CN116970064A - GLP-1/GCG receptor co-agonists, pharmaceutical compositions comprising same and uses thereof - Google Patents

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

Info

Publication number
CN116970064A
CN116970064A CN202211587746.8A CN202211587746A CN116970064A CN 116970064 A CN116970064 A CN 116970064A CN 202211587746 A CN202211587746 A CN 202211587746A CN 116970064 A CN116970064 A CN 116970064A
Authority
CN
China
Prior art keywords
lys
ser
glu
gly
ala
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
CN202211587746.8A
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 CN116970064A publication Critical patent/CN116970064A/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
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Abstract

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

Description

GLP-1/GCG receptor co-agonists, pharmaceutical compositions comprising same and uses thereof
Technical Field
The present disclosure relates to GLP-1/GCG receptor co-agonists, pharmaceutical compositions comprising the same, and uses and methods for the treatment and/or prevention of 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.
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. (see diabetes, 2017,60,1851-1861), and long-term administration can exhibit weight-reducing effects by increasing energy metabolism, but GCG has not been widely used for its inherent glycemic effect.
Oxyntomodulin OXM (oxyntomodulin) can activate GLP-1 receptor and GCG receptor simultaneously, and has effects of reducing weight gain and lowering blood sugar. However, the rapid degradation of dipeptidyl peptidase IV and other peptidases in the endogenous OXM body greatly limits its clinical application.
Drug developers developed a range of GLP-1 receptor agonists and GCG receptor agonists. GLP-1 receptor agonists and GCG receptor agonists exert the same biological effects as native GLP-1 and GCG, and are prevented from being degraded and deactivated, thereby prolonging the duration of action.
However, there remains a need for alternative co-agonists that have co-agonism for the GLP-1 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 -Leu 27 -Ala 28 -X 29 -X 30 -Pro 31 -Ser 32 -Ser 33 -Gly 34 -NH 2 the compound of the formula I,
wherein, the liquid crystal display device comprises a liquid crystal display device,
L 1 peptide analogues of the OXM (1-26) peptide, said L 1 Is a peptide consisting of 26 amino acids, and the L 1 Has at least 69% identity with SEQ ID NO. 1,
X 29 and X 30 Each independently is Gly, or X 29 And X 30 representing-NH- (CH) as a whole 2 ) n -C(O)-,
n is any integer from 2 to 6, and
the compounds have GLP-1 receptor agonist activity and GCG receptor agonist activity.
By peptide chain engineering of the OXM (1-26) peptide, in particular by using glycine residues Gly at both positions 29 and 30 of the peptide chain, or by using-NH- (CH) at positions 29 and 30 as a whole and at that position 2 ) n -C (O) -units (n is an integer from 2 to 6) to replace the common-NH- (CH) in the peptide chain 2 ) -C (O) -units, the resulting compounds of formula I unexpectedly retain high activity at GLP-1 receptor and GCG receptor, providing a hypoglycemic and weight-reducing effect.
In another aspect, the present disclosure provides a compound of formula II:
His 1 -X 2 -X 3 -Gly 4 -Thr 5 -Phe 6 -X 7 -Ser 8 -Asp 9 -Tyr 10 -Ser 11 -X 12 -Tyr 13 -X 14 -Asp 15 -Ser 16 -Lys 17 -Lys 18 -Ala 19 -Lys 20 -X 21 -Phe 22 -Val 23 -Glu 24 -Trp 25 -Leu 26 -Leu 27 -Ala 28 -X 29 -X 30 -Pro 31 -Ser 32 -Ser 33 -Gly 34 -NH 2
The compound of the formula II is shown in the specification,
wherein X is 2 、X 3 、X 7 、X 12 、X 14 And X 21 Each independently selected from natural amino acids or non-natural amino acid residues, and X 29 And X 30 Each independently is Gly, or X 29 And X 30 representing-NH- (CH) as a whole 2 ) 4 -C(O)-。
By peptide chain engineering of the OXM (1-26) peptide, in particular by modification of the OXM at position 29 and in the peptide chainGlycine residue Gly is used at position 30, or by taking positions 29 and 30 as a whole and using-NH- (CH) at that position 2 ) n -C (O) -units (n is an integer from 2 to 6) to replace the common-NH- (CH) in the peptide chain 2 ) -C (O) -units, the resulting compounds of formula II of the present disclosure unexpectedly possess co-agonism at the GLP-1 receptor and the GCG receptor, providing a therapeutic effect for lowering blood glucose and reducing body weight. At the same time, the compounds of formula II have high plasma stability and thus pharmacokinetic profile supporting once a week subcutaneous administration in humans, improving patient compliance.
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-C007 according to the present disclosure.
FIG. 2 is a high performance liquid chromatography analysis of compound NBB-C007 according to the present disclosure.
FIG. 3 is a mass spectrum of compound NBB-C002 according to the present disclosure.
FIG. 4 is a high performance liquid chromatography analysis of compound NBB-C002 according to the present disclosure.
Fig. 5 is a mass spectrum of compound NBB-C003 according to the present disclosure.
Fig. 6 is a high performance liquid chromatography diagram of compound NBB-C003 according to the present disclosure.
Fig. 7 is a mass spectrum of compound NBB-C004 according to the present disclosure.
Fig. 8 is a high performance liquid chromatography profile of compound NBB-C004 according to the present disclosure.
Fig. 9 is a mass spectrum of compound NBB-C006 according to the present disclosure.
Fig. 10 is a high performance liquid chromatography diagram of compound NBB-C006 according to the present disclosure.
FIG. 11 is a mass spectrum of compound NBB-C008 according to the present disclosure.
Fig. 12 is a high performance liquid chromatography analysis of compound NBB-C008 according to the present disclosure.
FIG. 13 is a mass spectrum of compound NBB-C009 according to the present disclosure.
Fig. 14 is a high performance liquid chromatography analysis of compound NBB-C009 according to the present disclosure.
Fig. 15 is a graph of potency-concentration change of compound NBB-C002 on a target according to the present disclosure.
Fig. 16 is a graph of potency-concentration change of compound NBB-C003 against a target according to the present disclosure.
Fig. 17 is a graph of potency-concentration change of compound NBB-C004 against target according to the present disclosure.
Fig. 18 is a graph of potency-concentration change of compound NBB-C006 on target according to the present disclosure.
Fig. 19 is a graph of potency-concentration change of compound NBB-C007 on a target according to the present disclosure.
Fig. 20 is a graph of potency-concentration change of compound NBB-C008 against a target according to the present disclosure.
Fig. 21 is a graph of potency-concentration change of compound NBB-C009 acting on a target according to the present disclosure.
FIG. 22 is a graph of blood glucose versus time for the db/db mice tested.
FIG. 23 is a graph of percent blood glucose reduction versus time for the db/db mice tested.
FIG. 24 is a graph of body weight versus time for db/db mice tested.
FIG. 25 is a graph showing the blood glucose versus time in normal mice tested.
Fig. 26 (a), 26 (b) and 26 (C) are concentration-time curves in plasma of compound NBB-C007 according to the present invention subcutaneously injected in SD rats tested, respectively.
Fig. 27 (a) is a concentration-time profile of cord Ma Lutai as a control in plasma injected subcutaneously in SD rats.
Fig. 27 (b) and 27 (c) are concentration-time curves of the control telipopeptide Tirzepatide in plasma subcutaneously injected in SD rats.
Figure 28 is a weight-time profile of the DIO mice tested.
Fig. 29 is a graph of blood glucose versus time for the DIO mice tested.
Fig. 30 is a graph of blood glucose versus time for the DIO mice tested.
FIG. 31 is a bar graph of serum insulin content change in the DIO mice tested.
FIG. 32 is a bar graph of serum biochemical marker changes (UREA, TG, CHO, HDL, LDL, CREA) in the DIO mice tested.
FIG. 33 is a bar graph of changes in serum biochemical indicators (ALT, AST, ALB, TBIL) of the DIO mice tested.
Figure 34 bar graph of body fat rate change for the DIO mice tested.
Fig. 35 is a bar graph of triglyceride content change in the tested DIO mice.
Fig. 36 is a weight-time profile of a subject DIO mouse over a prolonged period of time.
Figure 37 is a graph of percent weight loss versus time for the different doses of DIO mice tested.
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 the targets of 20 natural amino acids (also known as protein amino acids or encoded amino acids) that make up a proteinQuasi-single letter or three-letter codes. 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. Lys represents lysine, and has the structural formula of NH 2 (CH 2 ) 4 CH(NH 2 ) COOH, in the polypeptide backbone, lys is linked to other amino acid residues as-NH-CH-C (O) -NH 2 (CH 2 ) 4 - (i.e., epsilon-amino) attached as a pendant group to an alpha carbon atom, the alpha-amino group being located in the polypeptide backbone. epsilon-Lys also represents lysine, and the structural formula is NH 2 (CH 2 ) 4 CH(NH 2 ) COOH, except that epsilon-Lys was replaced by-NH- (CH) 2 ) 4 -CH-C- (O) -linked to other amino acid residues, epsilon-amino groups in the polypeptide backbone and alpha-amino groups in the side chains.
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.
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 histidine (His) at position 1, the C-terminal amino acid is glycine (Gly) at position 34, and lysine (Lys) at position 20.
In comparison with formulae III to V and VII to IX, in the compounds of formula VI, use is made of-CH 2 -CH 2 -unit replacement of two adjacent glycerins at positions 29 and 30acid-NH-CH 2 -C(O)-NH-CH 2 -C (O) -contained peptide bond. although-NH- (CH) thus obtained 2 ) 4 the-C (O) -unit is no longer 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, NH- (CH) is still formally provided 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 an individual, such as, for example, a decrease in blood glucose and/or a decrease in weight or fat) in such individual being diagnosed or treated after single or multiple doses are administered to an 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 of mammal, its size, age and general health, 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 of concomitant medication, and other relevant conditions.
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 having 12 to 24 carbon atomsLinear or branched dicarboxylic acids. 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, tirz's peptide (TZP) is a GLP-1/GIP receptor co-agonist having the structure shown below.
[ GLP-1 receptor and GCG receptor Co-agonist ]
The present disclosure provides a compound of formula I:
L 1 -Leu 27 -Ala 28 -X 29 -X 30 -Pro 31 -Ser 32 -Ser 33 -Gly 34 -NH 2 the compound of the formula I,
wherein, the liquid crystal display device comprises a liquid crystal display device,
L 1 peptide analogues of the OXM (1-26) peptide, said L 1 Is a peptide consisting of 26 amino acids, and the L 1 Has at least 69% identity with SEQ ID NO. 1,
X 29 and X 30 Each independently is Gly, or X 29 And X 30 representing-NH- (CH) as a whole 2 ) n -C(O)-,
n is any integer from 2 to 6, and
the compounds have GLP-1 receptor agonist activity and GCG receptor agonist activity.
SEQ ID NO. 1 is an OXM (1-26) sequence: HSQGTFTSDYSKYLDSRRAQDFVQWL it is His-Ser-Gln-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Lys-Tyr-Leu-Asp-Ser-Arg-Arg-Ala-Gln-Asp-Phe-Val-Gln-Trp-Leu.
“L 1 An amino acid sequence having at least 69% identity to SEQ ID NO. 1 means L 1 In comparison with SEQ ID NO. 1, of the 26 amino acids at positions 1 to 26, there are identical amino acids at a total of at least 18 positions, for example at a total of 18, 19, 20, 21, 22, 23, 24, 25 or 26 positions.
In some examples, L 1 Has the same amino acid sequence at 18, 19 or 20 positions in total as compared with SEQ ID NO. 1.
In some examples, L 1 Has at least 69% identity, for example 69%, 73%, 77%, 81%, 85%, 88%, 92%, 96% or 100% identity, to the amino acid sequence of SEQ ID NO. 1.
In some examples, L 1 Amino acids of (2)The sequence has 69%, 73% or 77% identity with SEQ ID NO. 1.
In some examples, n is 2, 3, 4, 5, or 6, preferably 4.
According to the present disclosure, by peptide chain engineering of wild type OXM, in particular by employing glycine residues Gly at both positions 29 and 30 of the peptide chain, or by integrating positions 29 and 30 and employing-NH- (CH) at that position 2 ) n -C (O) -units (n is an integer from 2 to 6) to replace the common-NH- (CH) in the peptide chain 2 ) -C (O) -units, the resulting compounds of formula I can surprisingly retain high activity at GLP-1 receptor and GCG receptor, providing a hypoglycemic and weight-reducing effect.
In some examples, L is compared to SEQ ID NO:1 1 Comprising a substitution selected from S2 (Aib) and S2 (beta-Ala).
In some examples, L is compared to SEQ ID NO:1 1 Comprising Q3E substitutions.
In some examples, L is compared to SEQ ID NO:1 1 Comprising a T7K substitution.
In some examples, L is compared to SEQ ID NO:1 1 Comprising a K12 (ε -K) substitution.
In some examples, L is compared to SEQ ID NO:1 1 Comprising an R17K substitution.
In some examples, L is compared to SEQ ID NO:1 1 Comprising R18K substitutions.
In some examples, L is compared to SEQ ID NO:1 1 Comprising a Q20K substitution.
In some examples, L is compared to SEQ ID NO:1 1 Comprising a substitution selected from D21E and D20A.
In some examples, L is compared to SEQ ID NO:1 1 Comprising Q24E substitutions.
In some examples, in formula I, when the amino acid at position 20 is lysine (Lys), C is bonded 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 to 2, thereby imparting excellent in vivo and in vitro activity to the compound;
preferably, the joint is (AEEA) 2 - (gamma-Glu), and said C 12 -C 24 The aliphatic diacid is octadecanedioic acid or eicosanedioic acid.
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 acid at position 21 is alanine (Ala), the α -carbon atom of alanine (Ala) at position 21 is linked to the α -carbon atom of alanine (Ala) at position 28 via an alkenyl group containing 16 carbon atoms.
In some examples, in formula I, when aspartic acid (Asp) or glutamic acid (Glu) containing a side chain carboxyl group, and lysine (Lys), arginine (Arg) or histidine (His) containing a side chain amino group are present together, 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;
Preferably, when the amino acid at position 3 is glutamic acid (Glu) and the amino acid at position 7 is lysine (Lys), the gamma-carboxyl group of glutamic acid (Glu) at position 3 and the epsilon-amino group of lysine (Lys) at position 7 form a ring by forming an amide bond.
According to the present disclosure, there is also provided a compound of formula II:
His 1 -X 2 -X 3 -Gly 4 -Thr 5 -Phe 6 -X 7 -Ser 8 -Asp 9 -Tyr 10 -Ser 11 -X 12 -Tyr 13 -X 14 -Asp 15 -Ser 16 -Lys 17 -Lys 18 -Ala 19 -Lys 20 -X 21 -Phe 22 -Val 23 -Glu 24 -Trp 25 -Leu 26 -Leu 27 -Ala 28 -X 29 -X 30 -Pro 31 -Ser 32 -Ser 33 -Gly 34 -NH 2
the compound of the formula II is shown in the specification,
wherein X is 2 、X 3 、X 7 、X 12 、X 14 And X 21 Each independently selected from natural amino acids or non-natural amino acid residues, and X 29 And X 30 Each independently is Gly, or X 29 And X 30 representing-NH- (CH) as a whole 2 ) 4 -C(O)-。
By peptide chain engineering of the OXM (1-26) peptide, in particular by using glycine residues Gly at both positions 29 and 30 of the peptide chain, or by using-NH- (CH) at positions 29 and 30 as a whole and at that position 2 ) n -C (O) -units (n is an integer from 2 to 6) to replace the common-NH- (CH) in the peptide chain 2 ) -C (O) -units, the resulting compounds of formula II of the present disclosure unexpectedly possess co-agonism at the GLP-1 receptor and the GCG receptor, providing a therapeutic effect for lowering blood glucose and reducing body weight. At the same time, the compounds of formula II have high plasma stability and thus pharmacokinetic profile supporting once a week subcutaneous administration in humans, 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 3 is an amino acid residue selected from Gln and Glu,
X 7 is an amino acid residue selected from Thr and Lys,
X 12 is an amino acid residue selected from the group consisting of Lys and (. Epsilon. -Lys),
X 14 is an amino acid residue selected from Leu and (. Alpha. -meL), and
X 21 is an amino acid residue selected from Glu and Ala.
As previously mentioned, the compounds of formula II of the present disclosure unexpectedly possess co-agonism at the GLP-1 receptor and the GCG receptor, providing a therapeutic effect in lowering blood glucose and reducing body weight. At the same time, the compounds of formula II have high plasma stability and thus pharmacokinetic profile supporting once a week subcutaneous administration in humans, improving patient compliance.
In some examples, the compound has a structure selected from any one of the following formulas III to IX:
His-(Aib)-Gln-Gly-(D-Thr)-Phe-(D-Thr)-Ser-Asp-Tyr-Ser-Lys-Tyr-Leu-Asp-Ser-Lys-Lys-Ala-Lys-Glu-Phe-Val-Glu-Trp-Leu-Leu-Ala-GLy-Gly-Pro-Ser-Ser-Gly-NH 2
the compound of the formula III,
His-(Aib)-Gln-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Lys-Tyr-(α-meL)-Asp-Ser-Lys-Lys-Ala-Lys-Glu-Phe-Val-Glu-Trp-Leu-Leu-Ala-GLy-Gly-Pro-Ser-Ser-Gly-NH 2
the compound of the formula IV,
His-(β-Ala)-Gln-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Lys-Tyr-Leu-Asp-Ser-Lys-Lys-Ala-Lys-Glu-Phe-Val-Glu-Trp-Leu-Leu-Ala-GLy-Gly-Pro-Ser-Ser-Gly-NH 2
the characteristic of the V-shaped alloy is that,
His-(Aib)-Gln-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Lys-Tyr-Leu-Asp-Ser-Lys-Lys-Ala-Lys-Glu-Phe-Val-Glu-Trp-Leu-Leu-Ala-NH-(CH 2 ) 4 -C(O)-Pro-Ser-Ser-Gly-NH 2
a compound of the formula VI,
His-(Aib)-Glu-Gly-Thr-Phe-Lys-Ser-Asp-Tyr-Ser-Lys-Tyr-Leu-Asp-Ser-Lys-Lys-Ala-Lys-Glu-Phe-Val-Glu-Trp-Leu-Leu-Ala-GLy-Gly-Pro-Ser-Ser-Gly-NH 2
the compound of the formula VII,
His-(Aib)-Gln-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-
(ε-Lys)-Tyr-Leu-Asp-Ser-Lys-Lys-Ala-Lys-Glu-Phe-Val-Glu-Trp-Leu-Leu-Ala-GLy-Gly-Pro-Ser-Ser-Gly-NH 2
formula VIII, and
His-(Aib)-Gln-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Lys-Tyr-Leu-Asp-Ser-Lys-Lys-Ala-Lys-Ala-Phe-Val-Glu-Trp-Leu-Leu-Ala-GLy-Gly-Pro-Ser-Ser-Gly-NH 2
formula IX.
In some examples, in any of formulas I-IX, each amino acid residue is in either the L-configuration or the D-configuration independently of the other.
In some examples, in any of formulas I-IX, each amino acid residue is independently of the other in the L-configuration.
In some examples, in any of formulas I to IX, 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, or 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 5 and 7 are both L-Thr or both D-Thr.
In some examples, in any of formulas I-IX, the carbon atoms of each amino acid residue may 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, decenyl or hexadecenyl; substituted or unsubstituted phenyl, such as p-chlorophenyl, pentafluorophenyl; and/or substituted or unsubstituted fused ring aryl, such as 1-naphthyl. 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.
The term "C", as used herein 12 -C 24 The conjugation of an aliphatic diacid to an "amino acid" refers to any natural or unnatural amino acid having the ability to be conjugated to the aliphatic diacid by covalent bonding, or preferably by linker meansIs a functional group of (a). 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 one embodiment, 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 one embodiment, the conjugation is acylation.
In one embodiment, 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 20.
In one embodiment, the compounds of the invention include an aliphatic diacid moiety conjugated directly to a natural or unnatural amino acid having functional groups available for conjugation, without a linker.
In some examples, wherein, in any of formulas II to IX, at lysine (Lys) at position 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 to 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 the present invention, when referring to a linker, 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, the joint is (AEEA) 2 - (gamma-Glu), and said C 12 -C 24 The aliphatic diacid is octadecanedioic acid or eicosanedioic acid.
In some examples, in any of formulas II through IX, via (AEEA) 2 - (gamma-Glu) chemical modification of octadecanedioic acid or eicosanedioic acid conjugated to the epsilon-amino group of lysine (Lys) side chain at position 20.
By way of example of the compound NBB-C007 (corresponding to formula VI) shown below, 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-IX helps to provide co-agonism of GLP-1 and GCP receptors to the compounds and provides the potential to produce long-acting compounds.
In some examples, in any of formulas I-IX, 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. 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 16 carbon atoms. In some examples, in formula IX, the α -carbon atom of alanine (Ala) at position 21 is attached to the α -carbon atom of alanine (Ala) at position 28 via an alkenyl group containing 16 carbon atoms.
In particular, in the compounds of the formula VI, the radicals-NH- (CH) is contained in positions 29 and 30 2 ) 4 -C (O) -units comprising two adjacent glycine residues-NH-CH, but not as in other compounds herein 2 -C(O)-NH-CH 2 -C (O) -, while unexpectedly maintaining co-agonism at the GLP-1 receptor and the GCG receptor, provides a therapeutic effect for lowering blood glucose and weight. At the same time, the compound of formula VI 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, in any of formulas II-IX, 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. In some examples, in formula VII, the gamma-carboxyl group of glutamic acid (Glu) at position 3 and the epsilon-amino group of lysine (Lys) at position 7 form a ring by amide linkage.
In some examples, in any of formulas I-IX, the histidine (His) at position 1 (i.e., the N-terminus) is amidated, e.g., by-C m H 2m+1 -C (O) -is amidated attached to the amino group of the histidine (His), 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.
/>
/>
[ preparation method of GLP-1/GCG 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 groups of tryptophan (Trp) and the side chain amino groups of lysine (Lys) are protected by Boc (tert-butoxycarbonyl protecting group); the side chain hydroxyl group of serine (Ser), 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, a carboxylic group having an Fmoc-protected (from the rightmost C-terminus) amino acid at position 1 was condensed as an amide bond onto a swelled and deprotected Rink-amide-AMresin using a condensing agent of 6-Chlorobenzotriazole-1, 3-tetramethyluronium hexafluorophosphate (O- (1H-6-Chlorobenzotriazole-1-yl) -1, 3-tetramethyluronium hexafluorophosphate, HCTU), and then the Fmoc-protected group on the amino group was deaminated with a 20% piperidine-containing N, N-Dimethylformamide (DMF) solution, followed by washing.
Then, in a similar manner, the condensing agent HCTU was employed, and the Fmoc-protected (from the rightmost C-terminus) amino acids at positions 2 through 4, respectively, were employed; 2 Fmoc-protected glycine (for other compounds of NBB-C series than compound NBB-C007), or Fmoc-protected 5-aminopentanoic acid (Fmoc-NH- (CH) 2 ) 4 -C (O) OH) (for compound NBB-C007); and Fmoc protected (from the rightmost C-terminus) amino acids 7 through 34; the previous cycles of amide bond formation coupling reaction, deprotection to remove Fmoc protecting group and washing were repeated sequentially.
iii) Deprotection and chemical modification of lysine 15 (corresponding to lysine 20 from leftmost N-terminus)
Palladium tetraphenylphosphine Pd (PPh) 3 ) 4 Removing (from the rightmost C-terminal) the Boc protecting group on the epsilon-amino group of lysine 15, and cleaning.
Then, the epsilon amino group of lysine 15 (from the rightmost C-terminus) was coupled and deprotected using Fmoc-protected linker Fmoc-AEEA. In a similar manner, such coupling and deprotection operations are repeated 1 time, thereby ligating the 2 nd linker AEEA.
Then, fmoc-and OtBu-protected glutamic acid (Fmoc-Glu (OtBu) -OH) and condensing agent (HCTU) were used to couple with the amino group of linker 2 AEEA and deprotect, thereby ligating the 3 rd linker gamma-Glu.
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 is cleaved from the resin carrier by reacting the cleavage reagent with Rink-Amide-AM resin using a trifluoroacetic acid (TFA, trifluoroacetic acid)/water/phenol/Triisopropylsilane (Tips, triisoopropylsilane) 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, e.g., P.Stahl et al, handbook of Pharmaceutical Salts: properties, selections, second revision (Wiley-VCH, 2011); S.M. Berge et al, "pharmaceutical salts", journal of PharmaceuticalSciences, 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, lipkincott, 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), and 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 piperidine was measured with a measuring cylinder and DMF was added to the measuring 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-C007 ]
(1) Swelling of resin
a) 0.63g (0.2 mmol) of 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) The solvent was drained after washing with 10mL of DMF.
(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 "(2) resin deprotection".
Then, the formation of the polypeptide is performed by: starting from the 1 st amino acid at the rightmost C-terminal of the polypeptide chain, the amino acids are connected one by one in the direction of the leftmost N-terminal to form the polypeptide.
(3) The 1 st amino acid (from the rightmost C-terminal)
a) 237mg (0.8 mmol) of Fmoc-Gly-OH (glycine) and 314mg (0.8 mmol) of condensing agent HCTU were weighed into a 10mL EP tube, 6mL of the mixture was dissolved in the EP tube and shaken well, 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 tube was removed by shaking in a shaking table at a constant temperature of 33℃for 10 minutes.
(4) From amino acid 2 to amino acid 4 (from the rightmost C-terminus)
The 2 nd to 4 th amino acids were sequentially accessed similarly to the 1 st amino acid from the rightmost C-terminal in the step (3), except that Fmoc-Ser (tBu) -OH (serine), and Fmoc-Pro-OH (proline) were used as starting materials for coupling the 2 nd to 4 th amino acids, respectively.
(5) "5 th and 6 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 tube was removed by shaking in a shaking table at a constant temperature of 33℃for 10 minutes.
(6) From the 7 th to 34 th amino acids (from the rightmost C-terminus)
Similar to the 1 st amino acid (from the rightmost C-terminal) in step (3), the 7 th to 34 th amino acids are sequentially accessed, the difference is only that Fmoc-Ala-OH (alanine), fmoc-Leu-OH (leucine), fmoc-Trp (Boc) -OH (tryptophan), fmoc-Glu (OtBu) -OH (glutamic acid), fmoc-Val-OH (valine), fmoc-Phe-OH (phenylalanine), fmoc-Glu (OtBu) -OH (glutamic acid), fmoc-Lys (Boc) -OH (lysine), fmoc-Ala-OH (alanine), fmoc-Lys (Boc) -OH (lysine), fmoc-Ser (tBu) -OH (serine), fmoc-Asp (OtBu) -OH (aspartic acid), fmoc-Leu-OH (leucine), fmoc-Tyr (tBu) -OH (tyrosine), fmoc-Lys (Boc) -OH (lysine), fmoc-Ser (tBu) -OH (serine), fmoc-Tyr (OH (tBu) -aspartic acid), fmoc-Ser (tBu) -OH (serine), fmoc-Thr (tBu) -OH (threonine), fmoc-Phe-OH (phenylalanine), fmoc-Thr (tBu) -OH (threonine), fmoc-Gly-OH (glycine), fmoc-Gln (Trt) -OH (glutamine), fmoc- (Aib) -OH (2-aminoisobutyric acid) and Fmoc-His (Trt) -OH (histidine) were used as starting materials for coupling (from the rightmost C-terminus) the 7 th to 34 th amino acid.
(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 LDCM and 10mL of LDMF, 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. 124. Mu.L was added to the EP tube
(1 mmol) phenylsilane, shaking thoroughly, and transferring the solution into a polypeptide synthesis tube. 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 was dissolved in DMF and 200. Mu.L (1.2 mmol) of DIEA was added thereto, and after mixing uniformly, it was transferred to a polypeptide 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 was dissolved in DMF and 200. Mu.L (1.2 mmol) of DIEA was added thereto, and after mixing uniformly, it was transferred to a polypeptide 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'.
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/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) 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 removing supernatant to obtain the crude peptide.
d) Air-drying at room temperature, and mashing to obtain purified polypeptide.
Purified polypeptides were isolated using Shimadzu semi-preparative liquid chromatography, and FIG. 1 is a representation of ESI-MS (electrospray mass spectrometry) performed on purified polypeptides NBB-C007, with peaks representing molecular weights of different mass to charge ratios. FIG. 2 is a high performance liquid chromatography analysis graph showing that NBB-C007 was synthesized with 95% purity.
[ Synthesis of other Compound of NBB-C series ]
Other compounds of the NBB-C series were prepared in a similar manner to the preparation of compound NBB-C007, 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 respective structural formulas.
The cyclization of NBB-C006 is as follows:
Fmoc-S8-OH (Cas No. 288617-75-4) and Fmoc-R8-OH (Cas No. 945212-26-0) were used as amino acid starting materials at positions 7 and 14 (from the rightmost C-terminus), respectively, 64mg of Grubbs catalyst (CasNo. 172222-30-9) was weighed into a 10mL EP tube and dissolved with 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 R8 unit and the S8 unit undergo olefin metathesis reaction, and cyclize. After this step, the peptide chain is extended, and the 15 th to 34 th amino acids are sequentially accessed (from the rightmost C-terminal).
The cyclization of NBB-C008 was performed as follows:
extension of peptide chain: fmoc-Lys (Dde) -OH, fmoc-Lys (Alloc) -OH and Fmoc-Glu (OAll) -OH were used as amino acid starting materials at positions 14, 28 and 32 (from the rightmost C-terminus), respectively. After the peptide chain extension is completed, the resin is washed.
Gamma-carboxydehydrative ring closure of epsilon-amino group of lysine 7 and glutamic acid 3 (from leftmost N-piece): 232mg (0.2 mmol) of palladium tetraphenylphosphine are weighed into a 10mL EP tube, 4mL of DCM and 4mL of DMF are added to the EP tube for dissolution, and 248. Mu.L (0.2 mmol) of phenylsilane is added to the EP tube after thoroughly mixing. The solution was transferred to a polypeptide synthesis tube. Transferring the polypeptide synthesis tube to a 33 ℃ constant temperature shaking table for shaking for 3 hours, and taking out and cleaning. This step removes the side chain protecting groups at positions 7 and 3 (from the leftmost N-piece) and exposes the epsilon-amino group of lysine at position 7 (from the leftmost N-piece) and the gamma-carboxyl group of glutamic acid at position 3. To a 10mL EP tube, 78mg (1.52 mmol) of HOBt was weighed, 6mL of DMF was added for dissolution, and 250. Mu.L (1.52 mmol) of DIC was added and shaken well. The above mixed solution is added into a polypeptide synthesis tube after being evenly mixed, and is transferred to a shaking table at a constant temperature of 33 ℃ for 12 hours and then taken out, thereby dehydrating the epsilon-amino group of the lysine at the 7 th position (from the leftmost N section) and the gamma-carboxyl group of the glutamic acid at the 3 rd position to form an amide group for cyclization.
Dde at position 20 Lys (Dde) was removed (from the leftmost N-terminus): 0.3mL of hydrazine hydrate was pipetted into a 10mL EP tube and 8mL of DMF was added and shaken well. And (3) uniformly mixing the mixed solution, transferring the mixed solution into a polypeptide synthesis tube, transferring the polypeptide synthesis tube to a 33 ℃ constant-temperature shaking table for oscillating reaction for 10min, taking out the polypeptide synthesis tube, and pumping the solvent. Repeating the steps for one time to completely remove the Dde protecting group.
FIGS. 1 to 14 show ESI-MS (electrospray mass spectrometry) characterization and HPLC analysis of NBB-C series compounds, respectively, and it can be seen that the purity of these compounds is not less than 95%.
[ in vitro Activity assay: cAMP detection ]
Experimental reagent
Experimental equipment
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 using DMEM containing 10% FBS, 1% P/S, 500. Mu.M IBMX, 20. Mu.L was removed for cell counting and diluted to 2X 10 6 cells/mL, 5. Mu.L of cells were plated in 384-well plates, and 5. Mu.L was addedlDMSO dissolved and diluted with a ten-fold gradient of DMEM containing 500. Mu. MIBMX (2X 10) -6 、2×10 -7 、···、2×10 -15 M) of the NBB-C series compound prepared previously, or alternatively of comparative cable Ma Lutai (homemade), were incubated at 37℃for 30 minutes, and after addition of 5. Mu.L of each of cAMP-d2 and anti-cAMP in cAMP-GsDynamickit and incubation at room temperature for 1h, were read with a TECAN microplate reader, excitation at 340nm and emission at 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 GraphPadprism 8 by using a four-parameter equation to obtain EC 50 Values, see fig. 15-21 and tables below.
As can be seen from the above table, compounds NBB-C003, NBB-C004, NBB-C006 and NBB-C007 exhibited dual activity against human GLP-1 receptor and GCG receptor. In particular, for the compound NBB-C007, NH- (CH) is contained in the peptide chain as compared with other NBB-C series compounds 2 ) 4 -C (O) -units other than two adjacent glycine-NH-CH 2 -C(O)-NH-CH 2 -C (O) -units, which surprisingly still maintain excellent dual activity towards the human GLP-1 receptor and the GCG receptor.
[ 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, so each group had similar initial average body weight and blood glucose concentration. Excipient controls ("solvent set", 0.05% NaHCO) 3 Solution), dissolved in excipient (0.05% NaHCO) 3 Solution) of the cord Ma Lutai control (dose 50 nmol/kg) and the NBB-C007 compound prepared in the previous example (dose 50 nmol/kg) were taken freely by subcutaneous injectionThe fed db/db mice were dosed and blood was collected from the tail vein using a steady hao rapid blood glucose meter (OneTouch UltraEasy, predatory) and blood glucose values were measured for 72 consecutive hours. Meanwhile, the body weight of db/db mice was monitored at 0h, 24h and 72 h. The test results are shown in fig. 22 to 24 and the following table.
It can be seen that the NBB-C007 compound according to the present disclosure has significant hypoglycemic and weight-reducing effects comparable to or superior to that of cable Ma Lutai and can maintain the pharmacodynamic effects for 72 hours, indicating that the NBB-C007 compound can achieve clinical treatment of type II diabetes and weight reduction and achieve the possibility of once-a-week dosing.
[ 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), telogen (dose 30 nmol/kg), telogen Tirzepatide control (dose 30 nmol/kg), and NBB-C007 compound prepared in the previous example (dose 30 nmol/kg) were administered by subcutaneous injection to freely feeding db/db mice twice weekly for a 12 week dosing period. During the administration period, the body weight, food intake and blood glucose of the mice were monitored, and the glycated hemoglobin HbA1C was measured by taking blood at the 4 th week, 6 th week, 8 th week and 10 th week, respectively. The test results of the inhibition (%) of glycosylated hemoglobin are shown in the following table.
It can be seen that compound NBB-C007 according to the present disclosure had a significant lowering effect on glycosylated hemoglobin of type II diabetic mice after long-term administration, suggesting that blood glucose of db/db mice may 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, the next day the mice were weighed and assayed for 0min blood glucose using a steady blood rapid glucometer (onetouch easy, prednisone).
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-C007 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). The test results are shown in the following table and in fig. 25.
From the above glucose tolerance experiments, it can be seen that the NBB-C007 compounds according to the present disclosure had comparable hypoglycemic effects to that of cable Ma Lutai, and that the glucose AUC was reduced in both the first and second day of testing, indicating that the NBB-C007 compounds according to the present disclosure had good efficacy in postprandial glycemic control.
[ 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), as well as NBB-C007 compound (dose 1 mg/kg), cord Ma Lutai (dose 1 mg/kg) and telipopeptide tirzepair control (dose 0.5 mg/kg) prepared in the previous examples dissolved in excipients, 0.25h, 0.5h, 1h, 2h, 4h, 8h, 24h, 48h, 72h, 96h, 108h, 120h, 144h, 168h, 204h, 240h, 300h and/or 324h, respectively, blood venous collected in the jugular vein was placed in EDTA2K anticoagulant tube, plasma was collected by centrifugation at 8000rpm for 5 minutes, plasma concentration of the test substance was determined by LC-MS/MS method, and the plasma sample was extracted using methanol, the sample treatment steps were as follows:
30.0. Mu.L of the sample, 50.0. Mu.L of the internal standard solution (Soxhlet Ma Lutai, 20,000 ng/mL) and 200. Mu.L of methanol were vortexed for 10min, centrifuged for 10min (3,900 rpm), and the supernatant was taken into another clean 96-well plate for LC-MS/MS analysis. The test results are shown in the following table and fig. 26 (a) to 27 (c).
T 1/2 =half-life, T max Time to maximum concentration, C max The concentration of the plasma at the maximum value,
AUC last AUC of this period from the start of dosing to the last point.
It can be seen that compound NBB-C007 reached an average maximum plasma concentration about 24 hours after subcutaneous administration. Of these, compound NBB-C007 had a half-life of 11.88 hours in SD rats and had good metabolic stability 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-C007 (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, place in an EDTA-K2 test tube, temporarily store in an ice water bath after whole blood collection, centrifuge for 5min at 11000rpm within 30 min, separate plasma, and place in a separated plasma refrigerator for cold test. The plasma prototype concentration was measured by LC-MS/MS. 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 1/2 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 compound NBB-C007 had a half-life of 79.19 hours in SD rats, had good metabolic stability, supporting the possibility of once weekly dosing.
[ 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 normal feedsMice served as the 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 50nmol/kg, NBB-C007 group (administration dose 50 nmol/kg)), and the mice were subcutaneously administered with the above test substance twice weekly, with a administration volume of 5mL/kg, and a vehicle control (0.05% NaHCO) was administered to the normal model group and the 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. 28.
As can be seen from fig. 28, after 40 days, the body weight of both normal model group and model control group ("solvent group") mice increased; the weight of mice administered with cable Ma Lutai was almost unchanged, and the weight of the mice tested with the NBB-C007 compound prepared in the previous example was reduced by about 40%.
2. 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. 29.
As can be seen from fig. 29, the test mice administered the NBB-C007 compound prepared in the previous example had better insulin sensitivity than the mice administered the cable Ma Lutai.
3. 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. 30.
As can be seen from fig. 30, the test mice administered the NBB-C007 compound prepared in the previous example had better postprandial glycemic control than the mice administered the cable Ma Lutai.
4. 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 (totalbilirubin), glutamic pyruvic transaminase ALT, glutamic oxaloacetic transaminase AST, test results see fig. 31 to 33.
As can be seen from fig. 31 to 33, the model control group exhibited a typical diabetes index of high serum insulin content. The test mice administered the NBB-C007 compound prepared in the previous examples had a comparable or better trend of reduced serum insulin content and comparable or better function of protecting liver function, protecting kidney function and improving lipid metabolism compared to mice administered rope Ma Lutai.
5. 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. 34 and 35.
As can be seen from fig. 34 and 35, the test mice administered the NBB-C007 compound 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.
[ Long-term administration weight loss effect of obese DIO mice ]
C57BL/6 mice (Nanjing Ji kang), male and high fat feed (60%Kcal fat,D12492) are 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 30% higher than that of normal diet animals, and the mice are randomly grouped after random blood sugar and weight are detected, and 6 mice are in each group. 6 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 (30 nmol/kg administered), a NBB-C007 group (30 nmol/kg administered) and a Tirzepatide group (30 nmol/kg administered). DIO small Mice were given subcutaneously twice weekly with 5mL/kg of dosing volume and vehicle controls (0.05% NaHCO) were given to the normal model group and model control group ("solvent group") 3 Solution) for 50 days, during which time the body weight, food intake, etc. were monitored. The results of the body weight are shown in fig. 36.
As can be seen from fig. 36, the weight loss of obese DIO mice after administration of the NBB-C007 compound prepared in the previous example was significantly better than that of control administration of cord Ma Lutai, tirzepatide and solvent during the long-term administration period of 50 days, and surprisingly, the weight of obese DIO mice was even reduced to the weight level of normal mice.
[ screening of dose administered to obese DIO mice ]
C57BL/6 mice (Nanjing Ji kang), male and high fat feed (60%Kcal fat,D12492) are 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 30% higher than that of normal diet animals, and the mice are randomly grouped after random blood sugar and weight are detected, and 5 mice are in each group. 5 mice were fed normal feed as normal model group. The mice fed with the high fat diet were randomly divided into model control group ("solvent group"), NBB-C007 group (5 nmol/kg, 10nmol/kg, 15nmol/kg of administration dose). DIO 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 28 days, during which time the body weight, food intake, etc. were monitored. The results of the body weight are shown in fig. 37 and the following table.
As can be seen from FIG. 37 and the above table, the weight loss effect of the obese DIO mice became more and more pronounced as the dosing amount increased from 5nmol/kg to 15 nmol/kg.

Claims (24)

1. A compound of formula I:
L 1 -Leu 27 -Ala 28 -X 29 -X 30 -Pro 31 -Ser 32 -Ser 33 -Gly 34 -NH 2 the compound of the formula I,
wherein, the liquid crystal display device comprises a liquid crystal display device,
L 1 peptide analogues of the OXM (1-26) peptide, said L 1 Is a peptide consisting of 26 amino acids, and the L 1 Has at least 69% identity with SEQ ID NO. 1,
X 29 and X 30 Each independently is Gly, or X 29 And X 30 representing-NH- (CH) as a whole 2 ) n -C(O)-,
n is any integer from 2 to 6, and
the compounds have GLP-1 receptor agonist activity and GCG receptor agonist activity.
2. The compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein L is compared to SEQ id No. 1 1 Comprising a substitution selected from S2 (Aib) and S2 (beta-Ala).
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 Q3E substitutions.
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 T7K 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 K12 (ε -K) substitution.
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 R17K substitution.
7. According toThe compound of any one of claims 1-6, or a pharmaceutically acceptable salt thereof, wherein L is compared to SEQ ID No. 1 1 Comprising R18K substitutions.
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 a Q20K 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 D21E and D20A.
10. The compound according to any one of claims 1-9, or a pharmaceutically acceptable salt thereof, wherein L is compared to SEQ ID No. 1 1 Comprising Q24E substitutions.
11. A compound according to any one of claims 1 to 10, or a pharmaceutically acceptable salt thereof, wherein, in formula I, when the amino acid at position 20 is lysine (Lys), 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 to 2;
preferably, the joint is (AEEA) 2 - (gamma-Glu), and said C 12 -C 24 The aliphatic diacid is octadecanedioic acid or eicosanedioic acid.
12. A compound according to any one of claims 1-11, 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 acid at position 21 is alanine (Ala), the α -carbon atom of alanine (Ala) at position 21 is linked to the α -carbon atom of alanine (Ala) at position 28 via an alkenyl group containing 16 carbon atoms.
13. A compound according to any one of claims 1-12, or a pharmaceutically acceptable salt thereof, wherein in formula I, when aspartic acid (Asp) or glutamic acid (Glu) containing a side chain carboxyl group and lysine (Lys), arginine (Arg) or histidine (His) containing a side chain amino group are present together, 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;
Preferably, when the amino acid at position 3 is glutamic acid (Glu) and the amino acid at position 7 is lysine (Lys), the gamma-carboxyl group of glutamic acid (Glu) at position 3 and the epsilon-amino group of lysine (Lys) at position 7 form a ring by forming an amide bond.
14. A compound of formula II:
His 1 -X 2 -X 3 -Gly 4 -Thr 5 -Phe 6 -X 7 -Ser 8 -Asp 9 -Tyr 10 -Ser 11 -X 12 -Tyr 13 -X 14 -Asp 15 -Ser 16 -Lys 17 -Lys 18 -Ala 19 -Lys 20 -X 21 -Phe 22 -Val 23 -Glu 24 -Trp 25 -Leu 26 -Leu 27 -Ala 28 -X 29 -X 30 -Pro 31 -Ser 32 -Ser 33 -Gly 34 -NH 2 the compound of the formula II is shown in the specification,
wherein X is 2 、X 3 、X 7 、X 12 、X 14 And X 21 Each independently selected from natural amino acids or non-natural amino acid residues, and X 29 And X 30 Each independently is Gly, or X 29 And X 30 representing-NH- (CH) as a whole 2 ) 4 -C(O)-。
15. The compound or pharmaceutically acceptable salt thereof according to claim 14, wherein,
X 2 is an amino acid residue selected from the group consisting of 2-aminoisobutyric acid (Aib) and (. Beta. -Ala),
X 3 is an amino acid residue selected from Gln and Glu,
X 7 is an amino acid residue selected from Thr and Lys,
X 12 is an amino acid residue selected from the group consisting of Lys and (. Epsilon. -Lys),
X 14 is an amino acid residue selected from Leu and (. Alpha. -meL), and
X 21 is an amino acid residue selected from Glu and Ala.
16. The compound of claim 14 or 15, or a pharmaceutically acceptable salt thereof, wherein the compound has a structure selected from any one of the following formulas III to IX:
His-(Aib)-Gln-Gly-(D-Thr)-Phe-(D-Thr)-Ser-Asp-Tyr-Ser-Lys-Tyr-Leu-Asp-Ser-Lys-Lys-Ala-Lys-Glu-Phe-Val-Glu-Trp-Leu-Leu-Ala-GLy-Gly-Pro-Ser-Ser-Gly-NH 2
the compound of the formula III,
His-(Aib)-Gln-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Lys-Tyr-(α-meL)-Asp-Ser-Lys-Lys-Ala-Lys-Glu-Phe-Val-Glu-Trp-Leu-Leu-Ala-GLy-Gly-Pro-Ser-Ser-Gly-NH 2
the compound of the formula IV,
His-(β-Ala)-Gln-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Lys-Tyr-Leu-Asp-Ser-Lys-Lys-Ala-Lys-Glu-Phe-Val-Glu-Trp-Leu-Leu-Ala-GLy-Gly-Pro-Ser-Ser-Gly-NH 2
the characteristic of the V-shaped alloy is that,
His-(Aib)-Gln-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Lys
-Tyr-Leu-Asp-Ser-Lys-Lys-Ala-Lys-Glu-Phe-Val-Glu-Trp-Leu
-Leu-Ala-NH-(CH 2 ) 4 -C(O)-Pro-Ser-Ser-Gly-NH 2
a compound of the formula VI,
His-(Aib)-Glu-Gly-Thr-Phe-Lys-Ser-Asp-Tyr-Ser-Lys-Tyr
-Leu-Asp-Ser-Lys-Lys-Ala-Lys-Glu-Phe-Val-Glu-Trp-Leu
-Leu-Ala-GLy-Gly-Pro-Ser-Ser-Gly-NH 2
the compound of the formula VII,
His-(Aib)-Gln-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-(ε-Lys)-Tyr-Leu-Asp-Ser-Lys-Lys-Ala-Lys-Glu-Phe-Val-Glu-Trp-Leu-Leu-Ala-GLy-Gly-Pro-Ser-Ser-Gly-NH 2
formula VIII, and
His-(Aib)-Gln-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Lys
-Tyr-Leu-Asp-Ser-Lys-Lys-Ala-Lys-Ala-Phe-Val-Glu-Trp-Leu
-Leu-Ala-GLy-Gly-Pro-Ser-Ser-Gly-NH 2
formula IX.
17. A compound according to any one of claims 14 to 16, or a pharmaceutically acceptable salt thereof, wherein, in any one of formulae II to IX, at lysine (Lys) at position 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 to 2;
preferably, the joint is (AEEA) 2 - (gamma-Glu), and said C 12 -C 24 The aliphatic diacid is octadecanedioic acid or eicosanedioic acid.
18. A compound according to any one of claims 14-17, or a pharmaceutically acceptable salt thereof, wherein, in any one of formulas II to IX, the a-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, in formula IX, the alpha-carbon atom of alanine (Ala) at position 21 is linked to the alpha-carbon atom of alanine (Ala) at position 28 via an alkenyl group containing 16 carbon atoms.
19. A compound according to any one of claims 14-18, or a pharmaceutically acceptable salt thereof, wherein in any one of formulas II-IX, 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;
preferably, in formula VII, the gamma-carboxyl group of glutamic acid (Glu) at position 3 and the epsilon-amino group of lysine (Lys) at position 7 form a ring by amide bond formation.
20. A pharmaceutical composition comprising:
a compound according to any one of claims 1-19, or a pharmaceutically acceptable salt thereof, and
a pharmaceutically acceptable carrier, diluent or excipient.
21. Use of a pharmaceutical composition according to claim 20 or a compound according to any one of claims 1-19, 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.
22. 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 20 or a compound according to any one of claims 1-19, or a pharmaceutically acceptable salt thereof.
23. The method of claim 22, wherein the pharmaceutical composition, the compound, or a pharmaceutically acceptable salt thereof is administered to the individual by subcutaneous injection.
24. The method of claim 22 or 23, wherein the pharmaceutical composition, the compound, or a pharmaceutically acceptable salt thereof is administered to the individual once a week.
CN202211587746.8A 2022-04-29 2022-12-07 GLP-1/GCG receptor co-agonists, pharmaceutical compositions comprising same and uses thereof Pending CN116970064A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2022104781118 2022-04-29
CN202210478111 2022-04-29

Publications (1)

Publication Number Publication Date
CN116970064A true CN116970064A (en) 2023-10-31

Family

ID=88477268

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202211587746.8A Pending CN116970064A (en) 2022-04-29 2022-12-07 GLP-1/GCG receptor co-agonists, pharmaceutical compositions comprising same and uses thereof

Country Status (2)

Country Link
CN (1) CN116970064A (en)
WO (1) WO2023207107A1 (en)

Family Cites Families (7)

* 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
GB0511986D0 (en) * 2005-06-13 2005-07-20 Imp College Innovations Ltd Novel compounds and their effects on feeding behaviour
WO2016090628A1 (en) * 2014-12-12 2016-06-16 北京韩美药品有限公司 Oxyntomodulin (oxm) analogs, synthesis and use thereof
TWI783244B (en) * 2015-06-22 2022-11-11 美商美國禮來大藥廠 Glucagon and glp-1 co-agonist compounds
CN106986924A (en) * 2017-03-23 2017-07-28 中国药科大学 Oxyntomodulin(OXM)Analog and its application
JP7350851B2 (en) * 2018-11-12 2023-09-26 天津薬物研究院有限公司 Glucagon-derived peptides and their uses
CN114853908A (en) * 2019-05-16 2022-08-05 浙江道尔生物科技有限公司 Fusion protein for treating metabolic diseases

Also Published As

Publication number Publication date
WO2023207107A1 (en) 2023-11-02

Similar Documents

Publication Publication Date Title
KR102544419B1 (en) Glucagon and glp-1 co-agonist compounds
KR102440323B1 (en) Incretin analogues and uses thereof
TWI744579B (en) Incretin analogs and uses thereof
JP2018500282A (en) GIP agonist compounds and methods
NO328077B1 (en) Use of an exendin or exendin agonist for the preparation of a pharmaceutical formulation.
KR20130133103A (en) Analogues of glucose-dependent insulinotropic polypeptide (gip) modified at n-terminal
CN1976948A (en) Modified Exendins and application thereof
KR20200116085A (en) Modified lipidated relaxin B chain peptides and their therapeutic uses
KR20240013798A (en) Multiple agents and their uses
CN113429471B (en) Long-acting GLP-1 polypeptide analogue, and preparation method and application thereof
CN108676084B (en) Exenatide modifier and application thereof
KR20230051583A (en) CRF2 receptor agonists and their use in therapy
JP7450724B2 (en) Incretin analogs and their uses
JP2023525260A (en) Polypeptide derivative having dual receptor agonist activity and use thereof
CN116970064A (en) GLP-1/GCG receptor co-agonists, pharmaceutical compositions comprising same and uses thereof
CN115572326A (en) Triplex agonists of GLP-1, GCG and GIP receptors
CN116970063A (en) GLP-1/GIP receptor co-agonists, pharmaceutical compositions comprising same and uses thereof
CN111491658B (en) Incretin analogues and uses thereof
CN113754752B (en) Polypeptide and preparation method and application thereof
EA043950B1 (en) INCRETIN ANALOGUES AND THEIR APPLICATION
CN117143221A (en) GLP-1R, GIPR and GCGR triple agonist compound
CN115785249A (en) GLP-1 analogue and application thereof
CN115960258A (en) GLP-1/glucagon/Y 2 Receptor triple agonists and uses thereof
CN115073582A (en) Polypeptide compound and application thereof in preventing or treating diabetes or diabetic complications

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