receptor, providing a hypoglycemic and weight-reducing effect. At the same time, the compound of formula II has a long half-life, supporting pharmacokinetic profile of once a week administration to humans via subcutaneous injections, superior to known drugs administered once a day via subcutaneous injections, thereby improving patient compliance.
In some examples, X 2 Is an amino acid residue selected from the group consisting of 2-aminoisobutyric acid (Aib) and (. Beta. -Ala),
X 5 is an amino acid residue selected from Thr and Ser,
X 6 is an amino acid residue selected from the group consisting of Phe, homophenylalanine (homoPhe) and para-chlorophenylalanine (Cpa-Ala),
X 10 is an amino acid residue selected from Tyr and pentafluorophenylalanine (Fpa 5-Ala),
X 12 is an amino acid residue selected from Ile and Lys,
X 13 amino acid residues selected from the group consisting of 2-aminoisobutyric acid (Aib), tyr, and Ala,
X 16 is selected from Lys and AlaAn amino acid residue is present in the amino acid sequence,
X 18 is an amino acid residue selected from Ala and Aib,
X 21 is an amino acid residue selected from Ala, glu and 2-aminobutyric acid (Abu),
X 23 is an amino acid residue selected from Val and Leu,
X 24 is an amino acid residue selected from Gln and Asn, and
X 25 is an amino acid residue selected from Trp, 2-methyltryptophan (2-me-Tp) and 1-theaylalanine (Nal). In some examples, the compound has a structure selected from any one of the following formulas III to XX:
Tyr-(Aib)-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Ile-Ala-Leu-Asp-Ala-Ile-Ala-Gln-Lys-Ala-Phe-Val-Gln-Trp-Leu-Ile-Ala-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
the compound of the formula III,
Tyr-(Aib)-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Ile-(Aib)-Leu-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Val-Gln-Trp-Leu-Ile-Ala-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
the compound of the formula IV,
Tyr-(Aib)-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Ile-Tyr-(α-meL)-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Val-Gln-Trp-Leu-Ile-Ala-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
the characteristic of the V-shaped alloy is that,
Tyr-(β-Ala)-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Ile-(Aib)-Leu-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Val-Gln-Trp-Leu-Ile-Ala-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
a compound of the formula VI,
Tyr-(Aib)-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Ile-(Aib)-Leu-Asp-Lys-Ile-Ala-Gln-Lys-Glu-Phe-Val-Gln-Trp-Leu-Ile-Ala-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
the process for preparing VI includes such steps as,
Tyr-(Aib)-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Lys-(Aib)-Leu-Asp-Lys-Ile-Ala-Gln-Lys-Glu-Phe-Val-Asn-Trp-Leu-Ile-Ala-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
a worker of the type VI,
Tyr-(Aib)-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Ile-(Aib)-Leu-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Val-Asn-Trp-Leu-Ile-Ala-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
the composition of the material IX,
Tyr-(Aib)-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Lys-(Aib)-Leu-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Val-Asn-Trp-Leu-Ile-Ala-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
the X-ray tube is arranged on the inner side of the tube,
tyr- (Aib) -Glu-Gly- (D-Thr) - (D-Phe) -Thr-Ser-Asp-Tyr-Ser-Gole- (Aib) -Leu-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Val-Gln-Trp-Leu-Ile-Ala- [ NH- (CH) 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
The compounds of the formula XI are shown in the specification,
Tyr-(Aib)-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Ile-(Aib)-Leu-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Val-Gln-Trp-Leu-Ile-Ala-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-(D-Ala)-Pro-Pro-Pro-Ser-NH 2
The compound of the formula XII,
Tyr-(Aib)-(D-Glu)-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Ile-(Aib)-Leu-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Val-Gln-Trp-Leu-Ile-Ala-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
the compound of the formula XIII,
Tyr-(Aib)-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Ile-Tyr-(α-meL)-Asp-Lys-Ile-(Aib)-Gln-Lys-Ala-Phe-Val-Gln-Trp-Leu-Ile-Ala-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
the compound of formula XIV,
Tyr-(Aib)-Glu-Gly-Thr-(homo-Phe)-Thr-Ser-Asp-Tyr-Ser-Ile-(Aib)-Leu-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Val-Gln-Trp-Leu-Ile-Ala-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
the composition of the composition is XV,
Tyr-(Aib)-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Ile-(Aib)-Leu-Asp-Lys-Ile-Ala-Gln-Lys-(Abu)-Phe-Val-Gln-Trp-Leu-Ile-Ala-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
the first component of the first component is a component of the second component,
Tyr-(Aib)-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Ile-(Aib)-Leu-Asp-Lys-Ile-A1a-Gln-Lys-Ala-Phe-Val-Gln-(2-me-Trp)-Leu-Ile-Ala-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
the compound of formula XVII is represented by formula,
Tyr-(Aib)-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Ile-(Aib)-Leu-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Val-Gln-(Nal)-Leu-Ile-Ala-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
the compound of formula XVIII,
Tyr-(Aib)-Glu-Gly-Ser-Phe-Thr-Ser-Asp-Tyr-Ser-Ile-(Aib)-Leu-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Leu-Gln-Trp-Leu-Ile-Ala-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
formula XIX, and Tyr- (Aib) -Glu-Gly-Thr- (Cpa-Ala) -Thr-Ser-Asp- (Fpa 5-Ala) -Ser-Ile- (Aib) -Leu-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Val-Gln-Trp-Leu-Ile-Ala- [ NH- (CH) 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
Formula XX.
In some examples, in any of formulas I-XXI, each amino acid residue is in the L-configuration or the D-configuration independently of the other.
In some examples, in any of formulas I-XXI, each amino acid residue is, independently of the other, in the L-configuration.
In some examples, in any of formulas I-XXI, there are at least 1D-configuration amino acid residue, e.g., 1-5, e.g., 1 or 2D-configuration amino acid residues, e.g., D-Glu, D-Thr, D-Phe, D-Ala, etc.; the remaining amino acid residues are in the L-configuration.
In some examples, in formula I or formula II, the amino acid at position 3 is L-Glu or D-Glu.
In some examples, in formula I or formula II, the amino acid at position 5 is L-Thr or D-Thr.
In some examples, in formula I or formula II, the amino acid at position 6 is L-Phe or D-Phe.
In some examples, in formula I or formula II, the amino acid at position 35 is L-Ala or D-Ala.
In some examples, in any of formulas I-XXI, the carbon atoms of each amino acid residue can have pendant substituents independently of each other. The substituent is not particularly limited as long as it does not affect the desired properties of the compound according to the present disclosure. In some examples, the substituents are each independently selected from linear, branched, or cyclic, saturated or unsaturated aliphatic groups, or aromatic groups. Optionally, the substituents may be further substituted. In some examples, the substituents are, for example, C 1 -C 20 Alkyl groups such as methyl; c (C) 2 -C 20 Alkenyl groups such as pentenyl or decenyl; substituted or unsubstituted phenyl, such as p-chlorophenyl, pentafluorophenyl; and/or substituted or unsubstituted fused ring aryl, such as 1-tea. Substituted amino acid residues such as leucine residue substituted with methyl at the alpha carbon atom (alpha-meL), tryptophan residue substituted with methyl at the 2-position (2-me-Trp), alanine residue substituted with p-chlorophenyl (Cpa-Ala), and alanine residue substituted with pentafluorophenyl (Fpa 5-Ala).
In some examples, in formula I or formula II, the amino acid at position 14 is a leucine residue (α -meL) with an α carbon atom substituted with a methyl group.
In some examples, in formula I or formula II, the amino acid at position 25 is a tryptophan residue (2-me-Trp) in which the carbon atom at position 2 is substituted with a methyl group.
The term "C", as used herein 12 -C 24 By aliphatic diacid conjugated to an "amino acid" is meant any natural or unnatural amino acid having a functional group conjugated to the aliphatic diacid by covalent bonding, or preferably by way of a linker. Examples of conjugated functional groups of amino acids include amino, carboxyl, chloro, bromo, iodo, azido, alkynyl, alkenyl and mercapto, preferably amino. Examples of natural amino acids including such functional groups include lysine K (having an amino group), cysteine C (having a thiol group), glutamic acid E (having a carboxyl group), and aspartic acid D (having a carboxyl group).
In some examples, the amino acid that is conjugated is lysine K. In such embodiments, conjugation refers to an epsilon amino group conjugated to the lysine K side chain.
In some examples, the conjugation is acylation.
In some examples, the compounds of the invention include an aliphatic diacid moiety conjugated via a linker to the epsilon-amino group of the lysine K side chain at position 16 or 20.
In some examples, the compounds of the invention include aliphatic diacid moieties that are directly conjugated, without a linker, to natural or unnatural amino acids having functional groups available for conjugation.
In some examples, in any of formulas II through XX, at lysine (Lys) at position 16 or 20, C is attached via a direct bond or via a linker 12 -C 24 Chemical modification of the aliphatic diacid conjugated to the epsilon-amino group of the lysine (Lys) side chain, the linker being selected from (AEEA) 2 -(γ-Glu) a 、AEEA-Ahx-(γ-Glu) a 、(Ahx) 2 -(γ-Glu) a And (beta-Ala) 2 -(γ-Glu) a Wherein a is 1 or 2, thereby imparting excellent in vivo and in vitro activity to the compound. In some examples, the linker is selected from (AEEA) 2 -(γ-Glu)、AEEA-Ahx-(γ-Glu)、(Ahx) 2 - (gamma-Glu) and (beta-Ala) 2 -(γ-Glu)。
In the context of reference to linkers, AEEA is an abbreviation for [2- (2-amino-ethoxy) -ethoxy ] -acetyl, representing [2- (2-amino-ethoxy) -ethoxy ] -acetyl. gamma-Glu represents gamma-glutamyl. Ahx is an abbreviation for amino hexanoyl, representing aminocaproyl. beta-Ala represents beta-alanyl.
In some examples, in any of formulas II through XX, via a linker (AEEA) 2 -(γ-Glu)、AEEA-Ahx-(γ-Glu)、(Ahx) 2 - (gamma-Glu) or (beta-Ala) 2 - (gamma-Glu) chemically modifying octadecanedioic acid or eicosanedioic acid by conjugation to the epsilon-amino group of the lysine (Lys) side chain at position 16 or 20.
In some examples, in any of formulas III and V through XX, via (AEEA) 2 - (gamma-Glu) conjugated eicosanedioic acid to the epsilon-amino group of lysine (Lys) side chain at position 20.
In some examples, in formula IV, via (AEEA) 2 - (gamma-Glu) conjugated eicosanedioic acid to the epsilon-amino group of lysine (Lys) side chain at position 16.
In some examples, in formula IV, via (Ahx) 2 - (gamma-Glu) conjugated eicosanedioic acid to the epsilon-amino group of lysine (Lys) side chain at position 20.
In some examples, in formula IV, the chemical modification is performed via conjugation of eicosadiacid to the epsilon-amino group of the lysine (Lys) side chain at position 20, AEEA-Ahx- (gamma-Glu).
In some examples, in formula IV, via (. Beta. -Ala) 2 - (gamma-Glu) conjugated eicosanedioic acid to the epsilon-amino group of lysine (Lys) side chain at position 20.
Taking the compound NBB-T007-12 (corresponding to formula XVI) shown below as an example, at lysine at position 20, the first [2- (2-amino-ethoxy) -ethoxy group]An acetyl (AEEA) unit linked to the epsilon-amino group of the lysine side chain via an acyl group, a second [2- (2-amino-ethoxy) -ethoxy group]-acetyl (AEEA) unit with the first [2- (2-amino-ethoxy) -ethoxy ] through an acyl group]Amino linkage of the acetyl (AEEA) unit, gamma-glutamyl (gamma-Glu) with the second [2- (2-amino-ethoxy) -ethoxy ] via gammA-Acyl ]Amino linkage of an acetyl (AEEA) unit, eicosadioic acid (C 20 Diacid) and is linked to the amino group of gamma-glutamyl (gamma-Glu) via the terminal acyl group, thereby chemically modifying the epsilon-amino group of the side chain of lysine (Lys) at position 20.
According to the present disclosure, C is attached using a linker 12 -C 24 The conjugation of aliphatic diacids to the epsilon-amino group of the lysine (Lys) side chain of compounds of formulas I-XXI helps provide co-agonism to GLP-1 and GIP receptors for the compounds and provides the potential to produce long-acting compounds.
In some examples, in any of formulas II through XX, the histidine (His) or tyrosine (Tyr) at position 1 (i.e., the N-terminus) is amidated, e.g., by-C m H 2m+1 -C (O) -amidation linked to the amino group of histidine (His) or tyrosine (Tyr), and m is an integer from 1 to 30. In some examples, m is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30.
In some examples, m is 1 or 19. Preferably, in formula IV, the amino group of tyrosine 1 (Tyr) is bound to CH 3 -C (O) -or with C 19 H 39 C (O) -linked amidation as shown by the following compounds T007-19 (acetylation) and T007-20 (eicosanoylation), respectively.
In some examples, in any of formulas I-XXI, the α -carbon atoms of any two amino acids can be linked into a ring via a direct bond or via a linker selected from alkyl or alkenyl groups containing 2 to 20 carbon atoms. Such side chain modifications, also known as "stapling" alterations, serve to enhance the structural rigidity of the polypeptide and stabilize the activity of the polypeptide compound. When the linking group is an alkenyl group having 2 to 20 carbon atoms, the linking group may be introduced by using Grubbs catalyst. The cyclic alpha-carbon atoms may be derived from the side chains of two adjacent amino acids or from the side chains of two amino acids separated by at least 1 amino acid. The linker is an alkyl or alkenyl group containing 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 carbon atoms. In some examples, the linking group is an alkenyl group containing 10 carbon atoms. In some examples, in formula III, the α -carbon atom of alanine (Ala) at position 13 is linked to the α -carbon atom of alanine (Ala) at position 16 via an alkenyl group containing 10 carbon atoms.
In some examples, in any of formulas II-XX, the side chain carboxyl group of aspartic acid (Asp) or glutamic acid (Glu) and the side chain amino group of lysine (Lys), arginine (Arg), or histidine (His) may form a ring by forming an amide bond.
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
[ preparation method of GLP-1/GIP receptor Co-agonist ]
The compounds according to the present disclosure were prepared via a solid phase synthesis method using Rink-amide-AM resin (san fran and science and technology limited) as a synthesis carrier. The amino groups of the various amino acid starting materials used in the synthesis are protected by Fmoc groups (9-fluorenylmethoxycarbonyl, fluoronyl-methyloxy carbonyl, fmoc). For neutral polar amino acid, acidic amino acid and basic amino acid, proper protecting groups are selected to protect polar side chains of the amino acid raw materials according to different side chain functional groups. For example, but not limited to, the side chain thiol group of cysteine (Cys), the side chain amino group of glutamine (gin), the side chain imidazolyl group of histidine (His) and the side chain amino group of asparagine (Asn) are protected by Trt (trityl); the side chain guanidino group of arginine (Arg) is protected by Pbf (2, 4,6,7-pentamethyldihydrobenzofuran-5-sulfonyl, 2,4,6, 7-pentamethylhydrodynamic-5-sulfolane); the side chain indolyl group of tryptophan (Trp), the side chain hydroxyl group of serine (Ser) and the side chain amino group of lysine (Lys) are protected by Boc (tert-butoxycarbonyl protecting group); the side chain hydroxyl group of threonine (Thr) and the side chain phenol group of tyrosine (Tyr) are protected by tBu (tert-butyl); the side chain carboxyl group of aspartic acid (Asp) and the side chain carboxyl group of glutamic acid (Glu) are protected by OtBu (tert-butoxy).
In some examples, compounds according to the present disclosure are prepared by a method comprising the steps of:
i) Swelling and deprotection of resin Carrier
The Rink-Amide-AM resin, which had been protected by Fmoc, was swollen and then Fmoc protecting groups of the Rink-Amide-AM resin were removed with a solution of 20% piperidine in N, N-Dimethylformamide (DMF).
ii) formation of a polypeptide
Starting from the rightmost C-terminal of the compound structural formula, the amino acids are linked one by one, toward the leftmost N-terminal, to form a polypeptide in which an amide bond is formed by condensation of the carboxyl group of the amino acid whose amino group is protected by Fmoc:
first, the carboxyl group of the 1 st amino acid with Fmoc protection (from the rightmost C-terminus) was condensed as an amide bond onto the swelled and deprotected Rink-amide-AM resin using the condensing agent 6-Chlorobenzotriazole-1, 3-tetramethyluronium hexafluorophosphate (O- (1H-6-Chlorobenzotriazole-1-y 1) -1, 3-tetramethyluronium hexafluorophosphate, HCTU), and then the Fmoc protecting group on the amino group was removed with a solution of 20% piperidine in N, N-Dimethylformamide (DMF), followed by washing.
Then, in a similar manner, the condensing agent HCTU was used, and Fmoc-protected (from the rightmost C-terminus) amino acids at positions 2 through 9 were used, respectively, fmoc-protected 5-aminopentanoic acid (Fmoc-NH- (CH) 2 ) 4 -C (O) OH), and the amino acids at position 12 to 39 protected by Fmoc (from the rightmost C-terminus), the preceding cycles of amide bond forming coupling reaction, deprotection to remove Fmoc protecting group and washing are repeated sequentially.
iii) Deprotection and chemical modification of lysine at position 16 or 20 (from leftmost N-terminus)
Palladium tetraphenylphosphine Pd (PPh) 3 ) 4 Removing (from the leftmost N-terminal) the Boc protecting group on the epsilon-amino group of lysine at position 16 or 20, and washing.
Fmoc-protected linkers are then used, such as Fmoc-AEEA, fmoc-Ahx, or Fmoc-beta-Ala; and a condensing agent (HCTU) for coupling and deprotecting the epsilon-amino group of lysine at position 16 or 20 (from the leftmost N-terminus). In a similar manner, such coupling and deprotection operations are repeated 1 time, thereby ligating the 2 nd linker AEEA, ahx, or beta-Ala.
Then, fmoc-and OtBu-protected glutamic acid (Fmoc-Glu (OtBu) -OH) and condensing agent (HCTU) were used to couple with the amino group of the 2 nd linker, deprotected, and thereby the 3 rd linker gamma-Glu was ligated.
Then, adopt C 12 -C 24 An aliphatic diacid or derivative thereof (e.g., mono-t-butyl ester of the diacid) and a condensing agent (HCTU) coupled to the amino group of gamma-Glu to thereby join C 12 -C 24 Aliphatic diacids.
iv) cleavage and characterization of the Polypeptides
The polypeptide was cleaved from the resin support by reacting the cleavage reagent with Rink-Amide-AM resin using a trifluoroacetic acid (TFA, trifluoroacetic acid)/water/phenol/triisopropylsilane (Tips, triisopropyl silane) mixture as the cleavage reagent. And (5) settling by frozen glacial ethyl ether to obtain a solid crude product of the polypeptide. And (3) carrying out centrifugal separation, airing and triturating on the polypeptide solid crude product to obtain the purified polypeptide.
Electrospray mass spectrometry was performed on the purified polypeptide to determine molecular structure, and high performance liquid chromatography was used to analyze the purity of the purified polypeptide.
[ pharmaceutical composition ]
The present disclosure provides a pharmaceutical composition comprising:
a compound according to the present disclosure or a pharmaceutically acceptable salt thereof, and
a pharmaceutically acceptable carrier, diluent or excipient.
In addition to one or more compounds according to the present disclosure or pharmaceutically acceptable salts thereof, the pharmaceutical compositions may also contain other components, such as physiological/pharmaceutically acceptable carriers, diluents and excipients, to facilitate administration to a subject, facilitate absorption of the compound or pharmaceutically acceptable salt thereof as an active ingredient for biological activity.
By "pharmaceutically acceptable salts" is meant salts of the compounds according to the present disclosure which are safe and effective when used in a subject, and which are biologically active.
The compounds according to the present disclosure may react with a variety of inorganic and organic acids to form pharmaceutically acceptable salts. Pharmaceutically acceptable salts and common methods for preparing them are well known in the art. See, for example, PStahl et al, handbook of Pharmaceutical Salts: properties, selection and Use, second revision (Wiley-VCH, 2011); S.M. Berge et al, "Pharmaceutical Salts", journal of Pharmaceutical Sciences, vol.66, no.1, 1977, month 1.
In some examples, a compound according to the present disclosure or a pharmaceutically acceptable salt thereof may be formulated into a pharmaceutical composition for administration by parenteral route (e.g., subcutaneous, intravenous, intraperitoneal, intramuscular, or transdermal). Such pharmaceutical compositions and methods for their preparation are well known in the art. See, for example, remington: the Science and Practice of Pharmacy (D.B. Troy edit, 21 st edition, lippincott, williams & Wilkins, 2006).
Pharmaceutically acceptable salts according to the present disclosure include, but are not limited to, trifluoroacetate, hydrochloride, and acetate salts.
[ use ]
The present disclosure provides the use of a pharmaceutical composition according to the present disclosure or a compound according to the present disclosure or a pharmaceutically acceptable salt thereof for the preparation of a medicament for the treatment and/or prevention of a metabolic disease or disorder;
in particular, the metabolic diseases or disorders include diabetes and diabetes-related conditions, as well as obesity and obesity-related conditions;
in particular, the diabetes and diabetes-related disorders include insulin resistance, glucose intolerance, elevated fasting blood glucose, prediabetes, type I diabetes, type II diabetes (T2 DM), gestational diabetes hypertension, dyslipidemia, atherosclerosis, arteriosclerosis, coronary heart disease, peripheral arterial disease, and dyslipidemia that actuates atherosclerosis, dyslipidemia, elevated blood pressure, hypertension, pre-thrombotic and pro-inflammatory states, and combinations thereof;
in particular, obesity and obesity-related disorders include obesity-related inflammation, obesity-related cholecystitis, obesity-induced sleep apnea, non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), and combinations thereof.
In some examples, the medicaments of the present disclosure are used to treat type II diabetes.
[ methods of treating and/or preventing metabolic diseases or disorders ]
The present disclosure provides a method of treating and/or preventing a metabolic disease or disorder comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition according to the present disclosure or a compound according to the present disclosure or a pharmaceutically acceptable salt thereof.
In some examples, the pharmaceutical composition, the compound, or a pharmaceutically acceptable salt thereof is administered to the individual by subcutaneous injection.
In some examples, the pharmaceutical composition, the compound, or a pharmaceutically acceptable salt thereof is administered to the subject once a week.
In some examples, the pharmaceutical composition, the compound, or a pharmaceutically acceptable salt thereof is administered to the subject once a week by subcutaneous injection.
Examples (example)
[ preparation example ]
Experimental reagent:
as used herein, unless otherwise indicated, N-Dimethylformamide (DMF) and Dichloromethane (DCM) are common reagents having a purity of 99.7% and excipients are NaHCO 0.05% 3 A solution. The 20% piperidine solution refers to a volume percentage, which can be obtained, for example, by: 100mL of piperidine was measured with a graduated cylinder and DMF was added to the graduated cylinder scale of 500mL.
Unless otherwise indicated, in embodiments of the present disclosure, draining the solvent refers to, for example, pumping the solvent in the polypeptide synthesis tube into a pump flask with an air pump, draining to the resin as a dry powder.
[ preparation of Compound NBB-T007 ]
(1) Swelling of resin
a) 0.63g (0.2 mmol) of Fmoc-protected Rink-Amide-AM resin was weighed and placed in a polypeptide synthesis tube.
b) To the polypeptide synthesis tube were added 10mL of DMF (N, N-dimethylformamide) and 10mL of DCM (dichloromethane), and the mixture was left at room temperature for 30min.
c) The solvent was pumped dry with an air pump.
d) After rinsing with 10ml of LDMF, the solvent was drained.
(2) Deprotection of resins
a) 10mL of 20% piperidine solution was added to the polypeptide synthesis tube to submerge the swollen resin obtained in step (1), and the swollen resin was transferred to a 33℃constant temperature shaker for 5min.
b) The polypeptide synthesis tube was removed from the shaker.
c) Cleaning: the resin was washed three times (10 mL each) with DMF and the solvent was drained; the resin was rinsed three times (10 mL each) with DCM and the solvent was drained; finally, the solvent was drained after three more DMF washes (10 mL each).
d) Deprotection: 10mL of 20% piperidine solution was added to the polypeptide synthesis tube, and the mixture was shaken in a shaking table at a constant temperature of 33℃for 10min, and the polypeptide synthesis tube was removed.
e) Repeating the washing step c) in the "resin deprotection" of the step (2).
Then, the formation of the polypeptide is performed by: the amino acids are linked one by one starting from the 1 st amino acid at the rightmost C-terminus of the polypeptide chain and proceeding toward the leftmost N-terminus.
(3) The 1 st amino acid (from the rightmost C-terminal)
a) 310mg (0.8 mmol) of Fmoc-Ser (Boc) -OH (serine) and 314mg (0.8 mmol) of condensing agent (HCTU) were weighed into a 10mL EP tube, 6mL of the EP tube was dissolved by adding 6mL of the solution to the EP tube, shaking the solution thoroughly, and 265. Mu.L (1.6 mmol) of N, N-Diisopropylethylamine (DIEA) was added to the EP tube to obtain a mixed solution.
b) Transferring the mixed solution into a polypeptide synthesis tube, transferring the polypeptide synthesis tube into a shaking table at a constant temperature of 33 ℃ for 1h, and taking out the polypeptide synthesis tube.
c) Cleaning: repeating the washing step c) in the "resin deprotection" of the step (2).
d) Deprotection: 10mL of 20% piperidine solution was added to the polypeptide synthesis tube, and the mixture was shaken in a shaking table at a constant temperature of 33℃for 10min, and the polypeptide synthesis tube was removed.
(4) From amino acid 2 to amino acid 9 (from the rightmost C-terminus)
Similarly to the 1 st amino acid from the rightmost C-terminal, the 2 nd to 9 th amino acids from the rightmost C-terminal are sequentially accessed (proline), except that Fmoc-Pro-OH (proline), fmoc-Ala-OH (alanine), fmoc-Gly-OH (glycine), fmoc-Ser (Boc) -OH (serine) and Fmoc-Pro-OH (proline) are used as the starting materials for the coupling (from the rightmost C-terminal) of the 2 nd to 9 th amino acids, respectively.
(5) "10 th and 11 th" amino acids (from the rightmost C-terminus)
a) 205mg (0.6 mmol) of Fmoc-5-aminopentanoic acid (formula: fmoc-NH- (CH) 2 ) 4 C (O) -OH) and 235mg (0.6 mmol) of condensing agent (HCTU) were placed in a 10mL EP tube, 6mL of LDMF was added to the EP tube for dissolution, and the mixture was thoroughly shaken and 265. Mu.L (1.6 mmol) of DIEA was added to the EP tube to obtain a mixed solution.
b) Shaking the mixed solution uniformly and transferring the mixed solution into a polypeptide synthesis tube; transferring the polypeptide synthesis tube to a shaking table at a constant temperature of 33 ℃ for shaking for 1h, and taking out the polypeptide synthesis tube.
c) Cleaning: repeating the washing step c) in the "resin deprotection" of the step (2).
d) Deprotection: 10mL of 20% piperidine solution was added to the polypeptide synthesis tube, and the mixture was shaken in a shaking table at a constant temperature of 33℃for 10min, and the polypeptide synthesis tube was removed.
(6) From amino acid 12 to 39 (from the rightmost C-terminus)
Similar to the 1 st amino acid from the rightmost C-terminus, the 12 th to 39 th amino acids from the rightmost C-terminus are sequentially accessed (from the rightmost C-terminus), except that Fmoc-Ala-OH (alanine), fmoc-Ile-OH (isoleucine), fmoc-Leu-OH (leucine), fmoc-Trp (Boc) -OH (tryptophan), fmoc-Gln (Trt) -OH (glutamine), fmoc-Val-OH (valine), fmoc-Phe-OH (phenylalanine), fmoc-Ala-OH (alanine), fmoc-Lys (Boc) -OH (lysine), fmoc-Gln (Trt) -OH (glutamine), fmoc-Ala-OH (alanine), fmoc-Ile-OH (isoleucine), fmoc-Leu-OH (Boc) -OH (leucine), fmoc-Asp (OtBu) -OH (tryptophan), fmoc-Val-OH (valine), fmoc-Phe-OH (phenylalanine), fmoc-Phe-OH (Ala) -OH (alanine), fmoc-Lys (Boc) -OH (lysine), fmoc-Ile-OH (isoleucine), fmoc-OH (Leu) and Fmoc-OH (Ile), fmoc-Ser (Boc) -OH (serine), fmoc-Thr (tBu) -OH (threonine), fmoc-D-Phe-OH (phenylalanine), fmoc-D-Thr (tBu) -OH (threonine), fmoc-Gly-OH (glycine), fmoc-Glu (OtBu) -OH (glutamic acid), fmoc- (Aib) -OH (2-aminoisobutyric acid) and Fmoc-Tyr (tBu) -OH (tyrosine) were used as starting materials for the coupling (from the rightmost C-terminal) of the 12 th to 39 th amino acids.
(7) Removing Boc at position 20 Lys (Boc) from the leftmost N-terminal
After completion of the peptide chain extension, the resin was washed with 10mL of DMF, 10mL of DCM, and 10mL of DMF, respectively.
Then, 116mg (0.1 mmol) of palladium tetraphenyl phosphine was weighed into a 10mL EP tube, 3mL of DCM and 3mL of DMF were added to the EP tube to dissolve, and the mixture was thoroughly mixed. To the EP tube was added 124. Mu.L (1 mmol) of phenylsilane, and the solution was transferred to the polypeptide synthesis tube by shaking thoroughly. Transferring the polypeptide synthesis tube to a 33 ℃ constant temperature shaking table for shaking for 2 hours, and taking out and cleaning.
Starting from the addition of the palladium tetraphenyl phosphine described above, the entire step of Boc removal was repeated 1 time until the washing step.
(8) Side chain modified ligation
a) After washing, the first AEEA was coupled to the side chain of lysine 20 (from the leftmost N-terminus): 231mg (0.6 mmol) of Fmoc-AEEA and 235mg (0.6 mmol) of HCTU were dissolved in DMF and 200. Mu.L (1.2 mmol) of DIEA were added and after mixing well transferred to a multipeptidation synthesis tube. The polypeptide synthesis tube was transferred to a thermostatically shaking table and shaken for 1h at room temperature. Repeating the step (2) 'resin deprotection'.
b) Coupling a second AEEA: 231mg (0.6 mmol) of Fmoc-AEEA and 235mg (0.6 mmol) of HCTU were dissolved in DMF and 200. Mu.L (1.2 mmol) of DIEA were added and after mixing well transferred to a multipeptidation synthesis tube. The polypeptide synthesis tube was transferred to a thermostatically shaking table and shaken for 1h at room temperature. Repeating the step (2) 'resin deprotection'.
c) Coupling gamma-Glu: 255mg (0.6 mmol) of Fmoc-Glu- (OtBu) -OH and 235mg (0.6 mmol) of HCTU were dissolved in DMF and 200. Mu.L (1.2 mmol) of DIEA were added and transferred to a polypeptide synthesis tube after mixing well. The polypeptide synthesis tube was transferred to a thermostatically shaking table and shaken for 1h at room temperature. Repeating the step (2) 'resin deprotection'.
d) Coupling eicosanedioic acid: 240mg (0.6 mmol) of mono-tert-butyl eicosadioate and 235mg (0.6 mmol) of HCTU were dissolved in DMF and 200. Mu.L (1.2 mmol) of DIEA were added and transferred to a multipeptidation synthesis tube after mixing well. The polypeptide synthesis tube was transferred to a thermostatically shaking table and shaken for 1h at room temperature.
(9) Cleavage of crude peptide
The polypeptide synthesis tube was removed and the resin was washed three times (10 mL each) with DMF, and the solvent was drained after each wash. The resin was rinsed three times (10 mL each) with DCM and the solvent was drained after each rinse (drained until the resin was dry). After draining, TFA (trifluoroacetic acid)/H was formulated in a 10mL EP tube 2 O/phenol/Tips (volume ratio: 10 mL/500. Mu.L/500 mg/500. Mu.L) cleavage reagent. Transferring the cutting reagent into the polypeptide synthesis tube, placing the polypeptide synthesis tube into a shaking table at a constant temperature of 26 ℃ for oscillating reaction for 2.5 hours, and taking out the polypeptide synthesis tube, wherein the solution in the tube is the peptide chain lysate.
(10) Post-treatment of polypeptides
a) 10mL of the peptide chain lysate was transferred to a 50mL centrifuge tube using an ear-washing ball, and the lysate was dried at room temperature with nitrogen as much as possible to 5mL or less.
b) Precipitation with glacial diethyl ether and centrifugation: adding 40mL of glacial diethyl ether into 50mL of centrifuge tube, properly vibrating the centrifuge tube, putting the centrifuge tube into a centrifuge, and centrifuging for 3min at 3500 rpm; after centrifugation, the supernatant was decanted.
c) Repeating the above steps of precipitation with glacial ethyl ether and centrifugation, and discarding supernatant to obtain precipitate as crude peptide.
d) Air-drying at room temperature, and mashing to obtain purified polypeptide.
Purified polypeptides were isolated using Shimadzu semi-preparative liquid chromatography. FIG. 1 is a representation of ESI-MS (electrospray mass spectrometry) performed on purified polypeptide compound NBB-T007, with peaks representing molecular weights of different mass to charge ratios. FIG. 2 is a high performance liquid chromatogram showing that NBB-T007 was synthesized with 95% purity.
[ preparation of other Compounds of NBB-T007 series and Compound NBB-EX4 ]
Other compounds of the NBB-T007 series and compound NBB-EX4 were prepared in a similar manner to the preparation of compound NBB-T007 except that Fmoc protected amino acid starting materials, linker starting materials and/or sources of aliphatic diacids, and other various modifying groups, if any, were varied according to the structural formula of each compound. Through high performance liquid chromatography analysis, the purity of the compounds is more than or equal to 95 percent.
Wherein Fmoc-S5-OH (Cas No.: 288617-73-2) and Fmoc-R5-OH (Cas No.: 288617-77-6) are used as amino acid raw materials at positions 24 and 27 (from the rightmost C-terminal) respectively, and after grafting (from the rightmost C-terminal) position 27 Fmoc-R5-OH, 64mg of Grubbs catalyst (Cas No. 172222-30-9) to a 10mL EP tube is weighed and dissolved in 4mL of DCM. After mixing well, the mixed solution was transferred to a polypeptide synthesis tube. And transferring the polypeptide synthesis tube to a constant temperature shaking table at 33 ℃, and taking out the polypeptide synthesis tube after shaking for 4 hours. Under the action of Grubbs catalyst, the side chains of the R5 unit and the S5 unit undergo olefin metathesis reaction, and cyclize. After this step is completed, the sequence of amino acids 28 to 39 (from the rightmost C-terminus) is continued.
[ in vitro Activity assay: cAMP detection ]
Experimental reagent
Experimental equipment
Name of the name Company (Corp) Model number
Enzyme label instrument TECAN INFINITE 200 PRO
Carbon dioxide incubator Siemens fly 3131
Biological clean workbench Suzhou AnQin air technologies Co.Ltd BLB-1300
Microscope Nikon ECLIPSE Ts2-FL
Cell counter BIO-RAD TC20 TM
Refrigerator with a refrigerator body Sea Er BCD-252WXPS
Medical low-temperature refrigerator Siemens fly 902-ULTS
The human GLP-1R receptor, the GIPR receptor and the GCGR receptor were cloned separately into pcDNA3.1 vector. pcDNA3.1-GLP1R, pcDNA3.1-GIPR and pcDNA3.1-GCGR were transfected into HKE293T cells cultured in 35mm dishes, respectively, and cultured in a carbon dioxide incubator for 24 hours using Lipofectamin3000 transffectionkit. Cells were resuspended with DMEM containing 10% FBS, 1% P/S and 500. Mu. MIBMX, 20. Mu.L was removed for cell counting and diluted to 2X 10 6 cells/mL, 5 μl of diluted DMEM resuspended cells were plated in 384-well plates. mu.L of DMSO was then added to each well of the well plate and diluted with a ten-fold gradient of DMEM containing 500. Mu.M IBMX (2X 10) -6 、2×10 -7 、…、2×10 -15 M) any of the following polypeptides prepared in the previous examples: NBB-T007 series compound, compound NBB-EX4, or comparative cable Ma Lutai (home made) were incubated at 37℃for 30 min. After addition of 5. Mu.L of each of cAMP-d2 and anti-cAMP in cAMP-Gs Dynamic kit and incubation at room temperature for 1 hour, reading was performed with a TECAN microplate reader at excitation wavelength of 340nm and emission wavelengths of 620nm and 655nm. Calculating the signal ratio (655 nm/620 nm. Times.10,000), and performing nonlinear fitting on the signal ratio and the sample concentration in GraphPad Prism 8 by using a four-parameter equation to obtain EC 50 Values, see fig. 3-26 and tables below.
As can be seen from the above table, somalupeptide has activity only against human GLP-1R, whereas the NBB-T007 series of compounds according to the present disclosure (except for the compound NBB-T007-7) surprisingly show dual activity against human GIP receptor and GLP-1 receptor. In particular, the compounds NBB-T007-1, NBB-T007-5, NBB-T007-8, NBB-T007-10, NBB-T007-12, NBB-T007-18, NBB-T007-19, NBB-T007-21, NBB-T007-22, NBB-T007-23 and NBB-T007-24 have triple activity on human GIP receptor, GLP-1 receptor and GCG receptor.
The compound NBB-EX4 has high activity against human GLP-1R, which indicates that by adopting-CH 2 -CH 2 -unit substitution of two adjacent glycine-NH-CH at positions 29 and 30 2 -C(O)-NH-CH 2 The scheme of modification of peptide chains by the peptide bonds comprised by-C (O) -is widely applicable to polypeptide drugs and is capable of maintaining high activity against human GLP-1R.
[ db/db mice (type II diabetes mice) hypoglycemic test ]
Male db/db mice (Kwangsi) 7-8 weeks old were used, each weighing 33-40g. These mice were individually placed in a temperature controlled (22-25 ℃) facility, cycled light/dark (illumination starting at 08:00) for 12 hours, and food and water were freely available. After 1 week of acclimation to the facility, the treatment groups (n=6/group) were randomly assigned according to body weight and blood glucose such that each group had similar initial average body weight and blood glucose concentration. Excipient controls ("solvent set", 0.05% NaHCO) 3 Solutions), cord Ma Lutai control dissolved in vehicle (dose 50 nmol/kg) and NBB-T007 series compound prepared in the previous example (dose 50 nmol/kg) were administered by subcutaneous injection to freely feeding db/db mice, blood glucose values were measured from the tail vein using a steady hao rapid glucometer (One Touch UltraEasy, prednisone) for 48 consecutive hours. Meanwhile, the body weight of db/db mice was monitored at 0h, 24h and 72 h. The test results are shown in the following table and fig. 27 to 29.
Formula IV: conjugation of eicosadioic acid using a linker (AEEA-Ahx- (γ -Glu) at lysine (Lys) at position 20 of formula IV;
formula IV: eicosadioic acid was conjugated at lysine (Lys) at position 20 of formula IV using linker (. Beta. -Ala) 2- (gamma. -Glu).
As can be seen, the NBB-T007 series compounds according to the present disclosure have significant hypoglycemic effects comparable to or better than cord Ma Lutai than cord Ma Lutai and can maintain the pharmacodynamic effects for 48 hours; of these, the compounds NBB-T007, NBB-T007-10 and NBB-T007-12 have particularly better hypoglycemic and weight-reducing effects.
[ Long-term administration hypoglycemic Effect of db/db mice (type II diabetic mice) ]
Male db/db mice (Kwangsi) 7-8 weeks old were used, each weighing 33-40g. These mice were individually placed in a temperature controlled (22-25 ℃) facility, cycled light/dark (illumination starting at 08:00) for 12 hours, and food and water were freely available. After 1 week of acclimation to the facility, the treatment groups (n=6/group) were randomly assigned according to body weight and blood glucose, so each group had similar initial average body weight and blood glucose concentration. Respectively using NaHCO dissolved in excipient (0.05% 3 Solution), the telogen control (dose 30 nmol/kg), the telogen Tirzepatide control (dose 30 nmol/kg), and the NBB-T007-12 compound prepared in the previous example (dose 30 nmol/kg) were administered by subcutaneous injection to freely fed db/db mice twice weekly for a period of 12 weeks. During the administration period, the body weight, food intake and blood glucose of the mice were monitored, and the measurement of glycosylated hemoglobin HbA1C was performed by taking blood at week 2, week 4, week 6, week 8 and week 10, respectively. The test results of the inhibition (%) of glycosylated hemoglobin are shown in the following table.
It can be seen that the tested compounds have a remarkable reduction effect on glycosylated hemoglobin of type II diabetic mice after long-term administration, suggesting that the blood glucose of db/db mice can be effectively improved.
[ sugar tolerance IPGTT test in Normal mice ]
C57BL/6 mice (Beijing vitamin Torilhua), males, 7-10 weeks old, and body weight 18-20g. These mice were individually placed in a temperature controlled (22-25 ℃) facility, cycled light/dark (illumination starting at 08:00) for 12 hours, and food and water were freely available. Mice were randomized after acclimation to the facility, 6 per group, randomized in terms of blood glucose and body weight, so that each group had similar initial average body weight and blood glucose concentration. Mice were fasted overnight for 12-16 hours, and the next day the mice were weighed and assayed for 0min blood glucose using a steady hao rapid glucometer (OneTouch UltraEasy, predominance).
The following test substances were used respectively: vehicle control ("solvent set", 0.05% NaHCO) 3 Solution), as well as cord Ma Lutai control (25 nmol/kg) dissolved in vehicle and NBB-T007 series compound prepared in the previous example (dose 25 nmol/kg), were administered to C57BL/6 mice fed freely by subcutaneous injection, and glucose solution (2 g/kg) was simultaneously administered intraperitoneally, and blood glucose values of 15min, 30min, 60min, 120min after administration were determined. The blood glucose levels at 15min, 30min, 60min and 120min after the administration were measured without administering the test substance and by administering a glucose solution (2 g/kg) to the abdominal cavity at the same time as the second day and the third day. The area under the blood glucose-time curve is calculated AUC (area under the curve). And, body weights of mice were monitored at 24h, 48h and 72h, respectively, after administration. The test results are shown in the following table and fig. 30 to 31.
From the above sugar tolerance experiments, it can be seen that the NBB-T007 series compounds according to the present disclosure had a significant hypoglycemic effect over that of cable Ma Lutai, and that the glucose AUC was reduced in the test on the first, second and third days.
[ in vivo Activity test: pharmacokinetic analysis of SD rats ]
SD rats (Beijing velutinal), males, 8-10 weeks, and body weight 180-200g were selected. Rats were individually placed in a temperature controlled (22-25 ℃) facility, cycled 12 hours light/dark (illumination starting at 08:00), and food and water were freely available. Rats were randomized after acclimation to the facility, 3 per group. Excipient control (0.05% NaHCO) 3 Solutions), NBB-T007 series compound (dose 1 mg/kg) prepared in previous examples dissolved in vehicle and Soxhlet Ma Lutai (dose 1 mg/kg), 0.25h, 0.5h, 1h, 2h, 4h, 8h, 24h, 48h, 72h after administration, jugular vein harvest 0.3m, respectivelyThe L venous blood was placed in EDTA2K anticoagulation tube, centrifuged at 8000rpm for 5 minutes to collect plasma, the plasma concentration of the test substance was determined by LC-MS/MS method, and the compound of the plasma sample was extracted with methanol, and the sample treatment steps were as follows:
samples were 30.0. Mu.L, 50.0. Mu.L of internal standard solution (Soxhlet Ma Lutai, 20,000 ng/mL) and 200. Mu.L of methanol, vortexed for 10min, centrifuged for 10min (3900 rpm), and the supernatants were removed to another clean 96-well plate for LC-MS/MS analysis. The test results are shown in the following table and in fig. 32 to 35.
T i/2 =half-life, T max Time to maximum concentration, C max Maximum plasma concentration, AUC last AUC, AUC of the period from the start of administration time to the last point INF AUC, MRT of_obs from the start of administration to the time of theoretical extrapolation to infinity INF_obs Average residence time from zero time to infinite time.
It can be seen that compounds NBB-T007, NBB-T007-10 and NBB-T007-12 reached an average maximum plasma concentration about 8 hours after subcutaneous administration. Wherein the half-lives of compounds NBB-T007 and NBB-T007-12 in rats were 11.82 and 9.29 hours, respectively, supporting the possibility of once weekly dosing.
[ in vivo Activity test: pharmacokinetic analysis of cynomolgus monkey
Male cynomolgus monkeys weighing 3-6kg were selected for pharmacokinetic analysis in non-rodents. Single intravenous administration of NaHCO dissolved in vehicle (0.05% NaHCO) 3 Solution) NBB-T007-12 (dose 0.5 mg/kg) and Tirzepatide control (dose 0.5 mg/kg). Before administration (0 h), and after administration 2h, 4h, 8h, 12h, 24h, 48h, 72h, 96h, 120h, 144h, 168h, 204h, 240h and 312h respectively take 0.5mL of blood through forelimb veins, put into an EDTA-K2 test tube, temporarily store in an ice water bath after whole blood collection, centrifuge for 5min at 11000rpm within 30min, separate plasma, and place in a separated plasma refrigerator for freezing to be tested. Detection of protogenic drug concentration in plasma Using LC-MS/MS method . Calculating relevant pharmacokinetic parameters T by adopting WinNonlin software max 、C max 、AUC last 、AUC 0-t (AUC from the start of administration to time t), AUC INF Obs (AUC from the start of administration to the time of theoretical extrapolation of infinity), T 1/2 CL, etc. The test results are shown in the following table.
T i/2 Half-life, C max Maximum plasma concentration, AUC last From the beginning to the end of the administration time
AUC for this time period for one point.
It can be seen that the half-life of compound NBB-T007-12 in cynomolgus monkeys for a single intravenous administration was 46.85 hours, further supporting the possibility of once weekly administration.
[ fat DIO mice weight-reducing and blood sugar-reducing Effect ]
70C 57BL/6 mice (Nanjing Jieqiang) are selected, the temperature of the male and the environment of the animal house are kept at 23+/-2 ℃ and the humidity is 40-70%, and the brightness and the darkness are alternated (illumination starts from 08:00) for 12 hours. 4-5 mice were kept per cage, with padding changed twice per week. The high-fat feed (60%Kcal fat,D12492) is fed for 10-12 weeks, the weight of the mice is 38-45g at the beginning of the experiment, the weight of the mice is measured to be more than 30% higher than that of normal diet animals, and the mice are randomly grouped after random blood sugar and weight are detected, and 8-10 mice are in each group. 8 mice were fed normal feed as normal model group. The mice fed with the high fat diet were randomly divided into a model control group ("solvent group"), a cable Ma Lutai group (administration dose 50 hmol/kg), a NBB-T007 group (administration dose 50 nmol/kg), a NBB-T007-10 group (administration dose 50 nmol/kg), and a NBB-T007-12 group (administration dose 50 nmol/kg). Mice were given subcutaneously twice weekly with 5mL/kg of dosing volume for the above test subjects, and vehicle controls (0.05% NaHCO) were given to the normal model group and model control group ("solvent group") 3 Solution), for 4 weeks. Stopping administration, or continuing administration. The following criteria were tested:
1. mice body weight and consumption were measured twice a week during the experiment, and the test results are shown in fig. 36.
As can be seen from fig. 36, after 40 days, the body weight of both normal model group and model control group ("solvent group") mice increased; the body weight of mice administered with cable Ma Lutai was almost unchanged, and the body weights of the mice administered with the compounds NBB-T007, NBB-T007-10 and NBB-T007-12 prepared in the previous examples were significantly reduced, wherein the body weights of the mice administered with the compounds NBB-T007-10 and NBB-T007-12 prepared in the previous examples were reduced by about 20%.
3. Insulin resistance ITT experiments: after 72 hours of the last dose study, the animals were enrolled for ITT studies. After the animals fasted for 1h, the blood glucose value of 0h was measured, and after 1U/kg of insulin injection was injected intraperitoneally, the blood glucose was measured for 15min, 30min, and 1h, and the test results are shown in FIG. 37.
As can be seen from FIG. 37, the test mice administered with the compounds NBB-T007, NBB-T007-10 and NBB-T007-12 prepared in the previous examples had better insulin sensitivity than the mice administered with cable Ma Lutai.
4. Sugar tolerance IPGTT experiment: animals were given the test subjects 2h after the ITT study was completed, and animals were given the IPGTT study 72h later. After 16h of overnight fasted animals, the tail tip was blood collected for testing for 0h of fasted blood glucose. The glucose solution with concentration of 2g/kg was injected intraperitoneally, and the test solution was simultaneously injected subcutaneously to test blood glucose of animals for 15min, 30min, 1h, and 2h (the second drop of blood was used for all blood glucose tests), and the test results are shown in fig. 38.
As can be seen from FIG. 38, the test mice administered the compounds NBB-T007, NBB-T007-10 and NBB-T007-12 prepared in the previous examples had comparable or better postprandial glycemic control than the mice administered the cable Ma Lutai.
5. Serum biochemistry/serum insulin, C-peptide/blood HbA1C: at the end of ITT study, animals were euthanized with carbon dioxide, hearts were collected, heparin sodium anticoagulated whole blood was collected, and blood was divided into two parts, one part was about 120 μl for testing HbA1C levels, and the other part was 500 μl whole blood was serum-separated for testing serum insulin, and conventional biochemical index tests (total cholesterol CHO, triglyceride TG, high density lipoprotein HDL, low density lipoprotein LDL, free fatty acid NEFA, UREA ura, inosine CREA (creatine), albumin ALB (album), total bilirubin TBIL (total bilirubin), glutamic pyruvic transaminase ALT, glutamic pyruvic transaminase AST), and test results were shown in fig. 39 (a), 39 (b) and 39 (C).
As can be seen from fig. 39 (a), 39 (b) and 39 (c), the model control group exhibited a typical diabetes index of high serum insulin content. The test mice administered the compounds NBB-T007, NBB-T007-10 and NBB-T007-12 prepared in the previous examples had a comparable or better trend of decreasing serum insulin levels and a comparable or better function of protecting liver function, protecting kidney function and improving lipid metabolism compared to mice administered rope Ma Lutai.
6. General anatomy/fat weight weighing: after the carbon dioxide of the animal is euthanized, the viscera change is observed, the fat of the abdomen or epididymis of the animal is collected, the weight is weighed, the weight coefficient is calculated, the liver tissue of the animal is collected, and the triglyceride TG is detected. The test results are shown in fig. 40 (a) and 40 (b).
From FIGS. 40 (a) and 40 (b), it can be seen that the mice tested administered the compounds NBB-T007, NBB-T007-10 and NBB-T007-12 prepared in the previous examples had comparable or better postprandial body fat rate reduction and triglyceride reduction compared to the mice administered the cord Ma Lutai.

Claims (37)

1. A compound of formula I:
L 1 -NH-(CH 2 ) n -C(O)-L 2 -NH 2 i is a kind of
Wherein, the liquid crystal display device comprises a liquid crystal display device,
L 1 peptide analogs of the GIP (1-28) peptide, said L 1 Is a peptide consisting of 28 amino acids, and the L 1 Has at least 39% identity with SEQ ID NO. 1,
L 2 is a peptide having an amino acid sequence consisting of SEQ ID NO. 2,
n is any integer from 2 to 6, and
the compounds have GLP-1 receptor agonist activity, or GIP receptor agonist activity, or both.
2. The compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein L is compared to SEQ ID No. 1 1 Comprising Y1H substitutions.
3. The compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof, wherein L is compared to SEQ ID No. 1 1 Comprising a substitution selected from the group consisting of A2G, A2 (Aib) and A2 (beta-Ala).
4. A compound according to any one of claims 1-3, or a pharmaceutically acceptable salt thereof, wherein L is compared to SEQ ID No. 1 1 Comprising a T5S substitution.
5. The compound according to any one of claims 1-4, or a pharmaceutically acceptable salt thereof, wherein L is compared to SEQ ID No. 1 1 Comprising a substitution selected from F6 (homo-Phe) and F6 (Cpa-Ala).
6. The compound according to any one of claims 1-5, or a pharmaceutically acceptable salt thereof, wherein L is compared to SEQ ID No. 1 1 Comprising an I7T substitution.
7. The compound according to any one of claims 1-6, or a pharmaceutically acceptable salt thereof, wherein L is compared to SEQ ID No. 1 1 Comprising a substitution selected from Y10L and Y10 (Fpa 5-Ala).
8. The compound of any one of claims 1-7, or a pharmaceutically acceptable salt thereof, wherein L is compared to SEQ ID No. 1 1 Comprising an I12K substitution.
9. The compound of any one of claims 1-8, or a pharmaceutically acceptable salt thereof, wherein L is compared to SEQ ID No. 1 1 Comprising a substitution selected from a13Q, A (Aib) and a 13Y.
10. According to claimThe compound of any one of claims 1-9, or a pharmaceutically acceptable salt thereof, wherein L is compared to SEQ ID No. 1 1 Comprising a substitution selected from M14L and M14 (. Alpha. -meL).
11. The compound according to any one of claims 1-10, or a pharmaceutically acceptable salt thereof, wherein L is compared to SEQ ID No. 1 1 Comprising a D15E substitution.
12. The compound according to any one of claims 1-11, or a pharmaceutically acceptable salt thereof, wherein L is compared to SEQ ID No. 1 1 Comprising a substitution selected from the group consisting of K16E and K16 (Ala).
13. The compound of any one of claims 1-12, or a pharmaceutically acceptable salt thereof, wherein L is compared to SEQ ID No. 1 1 Comprising an I17E substitution.
14. The compound of any one of claims 1-13, or a pharmaceutically acceptable salt thereof, wherein L is compared to SEQ ID No. 1 1 Comprising a substitution selected from H18A and H18 (Aib).
15. The compound of any one of claims 1-14, or a pharmaceutically acceptable salt thereof, wherein L is compared to SEQ ID No. 1 1 Comprising a Q19V substitution.
16. The compound of any one of claims 1-15, or a pharmaceutically acceptable salt thereof, wherein L is compared to SEQ ID No. 1 1 Comprising a substitution selected from Q20R and Q20K.
17. The compound of any one of claims 1-16, or a pharmaceutically acceptable salt thereof, wherein L is compared to SEQ ID No. 1 1 Comprising a substitution selected from the group consisting of D21L, D21A, D E and D21 (Abu).
18. A compound according to any one of claims 1-17 or a pharmaceutically acceptable thereofAn acceptable salt, wherein L is compared to SEQ ID NO. 1 1 Comprising a substitution selected from V23I and V23L.
19. The compound of any one of claims 1-18, or a pharmaceutically acceptable salt thereof, wherein L is compared to SEQ ID No. 1 1 Comprising a substitution selected from the group consisting of N24E and N24Q.
20. The compound of any one of claims 1-19, or a pharmaceutically acceptable salt thereof, wherein L is compared to SEQ ID No. 1 1 Comprising a substitution selected from the group consisting of W25 (Na 1) and W25 (2-me-Trp).
21. The compound of any one of claims 1-20, or a pharmaceutically acceptable salt thereof, wherein L is compared to SEQ ID No. 1 1 Comprising a substitution selected from the group consisting of L27K and L27I.
22. The compound of any one of claims 1-21, or a pharmaceutically acceptable salt thereof, wherein L is compared to SEQ ID No. 1 1 Comprising a28N substitution.
23. A compound of formula XXI:
His-Gly-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Leu-Ser-Lys-Gln-Met-Glu-Glu-Glu-Ala-Val-Arg-Leu-Phe-Ile-Glu-Trp-Leu-Lys-Asn
-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
formula XXI.
24. A compound according to any one of claims 1 to 23, or a pharmaceutically acceptable salt thereof, wherein, in formula I, when the amino acid at position 16 or 20 is lysine (Lys), C is linked via a direct bond or via a linker 12 -C 24 Chemical modification of the aliphatic diacid conjugated to the epsilon-amino group of the lysine (Lys) side chain, the linker being selected from (AEEA) 2 -(γ-Glu) a 、AEEA-Ahx-(γ-Glu) a 、(Ahx) 2 -(γ-Glu) a And (beta-Ala) 2 -(γ-Glu) a Wherein a is 1 or 2;
preferably via (AEEA) 2 - (gamma-Glu) conjugation of eicosanedioic acid to the epsilon-amino group of lysine (Lys) side chain at position 16 or 20 for chemical modification; or alternatively
Via (Ahx) 2 - (gamma-Glu), AEEA-Ahx- (gamma-Glu) or (. Beta. -Ala) 2 - (gamma-Glu) conjugated eicosanedioic acid to the epsilon-amino group of lysine (Lys) side chain at position 20.
25. A compound according to any one of claims 1 to 24, or a pharmaceutically acceptable salt thereof, wherein in formula I the histidine (His) or tyrosine (Tyr) in position 1 is amidated, e.g. by-C m H 2m+1 -C (O) -is attached to the amino group of histidine (His) or tyrosine (Tyr), and m is an integer from 1 to 20;
preferably, the amino group of tyrosine 1 (Tyr) is bound to CH by 3 C (O) or with C 19 H 39 -C (O) -linked amidation.
26. A compound according to any one of claims 1 to 25, or a pharmaceutically acceptable salt thereof, wherein in formula I the α -carbon atoms of any two amino acids may be linked via a direct bond or via a linker selected from alkyl or alkenyl groups containing 2 to 20 carbon atoms;
Preferably, when the amino acids at position 13 and 16 are both alanine, the alpha-carbon atom of alanine (Ala) at position 13 is attached to the alpha-carbon atom of alanine (Ala) at position 16 via an alkenyl group containing 10 carbon atoms.
27. A compound of formula II:
Tyr 1 -X 2 -Glu 3 -Gly 4 -X 5 -X 6 -Thr 7 -Ser 8 -Asp 9 -X 10 -Ser 11 -X 12 -X 13 -Leu 14 -Asp 15 -X 16 -Ile 17 -X 18 -Gln 19 -Lys 20 -X 21 -Phe 22 -X 23 -X 24 -X 25 -Leu 26 -Ile 27 -Ala 28 -NH-(CH 2 ) 4 -C(O)-Pro 31 -Ser 32 -Ser 33 -Gly 34 -Ala 35 -Pro 36 -Pro 37 -Pro 38 -Ser 39 -NH 2
the compound of the formula II is shown in the specification,
wherein X is 2 、X 5 、X 6 、X 10 、X 12 、X 13 、X 16 、X 18 、X 21 、X 23 、X 24 And X 25 Each independently selected from natural amino acids or non-natural amino acid residues.
28. The compound or pharmaceutically acceptable salt thereof according to claim 27, wherein,
X 2 is an amino acid residue selected from the group consisting of 2-aminoisobutyric acid (Aib) and (. Beta. -Ala),
X 5 is an amino acid residue selected from Thr and Ser,
X 6 is an amino acid residue selected from the group consisting of Phe, homophenylalanine (homoPhe) and para-chlorophenylalanine (Cpa-Ala),
X 10 is an amino acid residue selected from Tyr and pentafluorophenylalanine (Fpa 5-Ala),
X 12 is an amino acid residue selected from Ile and Lys,
X 13 amino acid residues selected from the group consisting of 2-aminoisobutyric acid (Aib), tyr, and Ala,
X 16 is an amino acid residue selected from Lys and Ala,
X 18 is an amino acid residue selected from Ala and Aib,
X 21 is an amino acid residue selected from Ala, glu and 2-aminobutyric acid (Abu),
X 23 is an amino acid residue selected from Val and Leu,
X 24 is an amino acid residue selected from Gln and Asn, and
X 25 is an amino acid residue selected from Trp, 2-methyltryptophan (2-me-Trp) and 1-naphthylalanine (Nal).
29. The compound of claim 27 or 28, or a pharmaceutically acceptable salt thereof, having a structure selected from any one of formulas III to XX below:
Tyr-(Aib)-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Ile-Ala-Leu
-Asp-Ala-Ile-Ala-Gln-Lys-Ala-Phe-Val-Gln-Trp-Leu-Ile-Ala
-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
formula III, tyr- (Aib) -Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Ile- (Aib) -Leu
-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Val-Gln-Trp-Leu-Ile-Ala
-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
The compound of the formula IV,
Tyr-(Aib)-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Ile-Tyr
-(α-meL)-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Val-Gln-Trp-Leu-Ile
-Ala-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
the characteristic of the V-shaped alloy is that,
Tyr-(β-Ala)-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Ile-(Aib)-Leu-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Val-Gln-Trp-Leu-Ile-Ala
-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
a compound of the formula VI,
Tyr-(Aib)-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Ile-(Aib)-Leu
-Asp-Lys-Ile-Ala-Gln-Lys-Glu-Phe-Val-Gln-Trp-Leu-Ile-Ala
-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
the compound of the formula VII,
Tyr-(Aib)-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Lys-(Aib)-Leu
-Asp-Lys-Ile-Ala-Gln-Lys-Glu-Phe-Val-Asn-Trp-Leu-Ile-Ala
-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
formula VIII, tyr- (Aib) -Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Ile- (Aib) -Leu
-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Val-Asn-Trp-Leu-Ile-Ala
-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
Formula IX, tyr- (Aib) -Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Lys- (Aib) -Leu
-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Val-Asn-Trp-Leu-Ile-Ala
-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
Formula X, tyr- (Aib) -Glu-Gly- (D-Thr) - (D-Phe) -Thr-Ser-Asp-Tyr-Ser-Ile- (Aib) -Leu-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Val-Gln-Trp-Leu-Ile-Ala- [ NH- (CH) 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
Formula XI, tyr- (Aib) -Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Ile- (Aib) -Leu
-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Val-Gln-Trp-Leu-Ile-Ala
-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-(D-Ala)-Pro-Pro-Pro-Ser-NH 2
Formula XII, tyr- (Aib) - (D-Glu) -Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Ile- (Aib) -Leu-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Val-Gln-Trp-Leu-Ile
-Ala-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
Formula XIII, tyr- (Aib) -Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Ile-Tyr
-(α-meL)-Asp-Lys-Ile-(Aib)-Gln-Lys-Ala-Phe-Val-Gln-Trp-Leu-Ile-Ala-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
Formula XIV, tyr- (Aib) -Glu-Gly-Thr- (homo-Phe) -Thr-Ser-Asp-Tyr-Ser-Ile- (Aib) -Leu-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Val-Gln-Trp-Leu-Ile-Ala- [ NH- (CH) 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
The composition of the composition is XV,
Tyr-(Aib)-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Ile-(Aib)-Leu
-Asp-Lys-Ile-Ala-Gln-Lys-(Abu)-Phe-Val-Gln-Trp-Leu-Ile-Ala
-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
the first component of the first component is a component of the second component,
Tyr-(Aib)-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Ile-(Aib)-Leu
-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Val-Gln-(2-me-Trp)-Leu-Ile
-Ala-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
the compound of formula XVII is represented by formula,
Tyr-(Aib)-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Ile-(Aib)-Leu
-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Val-Gln-(Nal)-Leu-Ile-Ala
-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
the compound of formula XVIII,
Tyr-(Aib)-Glu-Gly-Ser-Phe-Thr-Ser-Asp-Tyr-Ser-Ile-(Aib)-Leu-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Leu-Gln-Trp-Leu-Ile-Ala
-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
of formula XIX, and
Tyr-(Aib)-Glu-Gly-Thr-(Cpa-Ala)-Thr-Ser-Asp-(Fpa5-Ala)-Ser-Ile-(Aib)-Leu-Asp-Lys-Ile-Ala-Gln-Lys-Ala-Phe-Val-Gln-Trp-Leu-Ile-Ala-[NH-(CH 2 ) 4 -C(O)]-Pro-Ser-Ser-Gly-Ala-Pro-Pro-Pro-Ser-NH 2
formula XX.
30. A compound according to any one of claims 27 to 29, or a pharmaceutically acceptable salt thereof, wherein, in any one of formulae II to XX, at lysine (Lys) at position 16 or 20, C is linked via a direct bond or via a linker 12 -C 24 Chemical modification of the aliphatic diacid conjugated to the epsilon-amino group of the lysine (Lys) side chain, the linker being selected from (AEEA) 2 -(γ-Glu) a 、AEEA-Ahx-(γ-Glu) a 、(Ahx) 2 -(γ-Glu) a And (beta-Ala) 2 -(γ-Glu) a Wherein a is 1 or 2;
preferably, in any of formulas III and V to XX, via (AEEA) 2 - (gamma-Glu) conjugation of eicosanedioic acid to the epsilon-amino group of lysine (Lys) side chain at position 20; or alternatively
In formula IV, via (AEEA) 2 - (gamma-Glu) conjugation of eicosanedioic acid to the epsilon-amino group of lysine (Lys) side chain at position 16 for chemical modification; or alternatively
In formula IV, via (Ahx) 2 - (gamma-Glu), AEEA-Ahx- (gamma-Glu) or (. Beta. -Ala) 2 - (gamma-Glu) conjugated eicosanedioic acid to the epsilon-amino group of lysine (Lys) side chain at position 20.
31. A compound according to any one of claims 27-30, or a pharmaceutically acceptable salt thereof, wherein, in any one of formulae II to XX, histidine (His) or tyrosine (Tyr) in position 1 is amidated, e.g. by-C m H 2m+1 -C (O) -amidation linked to the amino group of histidine (His) or tyrosine (Tyr), and m is an integer from 1 to 30;
preferably, in formula IV, the amino group of tyrosine 1 (Tyr) is bound to CH 3 -C (O) -or with C 19 H 39 -C (O) -linked amidation.
32. A compound according to any one of claims 27 to 31, or a pharmaceutically acceptable salt thereof, wherein in any one of formulae II to XX, the α -carbon atoms of any two amino acids can be linked to form a ring via a direct bond or via a linker selected from alkyl or alkenyl groups containing from 2 to 20 carbon atoms;
preferably, in formula III, the alpha-carbon atom of alanine (Ala) at position 13 is linked to the alpha-carbon atom of alanine (Ala) at position 16 via an alkenyl group containing 10 carbon atoms.
33. A pharmaceutical composition comprising:
a compound according to any one of claims 1-32, or a pharmaceutically acceptable salt thereof, and
a pharmaceutically acceptable carrier, diluent or excipient.
34. Use of a pharmaceutical composition according to claim 33 or a compound according to any one of claims 1-32, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment and/or prevention of a metabolic disease or disorder;
in particular, the metabolic diseases or disorders include diabetes and diabetes-related conditions, as well as obesity and obesity-related conditions;
in particular, the diabetes and diabetes-related disorders include insulin resistance, glucose intolerance, elevated fasting blood glucose, prediabetes, type I diabetes, type II diabetes (T2 DM), gestational diabetes hypertension, dyslipidemia, atherosclerosis, arteriosclerosis, coronary heart disease, peripheral arterial disease, and dyslipidemia that actuates atherosclerosis, dyslipidemia, elevated blood pressure, hypertension, pre-thrombotic and pro-inflammatory states, and combinations thereof;
In particular, obesity and obesity-related disorders include obesity-related inflammation, obesity-related cholecystitis, obesity-induced sleep apnea, non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), and combinations thereof.
35. A method of treating and/or preventing a metabolic disease or disorder comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition according to claim 33 or a compound according to any one of claims 1-32, or a pharmaceutically acceptable salt thereof.
36. The method of claim 35, wherein the pharmaceutical composition, the compound, or a pharmaceutically acceptable salt thereof is administered to the individual by subcutaneous injection.
37. The method of claim 35 or 36, wherein the pharmaceutical composition, the compound, or a pharmaceutically acceptable salt thereof is administered to the individual once a week.
CN202211568720.9A 2022-04-29 2022-12-07 GLP-1/GIP receptor co-agonists, pharmaceutical compositions comprising same and uses thereof Pending CN116970063A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210477959 2022-04-29
CN2022104779599 2022-04-29

Publications (1)

Publication Number Publication Date
CN116970063A true CN116970063A (en) 2023-10-31

Family

ID=88482012

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202211568720.9A Pending CN116970063A (en) 2022-04-29 2022-12-07 GLP-1/GIP receptor co-agonists, pharmaceutical compositions comprising same and uses thereof

Country Status (2)

Country Link
CN (1) CN116970063A (en)
WO (1) WO2023207106A1 (en)

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101155828A (en) * 2005-02-11 2008-04-02 安米林药品公司 Gip analog and hybrid polypeptides with selectable properties
CN111825758A (en) * 2019-04-19 2020-10-27 上海翰森生物医药科技有限公司 GLP-1 and GIP co-agonist compounds
CA3163522A1 (en) * 2019-12-04 2021-06-10 The Scripps Research Institute Glp2 receptor agonists and methods of use
BR112022023834A2 (en) * 2020-05-29 2022-12-27 Beijing Tuo Jie Biopharmaceutical Co Ltd DOUBLE-AGONIST COMPOUND FOR GLP-1 AND GIP RECEPTORS AND ITS APPLICATION
CN112409460B (en) * 2020-11-27 2022-02-01 江苏师范大学 GLP-1/glucagon receptor dual agonist and application thereof

Also Published As

Publication number Publication date
WO2023207106A1 (en) 2023-11-02

Similar Documents

Publication Publication Date Title
CN107735100B (en) Glucagon and GLP-1 co-agonist compounds
TWI744579B (en) Incretin analogs and uses thereof
US8097586B2 (en) Modified exedins and uses thereof
CN106928341B (en) Fixed-point mono-substituted pegylated Exendin analogue and preparation method thereof
US20230220033A1 (en) Glp-1 agonist polypeptide compound and salt thereof, synthesis method therefor and use thereof
JP2018500282A (en) GIP agonist compounds and methods
US20120196802A1 (en) Modified exendins and uses thereof
TW201119670A (en) Sugar chain adduct of antigenicity GLP-1 analogue
KR20230106616A (en) Compounds and uses thereof in the treatment of tachykinin receptor mediated disorders
CA3222051A1 (en) Polypeptide derivative having effect of dual targeted activation of glp-1r and gipr, preparation method therefor, and use thereof
CN113429471B (en) Long-acting GLP-1 polypeptide analogue, and preparation method and application thereof
CN108676084B (en) Exenatide modifier and application thereof
KR20240013798A (en) Multiple agents and their uses
JP7450724B2 (en) Incretin analogs and their uses
CN113621045A (en) Polypeptide derivative with double-receptor agonism and application thereof
Niida et al. Antiobesity and emetic effects of a short-length peptide YY analog and its PEGylated and alkylated derivatives
CN116970063A (en) GLP-1/GIP receptor co-agonists, pharmaceutical compositions comprising same and uses thereof
WO2023207107A1 (en) Glp-1/gcg receptor co-agonist, pharmaceutical composition comprising same and use thereof
CN115572326A (en) Triplex agonists of GLP-1, GCG and GIP receptors
CN116514952B (en) GLP-1 analogues and application thereof
CN111491658B (en) Incretin analogues and uses thereof
EA043950B1 (en) INCRETIN ANALOGUES AND THEIR APPLICATION
CN117143221A (en) GLP-1R, GIPR and GCGR triple agonist compound
CN115819551A (en) GLP-1/glucagon/gastrin receptor triple agonist with site-specific modification and application thereof
CN115960258A (en) GLP-1/glucagon/Y 2 Receptor triple agonists and uses thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination