Incretin analogue and preparation method and application thereof

文档序号:93508 发布日期:2021-10-12 浏览:40次 中文

阅读说明:本技术 一种肠促胰岛素类似物及其制备方法和用途 (Incretin analogue and preparation method and application thereof ) 是由 黄岩山 陈永露 段文文 温晓芳 柳媛媛 王渊 盛世美 刘颖 倪晟 朱明月 方晨 于 2020-04-08 设计创作,主要内容包括:本发明提供了一种肠促胰岛素类似物及其制备方法和用途。所述肠促胰岛素类似物兼具GLP-1R/GIPR/GCGR激动活性,为三效激动剂,其在降糖、减脂以及减重方面效果显著。(The invention provides an incretin analogue and a preparation method and application thereof. The incretin analogue has GLP-1R/GIPR/GCGR agonistic activity, is a triple-effect agonist, and has obvious effects on reducing blood sugar, fat and weight.)

1. An incretin analog comprising a glucagon-like polypeptide linked to a long chain fatty acid; wherein, the amino acid sequence of the glucagon-like polypeptide is shown as formula (I):

YSEGTFTSDX10SKYLDSQAAQDFVQWLLAGGPSSGAPPPSX40 (I);

in the formula (I), X10Is amino acid K, X40Selected from the group consisting of OH or NH2

2. The incretin analog of claim 1, wherein the long chain fatty acid is a fatty acid having 14 to 20 carbons; preferably a fatty acid containing 16 to 18 carbon atoms; more preferably the long chain fatty acid is a straight chain saturated monocarboxylic acid; more preferably, the long chain fatty acid is palmitic acid.

3. The incretin analog of claim 1, wherein the long chain fatty acid is linked to amino acid K of the peptide chain of the glucagon-like polypeptide; preferably, is connected to X10To the amino acid of (a); preferably, the linkage is cross-linked; and/or

The glucagon-like polypeptide is linked to the long chain fatty acid through a linker; preferably, the linker is a linker capable of reacting with amino acid K and with the reactive group of a long chain fatty acid; more preferably, the linker is a linker comprising at least 1 unit of- γ -glutamyl-; more preferably, the incretin analogue has the structure shown in the formula (II):

4. use of an incretin analogue according to any one of claims 1 to 3 for the preparation of a composition for:

activating the human glucagon-like peptide-1 receptor, glucose-dependent insulinotropic polypeptide receptor and/or glucagon receptor;

preventing, ameliorating or treating metabolic disorders; or

Reducing food intake, reducing fat, reducing body weight, or reducing blood glucose.

5. The use according to claim 4, wherein the metabolic disease comprises: hyperglycemia-related metabolic diseases or hyperlipidemia-related metabolic diseases;

preferably, the hyperglycemia-related metabolic disease includes: diabetes or metabolic syndrome associated with diabetes; preferably, the metabolic syndrome associated with diabetes includes insulin resistance, glucose intolerance; or

The hyperlipidemia-associated metabolic diseases include: obesity, hyperlipidemia, fatty liver, hypertriglyceridemia, hypercholesterolemia, low HDL cholesterol, high LDL cholesterol; preferably, the fatty liver comprises non-alcoholic fatty liver disease, more preferably non-alcoholic fatty hepatitis.

6. A composition comprising an incretin analog according to any of claims 1 to 3, and a carrier; the carrier is a pharmaceutically, dietotherapy or health care dietotherapy acceptable carrier.

7. A glucagon-like polypeptide for use in the preparation of an incretin analog of claim 1, having an amino acid sequence according to formula (I):

YSEGTFTSDX10SKYLDSQAAQDFVQWLLAGGPSSGAPPPSX40 (I);

in the formula (I), X10Is amino acid K, X40Selected from the group consisting of OH or NH2

8. A method of preparing the incretin analog of claim 1, comprising: linking the glucagon-like polypeptide of claim 7 to a long chain fatty acid;

preferably, the long-chain fatty acid is a fatty acid containing 14-20 carbons; more preferably a fatty acid containing 16 to 18 carbon atoms; more preferably the long chain fatty acid is a straight chain saturated monocarboxylic acid; more preferably, the long-chain fatty acid is palmitic acid;

preferably, the long chain fatty acid is linked to amino acid K of the peptide chain of the glucagon-like polypeptide; more preferably, is connected to X10To the amino acid of (a); more preferably, the linkage is cross-linking;

preferably, the glucagon-like polypeptide is linked to the long chain fatty acid through a linker; more preferably, the linker is a linker that is reactive with amino acid K and reactive with the reactive group of a long chain fatty acid; more preferably, the linker is a linker comprising at least 1 unit of- γ -glutamyl-.

9. A method of non-therapeutically reducing food intake, fat, weight or blood glucose comprising administering to a subject in need of such reduction the incretin analogs of any one of claims 1 to 3, or the composition of claim 6.

10. A kit comprising:

an incretin analog according to any one of claims 1 to 3; or

The composition of claim 6.

Technical Field

The invention relates to the field of biotechnology, in particular to an incretin analogue and a preparation method and application thereof.

Background

Hyperglycemia is caused by a defect in insulin secretion or an impaired biological action thereof, or both. Clinically, diabetes is generally classified into type i diabetes, type ii diabetes, and the like. Hyperglycemia symptoms exist in the long term of diabetes, and severe hyperglycemia can cause chronic damage and dysfunction of various tissues, particularly eyes, kidneys, hearts, blood vessels and nerves, and threaten human health. Especially, with the change of life style and the aging of population, diabetes and its complications become more and more important threats to human health.

The main categories of clinical treatments for type two diabetes in the prior art include: biguanide diabetes drugs (metformin, or phenformin), sulfonylurea diabetes drugs (glibenclamide, glipizide, glibornuride, glimepiride, or gliquidone), glucosidase inhibitor drugs (acarbose, voglibose, or miglitol), insulin sensitizing drugs (ciglitazone, troglitazone, rosiglitazone, or pioglitazone), aldose reductase inhibitor drugs (astatin, epalrestat, bolalastat, or torenostat), insulin release-promoting drugs (repaglinide), or nateglinide. Although various diabetes therapeutic drugs have been developed, there are also a number of negative factors that restrict the use of some drugs.

Among the currently marketed protein drugs, the main therapeutic agents for type II diabetes mellitus are human glucagon-like peptide-1 receptor (GLP-1R) agonists, such as Liraglutide (Liraglutide, trade name)And) Semaglutide (trade name)) And the like. Liraglutide is a chemically modified GLP-1 analogue, fatty acid (hexadecanoic acid) is connected to 26 th lysine of a GLP-1 protein skeleton through gamma-Glu, and the fatty acid can be combined with serum albumin, is clinically administered once a day and is respectively used for two indications of blood sugar reduction and weight loss. From the structural point of view, the somagluteptide is formed by replacing Ala with Aib at the 8-position, Lys with Arg at the 34-position and connecting octadecane fatty acid chain with Lys at the 26-position on a GLP-1(7-37) chain. Compared with liraglutide, the fatty acid chain of the somaglutide is longer, the affinity with serum albumin is higher, and clinically 1 subcutaneous injection is performed every week.

Patients with diabetes are generally obese, and weight loss is significantly improved in diabetes. Therefore, weight loss is an important indicator for GLP-1 analogues. Liraglutide, although approved for the treatment of obesity, actually lost only about 5.6 kg. While the average weight loss of the clinical treatment groups of somaglutide (0.5mg) and somaglutide (1.0mg) was 4.2kg and 5.5 kg. Current drugs for obesity generally lose weight by about 5-10% (compared to placebo), i.e. the average weight loss ratio as a whole does not exceed 10% of the patient's body weight (Rudolph l. leibel et al, Diabetes, 64(7): 2299-.

Currently, although many pharmaceutical companies and research institutes are developing multispecific drugs based on native incretin sequences, there is still a lack of clinically superior drugs, and further research and development in this regard is needed in the art.

Disclosure of Invention

The invention aims to provide an incretin analogue, and a preparation method and application thereof.

In a first aspect of the invention, there is provided an incretin analog comprising a glucagon-like polypeptide and, attached thereto, a long chain fatty acid; wherein, the amino acid sequence of the glucagon-like polypeptide is shown as formula (I):

YSEGTFTSDX10SKYLDSQAAQDFVQWLLAGGPSSGAPPPSX40(I);

in the formula (I), X10Is amino acid K, X40Selected from the group consisting of OH or NH2

In a preferred embodiment, the long-chain fatty acid is a fatty acid containing 14-20 carbons; preferably a fatty acid containing 16 to 18 carbon atoms; more preferably the long chain fatty acid is a straight chain saturated monocarboxylic acid; more preferably, the long chain fatty acid is palmitic acid.

In another preferred embodiment, said long chain fatty acid is linked to amino acid K of the peptide chain of said glucagon-like polypeptide; preferably, is connected to X10To the amino acid of (a); preferably, the linkage is cross-linked.

In a preferred embodiment, said glucagon-like polypeptide is linked to said long chain fatty acid via a linker; preferably, the linker is a linker that is reactive with amino acid K and reactive with the reactive group of the long chain fatty acid.

In a preferred embodiment, the linker is a linker comprising at least 1 (e.g., 1 to 6) units of- γ -glutamyl- (- γ Glu-).

In a preferred embodiment, the incretin analog has the structure of formula (II):

in another aspect of the invention there is provided the use of said incretin analogue in the preparation of a composition for:

activating the human glucagon-like peptide-1 receptor (GLP-1R), glucose-dependent insulinotropic polypeptide receptor (GIPR) and/or glucagon receptor (GCGR);

preventing, ameliorating or treating metabolic disorders; or

Reducing food intake, reducing fat, reducing body weight, or reducing blood glucose.

In a preferred embodiment, said reduction of food intake, fat, weight or blood glucose may be in a non-therapeutic situation.

In a preferred embodiment, the metabolic disease comprises: hyperglycemia-related metabolic diseases or hyperlipidemia-related metabolic diseases.

In another preferred example, the hyperglycemia-related metabolic disease includes: diabetes or diabetes-related metabolic syndrome; preferably, the metabolic syndrome associated with diabetes comprises insulin resistance, glucose intolerance.

In another preferred embodiment, the hyperlipidemia-associated metabolic disease comprises: obesity, hyperlipidemia, fatty liver, hypertriglyceridemia, hypercholesterolemia, low HDL cholesterol, high LDL cholesterol; preferably, the fatty liver comprises nonalcoholic fatty liver disease (NAFLD), more preferably nonalcoholic steatohepatitis (NASH).

In another aspect of the present invention, there is provided a composition comprising the incretin analog as described above, and a carrier; the carrier is a pharmaceutically, dietotherapy or health care dietotherapy acceptable carrier.

In a preferred embodiment, the incretin analog is in an effective amount.

In another preferred embodiment, the composition includes, but is not limited to: pharmaceutical compositions, food compositions or nutraceutical compositions, and the like.

In another aspect of the present invention, there is provided a glucagon-like polypeptide useful for the preparation of an incretin analog of claim 1, having an amino acid sequence according to formula (I):

YSEGTFTSDX10SKYLDSQAAQDFVQWLLAGGPSSGAPPPSX40(I);

in the formula (I), X10Is amino acid K, X40Selected from the group consisting of OH or NH2

In another aspect of the present invention, there are provided polynucleotides encoding said glucagon-like polypeptides, expression vectors comprising said polynucleotides and/or recombinant cells comprising said polynucleotides.

In another aspect of the present invention, there is provided a method for preparing the incretin analog, comprising: linking the glucagon-like polypeptide with long-chain fatty acid.

In another aspect of the invention, there is provided a method of non-therapeutically reducing food intake, fat, weight or blood glucose comprising administering to a subject in need of such reduction the incretin analog, or the composition.

In another aspect of the present invention, there is provided a kit comprising: the incretin analog; or comprising said composition.

Other aspects of the invention will be apparent to those skilled in the art in view of the disclosure herein.

Drawings

FIG. 1 is a graph showing the results of mass spectrometry of the incretin analog P3 YELAN.

FIG. 2 is a graph showing the results of measurement of GLP-1R agonistic activity in vitro of the incretin analog P3 YELAN; dolastatin was used as a control.

FIG. 3 is the measurement result of in vitro GIPR agonistic activity of incretin analog P3 YELAN.

FIG. 4 is a graph showing the results of measurement of in vitro GCGR agonistic activity of the incretin analog P3 YELAN.

FIG. 5 shows the results of the measurement of the in vitro serum stability of the incretin analog P3 YELAN; YELAN and dolastatin were used as controls.

FIG. 6 is a graph showing the results of random blood glucose changes in db/db mice for the incretin analog P3 YELAN; the dolastatin is used as a positive control; normal control (Normal control) is a Normal non-diseased animal; the Negative control (Negative control) was a solvent control (no P3YELAN or positive control drug was administered).

FIG. 7 shows the results of the body weight changes of the incretin analog P3YELAN in DIO mice; liraglutide was used as a positive control; normal control (Normal control) is Normal non-induced obese animals; the Negative control (Negative control) was a solvent control (no P3YELAN or positive control drug was administered).

FIG. 8 is the cumulative feeding change results of the incretin analog P3YELAN in DIO mice; liraglutide was used as a positive control; normal control (Normal control) is Normal non-induced obese animals; the Negative control (Negative control) was a solvent control (no P3YELAN or positive control drug was administered).

FIG. 9 shows the fasting blood glucose measurement of the incretin analog P3YELAN in DIO mice; liraglutide was used as a positive control; normal control (Normal control) is Normal non-induced obese animals; the Negative control (Negative control) was a solvent control (no P3YELAN or positive control drug was administered).

Detailed Description

In view of the defects of the incretin medicines in the prior art, the inventor provides the incretin analogue which has GLP-1R/GIPR/GCGR agonistic activity and is a triple-effect agonist with obvious effects on reducing blood sugar, fat and weight. The invention also provides a preparation method and application thereof.

Incretin analogue and preparation thereof

The invention provides an incretin analogue which comprises a glucagon-like polypeptide and a long-chain fatty acid connected with the glucagon-like polypeptide.

The sequence of the glucagon-like polypeptide used to establish the incretin analog is (SEQ ID NO: 2):

YSEGT FTSDX10SKYLD SQAAQ DFVQW LLAGG PSSGA PPPSX40(I)

wherein, X10=K,X40Is OH or NH2

The glucagon-like polypeptides of the invention can be recombinant polypeptides, synthetic polypeptides. It can be a product of chemical synthesis or produced from prokaryotic or eukaryotic hosts (e.g., bacterial, yeast, higher plant, insect, and mammalian cells) using recombinant techniques.

The invention also includes fragments, derivatives and analogs of the glucagon-like polypeptide of SEQ ID NO. 2. As used herein, the terms "fragment," "derivative," and "analog" refer to a polypeptide that retains substantially the same biological function or activity of the glucagon-like polypeptide. The polypeptide fragment, derivative or analogue may be (i) a polypeptide having one or more (e.g. 1-5, 1-3 or 1-2) conserved or non-conserved amino acid residues (preferably conserved amino acid residues) substituted for such a substituted amino acid residue, which may or may not be encoded by the genetic code, or (ii) a polypeptide having a substituent group in one or more (e.g. 1-5, 1-3 or 1-2) amino acid residues, or (iii) a polypeptide in which an additional amino acid sequence is fused to the polypeptide sequence (e.g. a leader or secretory sequence or a sequence used to purify the polypeptide or a proprotein sequence, or a fusion protein). Such fragments, derivatives and analogs are within the purview of those skilled in the art in view of the definitions herein.

Also included in the invention are polypeptides having at least 75% (preferably at least 80%, 85%, 90%, 95%) sequence identity (sequence identity) to the amino acid sequence of the glucagon-like polypeptide set forth in SEQ ID NO:2 and having the function of the glucagon-like polypeptide set forth therein.

In the present invention, modified forms of polypeptides (usually without changing the primary structure) comprising one or more amino acids modified to increase the stability, half-life, or efficacy of the polypeptide are also included, including: chemically derivatized forms of the polypeptide, such as acetylation or carboxylation, in vivo or in vitro. Modifications also include glycosylation. Modified forms also include sequences having phosphorylated amino acid residues (e.g., phosphotyrosine, phosphoserine, phosphothreonine). Also included are polypeptides modified to improve resistance to hydrolysis or to optimize solubility.

The invention also provides polynucleotide sequences encoding the glucagon-like polypeptides of the invention or conservative variant polypeptides thereof. The polynucleotide of the present invention may be in the form of DNA or RNA. The DNA may be the coding strand or the non-coding strand. That is, a "polynucleotide encoding a polypeptide" may include a polynucleotide encoding the polypeptide, and may also include additional coding and/or non-coding sequences.

The present invention also relates to expression vectors comprising the polynucleotides of the present invention, as well as genetically engineered host cells produced with the coding sequences for the expression vectors or glucagon-like polypeptides, and methods for producing the polypeptides by recombinant techniques.

The invention also provides application of the glucagon-like polypeptide in preparation of the incretin analogue.

The long-chain fatty acid used for establishing the incretin analogue is a fatty acid containing 14-20 carbon atoms, preferably 16-18 carbon atoms. In the present invention, esters, ethers or derivatives of the long chain fatty acids are also included, as are salts (e.g., sodium salts) of the long chain fatty acids. In a preferred embodiment of the present invention, the long-chain fatty acid is a straight-chain saturated monocarboxylic acid.

In a preferred embodiment of the present invention, the linkage is chemical crosslinking. For the purpose of chemical crosslinking, the present inventors introduced amino acids K or C on the basis of the original sequence of the polypeptide and crosslinked fatty acids to K or C.

After the glucagon-like polypeptide is crosslinked with long-chain fatty acid, the activity of the glucagon-like polypeptide is obviously improved, and the half-life period of the medicine in vivo is obviously prolonged. Preferably, Y at position 10 is mutated to K (in formula I: X)10K) so that the fatty acid chains can be cross-linked to the incretin analog polypeptide fragment by K.

In a preferred embodiment of the present invention, the incretin analog provided by the present invention, wherein the fatty acid chain is palmitic acid (C16), which is a linear monocarboxylic acid, and the chemical structural formula of which is a group shown below:

in the incretin analogue provided by the invention, a joint can be arranged between the glucagon-like polypeptide fragment and the long-acting carrier. The linker may generally be reacted with a lysine residue K and/or a cysteine residue C on the glucagon-like polypeptide fragment, respectively, and an active group on the long-acting carrier (for example, the linker may include an active group such as carboxyl, maleimide, etc.), so that the two ends of the linker are connected to the long-acting carrier and the incretin analog polypeptide fragment, respectively, to achieve cross-linking of the long-acting carrier and the incretin analog polypeptide fragment, for example, various types of condensation reactions.

The linker may be any of a variety of linkers suitable for use in the art for linking the incretin analog polypeptide fragment to a long acting carrier, and in some embodiments of the invention, the linker may be-gammaglu- (-gammaglutamyl-). A group of the formula:

as described above, the C-terminal amino acid of the incretin analog provided by the present invention may be modified, such as amidated.Amidation generally refers to the conversion of the C-terminal-COOH group to-CONH2Radicals, for example of formula (I): x40Is NH2

In some embodiments of the invention, the specific sequence of the incretin analog is shown as SEQ ID NO 6 in Table 1. In Table 1, the alignment of the polypeptide fragments with Glucagon (SEQ ID NO:1, GCG for short), GLP-1(SEQ ID NO:3) and GIP (SEQ ID NO:4) is also presented.

TABLE 1

In Table 1, -gamma E-or-gamma Glu (- γ -glutamyl-), "gamma E-C16" indicates that palmitoyl group is conjugated to the epsilon nitrogen of lysine via a-gamma-glutamyl-linker. "2 xOEG" means 2-OEG- (-2- (2- (2-aminoethoxy) ethoxy) acetyl-) linkages.

The invention also provides a preparation method of the incretin analogue, which comprises the step of connecting the glucagon-like polypeptide with long-chain fatty acid.

The preparation method can comprise the following steps: preparing the incretin analogue by using a chemical synthesis method; the preparation method can also comprise the following steps: culturing a suitable host cell under suitable conditions to express the incretin analog polypeptide fragment, separating and purifying to obtain the incretin analog polypeptide fragment, and chemically crosslinking the long-acting carrier to the incretin analog polypeptide fragment. The incretin analogs of the invention can be prepared by standard peptide synthesis methods, e.g., by standard solid or liquid phase methods, stepwise or by fragment assembly, and isolation and purification of the final incretin analog polypeptide fragment, incretin analog product, or by any combination of recombinant and synthetic methods.

Application of incretin analogue

The invention also provides application of the incretin analogue in preparing a medicament for treating metabolic diseases and GLP-1R/GIPR/GCGR pleiotropic agonist. The metabolic disease may be selected from diabetes, obesity, dyslipidemia, nonalcoholic fatty liver disease (NAFLD)/nonalcoholic steatohepatitis (NASH), other metabolic syndromes associated with diabetes, including hypertriglyceridemia, low HDL cholesterol and high LDL cholesterol, insulin resistance, obesity, glucose intolerance, etc.

The invention also provides a method for treating a disease, comprising the steps of: administering to the subject the incretin analogue provided in the first aspect of the invention. In a random blood sugar detection test, the blood sugar reducing effect and the weight losing effect of a diabetes model mouse applied with the incretin analogue are obviously superior to those of a control group sample.

In addition to applications in the prevention, alleviation or treatment of disease, the incretin analogs of the present invention can also be applied in some non-therapeutic applications. Partial results according to embodiments of the present invention show that the incretin analogues are capable of very significantly reducing food intake, reducing fat, reducing body weight or reducing blood glucose. Thus, the incretin analogs of the present invention may also be used in subjects who have no disease characteristics of the body, but who have a need to control food intake, reduce fat, and lose weight.

Although the art has been studying the development of molecules with multiple agonistic activities, it is actually very difficult to obtain an ideal drug of this type. The first is the problem of safety, in particular immunogenicity. The hypoglycemic slimming medicine needs to be used for a long time and has extremely high requirement on safety. In order to design and obtain a polypeptide with high multiple agonistic activity and stability in vivo, the prior technical scheme often introduces more mutation sites, and often introduces unnatural amino acids and other modifications. Both these mutations, as well as the introduction of unnatural amino acids, increase the risk of potential immunogenicity. The safety of the medicine for treating diabetes, obesity and other diseases is extremely important. In addition, for small peptides of about 30 amino acids in length such as GLP-1, Glucagon, the change in sequence is extremely sensitive to the change in activity; in the case of multiple active polypeptides, the changes are more complex due to the involvement of agonism at multiple different receptors, and it is not at all predictable what the effect of agonistic activity at a receptor will be after any amino acid change. For example, Joseph channel et al reported (Joseph channel et al, Molecular Metabolism, 3: 293-. However, it can be seen from other reports that, when a single or several of the above-mentioned sites are simultaneously mutated, and substituted with other amino acids, the activity change does not always coincide with the result of alanine scanning. As reported by Jonathan W Day et al (Jonathan W Day et al, Nature Chemical Biology, 5: 749-. Second, the Joseph channenne study suggested that substitution with alanine at position 23 would result in almost complete loss of GCGR agonist activity (retention of only 1.1%); however, Jonathan W Day et al mutated 23 to Ile did not show a decrease in GCG activity. For example, the second position S is considered to be very important for maintaining the GCG activity (the activity is only 1/3 when the mutation is Ala) as a result of alanine scanning, but Brian Finan et al report (Finan B et al, Nat Med.2015; 21:27-36.) that the relative agonistic activity of GCGR is improved to 200% -640% by performing 2S → Aib, 2S → dSer, 2S → G, 2S → dAla substitution mutations on the second amino acid of GCG, respectively, and combining the mutations at other sites. In the studies of the present inventors, it was also found that when a combination of mutations which are advantageous for increasing GLP-1, GCG or GIP activity is introduced, the effect is quite often not consistent with that of single site mutation. In addition, polypeptides such as GLP-1, Exendin-4, GCG or GIP, in which amino acids are added or reduced at the N-and C-termini, affect their biological activities. If one or two amino acids are removed from the N-terminus, the agonistic activity of GLP-1, GCG, etc. is completely lost. For example, Oxyntomodulin has only 8 amino acids more than the C-terminus of Glucagon, and thus about 90% of its GCGR agonistic activity is lost (Alessandro Pocai et al, Diabetes; 58(10): 2258-.

The incretin analogue provided by the invention has extremely high GLP-1R and GIPR (glutathione peptide receptor) agonist activities and slightly weak GCGR agonist activities, and surprisingly, the in vitro activity of the incretin analogue polypeptide is remarkably changed before and after fatty acid crosslinking.

The embodiments of the present invention are described below with reference to specific embodiments, and other advantages and effects of the present invention will be easily understood by those skilled in the art from the disclosure of the present specification. The invention is capable of other and different embodiments and of being practiced or of being carried out in various ways, and its several details are capable of modification in various respects, all without departing from the spirit and scope of the present invention.

Composition comprising a metal oxide and a metal oxide

The invention also provides a composition comprising an effective amount of an incretin analog of the invention and a carrier; the carrier is a pharmaceutically, dietotherapy or health care dietotherapy acceptable carrier. Such compositions include, but are not limited to: pharmaceutical compositions, food compositions or nutraceutical compositions, and the like.

In the present invention, the pharmaceutical composition may contain the incretin analog in a weight ratio of 0.01 to 95% (e.g., 0.1%, 1%, 5%, 10%, 20%, 30%, 50%, 80%, etc.).

As used herein, an ingredient that is "pharmaceutically, dietetically or nutraceutically acceptable" is one that is suitable for use in humans and/or animals without undue adverse side effects (such as toxicity, irritation and allergic response), i.e., at a reasonable benefit/risk ratio; such as pharmaceutical carriers or excipients commonly used in the art.

As used herein, "effective amount" or "effective dose" refers to an amount that produces a function or activity in a human and/or animal and is acceptable to the human and/or animal as used herein.

The dosage form of the pharmaceutical composition of the present invention may be various, as long as it is a dosage form that enables the active ingredient to efficiently reach the body of a mammal. Such as may be selected from: gels, aerosols, tablets, capsules, powders, granules, syrups, solutions, or suspensions. Depending on the type of disease to be treated by the compounds of the present invention, one skilled in the art may select a dosage form that is convenient to use.

Suitable pharmaceutically acceptable carriers are well known to those of ordinary skill in the art. Sufficient details regarding pharmaceutically acceptable carriers can be found in Remington's Pharmaceutical Sciences (Mack pub. co., n.j.1991). Pharmaceutically acceptable carriers in the compositions may comprise liquids such as water, phosphate buffered saline, ringer's solution, physiological saline, balanced salt solution, glycerol or sorbitol, and the like. In addition, auxiliary substances, such as lubricants, glidants, wetting or emulsifying agents, pH buffering substances and stabilizers, such as albumin and the like, may also be present in these carriers.

Preferred pharmaceutical compositions are solid compositions, especially tablets and solid-filled or liquid-filled capsules, from the standpoint of ease of preparation and administration. The compounds of the invention or pharmaceutical compositions thereof may also be stored in sterile devices suitable for injection or instillation.

The effective administration dose of the incretin analog as an active ingredient of the present invention may vary depending on the mode of administration and the severity of the disease to be treated, for example, at a dose of about 0.00001 to 10mg/kg body weight per day. The timing of administration may also be adjusted, for example involving administration in a sustained release form, with the administration of one drug at intervals of days or several days. The dosage regimen may be adjusted to provide the optimum therapeutic response.

When the composition is applied to large animals and patients, the effective dosage (including solid or solution dosage conversion) of the large animals or the patients can be converted according to the dosage of the small animals and through a corresponding professional conversion formula. In the specific examples of the present invention, some dosing regimens for animals such as mice are given. The conversion from the administered dose in animals such as mice to the administered dose suitable for humans is easily done by the person skilled in the art, and can be calculated, for example, according to the Meeh-Rubner formula: Meeh-Rubner formula: a ═ k × (W2/3)/10,000. Wherein A is the body surface area, calculated as m 2; w is body weight, calculated as g; k is constant and varies with species of animal, in general, mouse and rat 9.1, guinea pig 9.8, rabbit 10.1, cat 9.9, dog 11.2, monkey 11.8, human 10.6. It will be appreciated that the conversion to a given dose may vary depending on the drug and clinical situation, as assessed by an experienced pharmacist.

The invention also provides a kit or kit comprising: the incretin analog; or the pharmaceutical composition. For clinical use, the kit or kit of the present invention may further comprise other auxiliary components, such as a syringe. The kit or kit may further comprise instructions for use, which will facilitate the use of the kit or kit in a proper manner by a person skilled in the art.

Before the present embodiments are further described, it is to be understood that the scope of the invention is not limited to the particular embodiments described below; it is also to be understood that the terminology used in the examples is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention.

When numerical ranges are given in the examples, it is understood that both endpoints of each of the numerical ranges and any value therebetween can be selected unless the invention otherwise indicated. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. In addition to the specific methods, devices, and materials used in the examples, any methods, devices, and materials similar or equivalent to those described in the examples may be used in the practice of the invention in addition to the specific methods, devices, and materials used in the examples, in keeping with the knowledge of one skilled in the art and with the description of the invention.

Unless otherwise indicated, the experimental methods, detection methods, and preparation methods disclosed herein all employ techniques conventional in the art of molecular biology, biochemistry, chromatin structure and analysis, analytical chemistry, cell culture, recombinant DNA technology, and related arts. These techniques are well described in the literature, and may be found in particular in the study of the MOLECULAR CLONING, Sambrook et al: a LABORATORY MANUAL, Second edition, Cold Spring Harbor LABORATORY Press, 1989and Third edition, 2001; ausubel et al, Current PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, New York, 1987and periodic updates; the series METHODS IN ENZYMOLOGY, Academic Press, San Diego; wolffe, CHROMATIN STRUCTURE AND FUNCTION, Third edition, Academic Press, San Diego, 1998; (iii) METHODS IN ENZYMOLOGY, Vol.304, Chromatin (P.M.Wassarman and A.P.Wolffe, eds.), Academic Press, San Diego, 1999; and METHODS IN MOLECULAR BIOLOGY, Vol.119, chromatography Protocols (P.B.Becker, ed.) Humana Press, Totowa, 1999, etc.

The abbreviations referred to in the examples have the following meanings:

RT: room temperature;

DMF: n, N-dimethylformamide;

fmoc: 9H-fluoren-9-ylmethoxycarbonyl;

trt: a trityl group;

boc: a tert-butoxycarbonyl group;

HOBt: 1-hydroxybenzotriazole;

tBu is tert-butyl;

DCM: dichloromethane;

DBLK: 20% N, N-dimethylformamide piperidine;

DIC: n, N' -diisopropylcarbodiimide;

MeOH: methanol;

TFA: trifluoroacetic acid;

Fmoc-Lys(Pal-Glu-OtBu)-OH:Nαfluorenylmethoxycarbonyl- (N)ε- (gamma-glutamyl (N)α-hexadecyl, α -tert-butyl))) lysine;

- γ E-: - γ -glutamyl-;

-OEG-: -2- (2- (2-aminoethoxy) ethoxy) acetyl-

DIEA: n, N-diisopropylethylamine;

MTBE: methyl tert-butyl ether.

TFEA: 2,2, 2-trifluoroethanol

Pd (PPh3) 4: tetrakis (triphenylphosphine) palladium

And (3) Alloc: allyloxycarbonyl radical

The various commercially available amino acids and amino acid fragments, and the various commercially available resins referred to in the examples were produced by the following manufacturers and commercial models:

the Fmoc protecting group amino acid raw material, the 2-CTC resin and the Rink amide MBHA resin are all conventional commercial reagents (protected amino acid manufacturer: Chengdong source Biochemical technology Co., Ltd., resin manufacturer: Tianjin Nankai and ChengTech Co., Ltd.);

the organic solvent and other raw materials are all commercially available products (manufacturer: national drug group chemical reagents limited company; chemical purity).

In addition, the conditions for HPLC and mass spectrometry and the equipment types and manufacturers used are as follows:

the instrument comprises the following steps: HPLC UltiMate 3000; the test conditions are shown in table 2 below.

TABLE 2

Preparing a liquid phase: beijing Innovation, LC 3000.

Mass spectrum: the instrument model is 5800MALDI-TOF (AB SCIEX), the analysis software is T0F/TOF Explorer, Data Explorer, MS adopts Reflector Positive parameters: CID (OFF), Mass ranging (700 + 6500Da) Focus Mass (1200Da) Fixed laser intensity (5600) Digitizer: bin Size (0.5ns)

EXAMPLE 1 preparation of incretin analogs

1. Preparation of incretin analogue P3YELAN

The structure of the incretin analogue P3YELAN is as follows:

namely: YSEGT FTSDX10 SKYLD SQAAQ DFVQW LLAGG PSSGA PPPS-NH2

X10K (palmitoyl- γ E). Synthesis of Fmoc-Ser (tBu) -Rink amide MBHA resin:

2.65g of Rink amide MBHA resin (Tianjin Nankai and science and technology Co., Ltd.) having a substitution degree of 0.38mmol/g was weighed into a solid phase reaction column, 10mL of DCM was added to swell the resin for 30 minutes, and the resin was washed 3 times with 10mL of DMF each time. 15mL of DBLK solution was added to the reaction column, reacted for 5 minutes, filtered, washed once with 20mL of DMF, and then added with 15mL of DBLK solution, reacted for 10 minutes, and Kaiser detected positive. Suction filtration and washing with DMF 3 times 20mL each time.

Dissolving 1.91g of Fmoc-Ser (tBu) -OH and 0.81g of HOBt by using 10ml of DMF, adding 0.69g of DIC at the temperature of 5-8 ℃ for activation for 5min, adding the mixture into the reaction column filled with the resin, and reacting for 2 hours. After the Kaiser test is negative, the peptide is directly used for the next step of peptide resin synthesis.

Synthesis of peptide resin:

the resin Fmoc-Ser (tBu) -Rink amide MBHA resin (1.0mmol) was weighed out, added to a reaction column, swollen with 20mL of DCM for 30 minutes, and washed 3 times with 20mL of DMF each time. After the completion of the washing, 10mL of DBLK solution (20% piperidine/DMF (V/V)) was added to the reaction column, reacted for 5 minutes, filtered with suction, washed once with 20mL of DMF, and added with 10mL of DBLK solution (20% piperidine/DMF (V/V)) and reacted for 10 minutes, which was positive by Kaiser test. Suction filtration and washing with DMF 3 times 20mL each time. And adding Fmoc-Pro-OH (1.69g, 5.0eq) and HOBt (0.81g, 6.0eq) into 10mL of DMF for dissolving, adding DIC (0.69g, 5.5eq) at 5-8 ℃ for activating for 5min, adding into a reaction column, reacting for 1 h, detecting by Kaiser to be negative, completely reacting, and washing with DMF for 3 times, 20mL each time. Repeating the above deprotection and coupling operations, and completing the coupling of other amino acids in sequence according to the sequence of the peptide, wherein X10Coupling was performed using Fmoc-Lys (Pal-Glu-OtBu) -OH (Chengdong Biochemical technology Co., Ltd.). After the last amino acid coupling was completed, deprotection was performed according to the above deprotection method, and after complete deprotection, DMF washing was performed sequentially for 2 times, MeOH washing 2 times, DCM washing 2 times, and MeOH washing 2 times, each time with 20mL of washing solvent. Collecting materials, and drying under reduced pressure at normal temperature to obtain the target peptide resin.

Cleavage of the crude peptide:

5.02g of the peptide resin was weighed, and slowly added to 60mL of a lysate (trifluoroacetic acid: thioanisole: ethylenedithioglycol: 90:5:3:2) at 20-30 ℃ to complete the reaction for 2 hours. After completion of the reaction, the resin was removed by filtration, the filtrate was poured into previously precooled methyl tert-ether (600mL) with vigorous stirring, and the resulting mixed solution was left to settle in a refrigerator for 2 hours. The supernatant was removed and washed 5 times with 400mL portions of precooled methyl tert-ether by centrifugation. After the completion, the crude peptide was collected and dried under reduced pressure at room temperature to obtain 2.23g of crude peptide.

Purification of the crude peptide:

the crude peptide was refined by multi-step purification using preparative liquid phase (beijing innovational sumen, LC 3000): the first step is as follows: stationary phase: c18 (Daisogel: sp-120-40/60-C18-RPS), mobile phase 0.1% TFA, acetonitrile; the second step is that: stationary phase: c8 (Daisogel: sp-120-10-C8-P), mobile phase: 0.5% phosphoric acid, acetonitrile, third step: stationary phase: c8 (Daisogel: sp-120-10-C8-P), mobile phase: 50mM ammonium acetate, 0.3% acetic acid, acetonitrile, and finally lyophilized (lyophilizer Beijing Bo Yi kang, FD-2A) to yield the refined peptide (98.0%). And finally, determining the precise molecular weight of the refined peptide by adopting MS: m/z4455.69(M + H)+. The MS is shown in figure 1.

2. Preparation of glucagon derivative P5YELAN

The structural formula of the glucagon derivative P5YELAN is as follows:

namely: YSEGT FTSDX10 SKYLD SQAAQ DFVQW LLAGG PSSGA PPPS-NH2 X10K (((octadecanedioic acid monoacyl) - γ E) -2 xeeg)

Solid phase synthesis of branched protected amino acid W1: Alloc-Lys ((octaneedioic Acid mono-tert-butyl ester) -Glu-OtBu) -OEG-OEG) -OH, structure as follows:

synthesis of W1:

weighing 20g of 2-CTC resin with the substitution degree of 1.0mmol/g, adding the resin into a solid phase reaction column, adding the resin into the solid phase reaction column, washing the solid phase reaction column with DMF for 1 time, swelling the resin with DMF for 30 minutes, dissolving 8.53g of Alloc-Lys (Fmoc) -OH (20mmol) with DMF, adding 7.5ml of DIEA (45mmol) into ice water bath for activation, adding the activated product into the reaction column filled with the resin, reacting for 2 hours, adding 30ml of anhydrous methanol for blocking for 1 hour, and washing the product with DMF for 3 times. The Fmoc protection was removed from the mixture of DMF and pyridine at a volume ratio of 4:1, followed by washing 6 times with DMF and weighing 15.42g of [2- [2- (Fmoc-amino) ethoxy ] ethyl]Ethoxy radical]Acetic acid and 5.41g of HOBt were dissolved in DMF, and 6.2ml of DIC was added in an ice water bath to activate the mixture, and then the activated product was put into the above reaction column filled with the resin to react at room temperature for 2 hours. Repeating the steps of removing Fmoc protection and adding corresponding materials for coupling, and sequentially finishing the [2- [2- (Fmoc-amino) ethoxy ] according to the sequence of the branched chain fragments]Ethoxy radical]Acetic acid, Fmoc-Glu-OtBu and mono-tert-butyl octadecanedioate. After coupling, the resin was washed 3 times with DMF, 5 times with MeOH and drained. The resin was added to 400ml TFEA/DCM 1:4 and reacted for 4h at room temperature. After filtration of the resin, DCM was removed from the filtrate and the filtrate was taken up in 500ml of MTBE for settling and drying by centrifugation to give 19.43g of the title compound M/Z1242.51 (M + H)+

The polypeptide is synthesized as P3YELAN, X10Coupling was carried out using W1 and the Alloc groups were removed using Pd (PPh3) 4. The crude peptide obtained was purified by RP-HPLC and finally lyophilized to give the refined peptide (96.5%). MS for precise molecular weight determination of protamine: m/z 4804.13(M + H)+

3. Preparation of glucagon derivative P9 YELAN:

the structural formula of the glucagon derivative P9YELAN is as follows:

namely: YSEGT FTSDX10 SKYLD SQAAQ DFVQW LLAGG PSSGA PPPS-NH2 X10K (((eicosanedioic acid monoacyl) - γ E) -2 xeeg)

Synthesis of protected amino acid with branched chain as in P5YELAN, the protected amino acid with branched chain W2 was synthesized by solid phase method: Alloc-Lys ((Eicosanedioic Acid mono-tert-butyl) -Glu-OtBu) -OEG-OEG) -OH (wherein the fatty Acid coupling employs mono-tert-butyl eicosanoate) has the following structure:

then performing polypeptide synthesis, wherein the polypeptide synthesis is the same as P3YELAN, and X10Coupling was carried out using W2 and the Alloc groups were removed using Pd (PPh3) 4. The resulting crude peptide was purified by RP-HPLC to give the refined peptide (97.1%). MS determination of precise molecular weight for protamine: m/z 4832.50(M + H)+

Example 2 in vitro cytological Activity assay

Measurement of GLP-1R agonistic Activity

GLP-1R agonistic activity was measured using a luciferase reporter assay (Jonathan W Day et al: Nat Chem biol.2009 Oct; 5(10): 749-57). Cloning the human GLP-1R gene into a mammalian cell expression plasmid pCDNA3.1 to construct a recombinant expression plasmid pCDNA3.1-GLP-1R, and simultaneously cloning the Luciferase (Luciferase) full-length gene into a pCRE plasmid to obtain the pCRE-Luc recombinant plasmid. The pcDNA3.1-GLP-1R and the pCRE-Luc plasmids transfect CHO-K1 cells according to the molar ratio of 1:10, and stably-expressed strains are screened.

Culturing cells in a 9-cm cell culture dish by using DMEM/F12 culture medium containing 10% FBS and 300 mu G/ml G418, when the confluency reaches about 90%, discarding the culture supernatant, adding 2ml pancreatin for digestion for 3min, adding 2ml DMEM/F12 culture medium containing 10% FBS and 300 mu G/ml G418 for neutralization, transferring to a 15ml centrifuge tube, centrifuging at 1000rpm for 5min, discarding the supernatant, adding 2ml DMEM/F12 culture medium containing 10% FBS and 300 mu G/ml G418 for resuspension, and counting. Cells were diluted to 1X 10 with DMEM/F12 medium containing 10% FBS5Perml, 100. mu.l per well in 96-well plates, i.e.1X 104Per well, after adherence, DMEM/F12 medium containing 0.2% FBS was used. After the supernatant was discarded from the cells plated in the 96-well plate, the purified glucagon derivative (positive control: Du: Du)Ladrolutide (Dulaglutide, trade name)) ) were diluted to a series of indicated concentrations with DMEM/F12 medium containing 1% BSA, added to cell culture wells at 100 μ l/well and tested after 6h of stimulation. Detection was performed according to the Lucifersae reporter kit (Ray Biotech, Cat:68-Lucir-S200) instructions. The viability assay was repeated 3 times for each sample.

(II) GIPR (general in vitro receptor) agonistic activity detection method

The GIPR agonistic activity assay also employs the luciferase reporter assay. Cloning the human GIPR gene into the expression plasmid pcDNA3.1 of the mammalian cell to construct a recombinant expression plasmid pCDNA3.1-GIPR, transfecting CHO-K1, and screening and constructing the stable transfer cell strain. The step of viability assay was the same as above (positive control group: native human GIP peptide), and the viability assay was repeated 3 times for each sample.

(III) GCGR agonistic activity detection method

The GCGR agonistic activity assay also employs the luciferase reporter assay. Cloning the humanized GCGR gene into the expression plasmid pcDNA3.1 of the mammalian cell to construct a recombinant expression plasmid pCDNA3.1-GCGR, transfecting CHO-K1, and screening and constructing stable transfer cell strains. The viability assay procedure was as above (positive control: native human GCG peptide) and the viability assay was repeated 3 times for each sample.

(IV) measuring the result of alive

FIGS. 2, 3, and 4 are graphs of the agonist activity results for GLP-1R, GIPR and GCGR, respectively, with specific EC50 shown in Table 3.

TABLE 3

The above results indicate that the P3YELAN of the present invention has high GLP-1R and GIPR agonistic activities, and also has significant GCGR agonistic activity. However, P5YELAN and P9YELAN having the same peptide sequence have a greatly decreased cell agonistic activity due to the difference in the linked fatty acid chains.

Example 3 serum stability

In this example, the serum stability of the incretin analog P3YELAN was determined using the following procedure:

(1) preparing the incretin analogue P3YELAN, YELAN and a reference substance (Dulaglutide) into a solution with the concentration of 1.0mg/ml by using 5mM Tris-HCl, pH8.5 and 0.02 percent TWEEN-80 solution, diluting the solution by 10 times by using rat serum after degerming and filtering (0.22 mu m and Millipore SLGP033RB), mixing the solution uniformly and subpackaging the mixture into a sterile centrifuge tube;

(2) freezing 3 tubes of the samples at-20 ℃ as a control, placing the rest in a constant temperature box at 37 ℃, and sampling for detecting the activity at 0 hour, 12 hours, 24 hours and 72 hours;

(3) GLP-1R agonistic activity is detected. Relative activity: activity value at 0 hour is 100%, and the values measured at subsequent time points are compared.

FIG. 5 shows the results of the residual activity of the incretin analog P3YELAN over time. The results show that P3YELAN retains higher GLP-1R agonistic activity over a longer period of time than YELAN and Dulaglutide.

Example 4 random blood glucose testing after dosing in db/db mice

Experiments on glucose lowering in mice with leptin receptor deficient type two diabetes mellitus (db/db). The db/db mice are screened and divided into groups according to three indexes of body weight, non-fasting blood glucose and pre-drug OGTT response, 6 mice in each group are excluded, and the non-fasting blood glucose is larger than 15 mM. P3YELAN was dissolved in 50mM phosphate buffer (pH 7.4), 5% sorbitol, 0.02% v/v Tween-80, and dolabrin or P3YELAN (multiple administration) was injected subcutaneously at a dose of 10nmol/kg/4d, and P3YELAN was used at low (1nmol/kg/d), medium (3nmol/kg/d), and high (6 nmol/kg/d). day0-day4 random blood glucose was monitored daily for all animals, and subsequently measured every 4 days on days scheduled at day6, 10, 14, 18, 22, 26, 30, 34.

The trend of blood glucose is shown in fig. 6. The results show that the blood glucose levels in the P3 YELAN-administered animals were much lower than that of dolaglutide (10nmol/kg/4d) at the dose of 3nmol/kg/d or 6 nmol/kg/d.

Example 5 weight loss experiments in diet-induced obese (DIO) mice

Preparation of DIO mouse model: male C57BL/6J male mice of about 7 weeks of age were given a high fat diet (60% kcal from fat) and kept on for about 16 weeks (23 weeks total) until they had a body weight of about 45g for the test. DIO mice were randomly divided into groups of 6 mice each, with no difference in basal body weight, and weighed daily. P3YELAN, liraglutide or PBS was injected subcutaneously. The administration dose of Liraglutide (Liraglutide) is 40 nmol/kg/d; the glucagon derivatives were used at low (5nmol/kg/d), high doses (20 nmol/kg/d). Body weights were weighed beginning on the first dosing Day and continuing until the end of the experiment Day30, with food intake recorded and weighed daily to be consistent. After the experiment, the blood fat and the fasting blood sugar are detected. Day28 fasted overnight and Day29 measured fasting blood glucose.

The results of measuring the weight change of the test animals are shown in fig. 7, and the results show that the P3YELAN of the three dose groups can remarkably reduce the weight of the test animals, and the weight reducing effect of the P3YELAN is better than that of the liraglutide.

The results of the food intake assay of the test animals are shown in fig. 8, and the results show that P3YELAN of the three dose groups can significantly reduce the food intake of the test animals, and the effect of reducing the food intake is better than that of liraglutide.

The fasting blood glucose measurement results of the tested animals are shown in fig. 9, and the results show that the P3YELAN of the three dose groups can remarkably reduce the fasting blood glucose of the tested animals, and the effect of reducing the fasting blood glucose is better than that of the liraglutide.

In conclusion, the P3YELAN peptide effectively overcomes some technical defects in the prior art and has good industrial application value.

The foregoing embodiments are merely illustrative of the principles and utilities of the present invention and are not intended to limit the invention. Any person skilled in the art can modify or change the above-mentioned embodiments without departing from the spirit and scope of the present invention. Accordingly, it is intended that all equivalent modifications or changes which can be made by those skilled in the art without departing from the spirit and technical spirit of the present invention be covered by the claims of the present invention.

Sequence listing

<110> Zhejiang Dalong Biotechnology Ltd

HANGZHOU HEZE PHARMACEUTICAL TECHNOLOGY Co.,Ltd.

<120> an incretin analogue, a preparing method and a use thereof

<130> 201452

<160> 8

<170> SIPOSequenceListing 1.0

<210> 1

<211> 29

<212> PRT

<213> Artificial Sequence (Artificial Sequence)

<220>

<221> PEPTIDE

<222> (1)..(29)

<223> Glucagon

<400> 1

His Ser Gln Gly Thr Phe Thr Ser Asp Tyr Ser Lys Tyr Leu Asp Ser

1 5 10 15

Arg Arg Ala Gln Asp Phe Val Gln Trp Leu Met Asn Thr

20 25

<210> 2

<211> 39

<212> PRT

<213> Artificial Sequence (Artificial Sequence)

<220>

<221> PEPTIDE

<222> (1)..(39)

<223> glucagon-like polypeptides

<220>

<221> BINDING

<222> (39)..(39)

<223> OH or NH2 modification

<400> 2

Tyr Ser Glu Gly Thr Phe Thr Ser Asp Lys Ser Lys Tyr Leu Asp Ser

1 5 10 15

Gln Ala Ala Gln Asp Phe Val Gln Trp Leu Leu Ala Gly Gly Pro Ser

20 25 30

Ser Gly Ala Pro Pro Pro Ser

35

<210> 3

<211> 31

<212> PRT

<213> Artificial Sequence (Artificial Sequence)

<220>

<221> PEPTIDE

<222> (1)..(31)

<223> GLP-1

<400> 3

His Ala Glu Gly Thr Phe Thr Ser Asp Val Ser Ser Tyr Leu Glu Gly

1 5 10 15

Gln Ala Ala Lys Glu Phe Ile Ala Trp Leu Val Lys Gly Arg Gly

20 25 30

<210> 4

<211> 42

<212> PRT

<213> Artificial Sequence (Artificial Sequence)

<220>

<221> PEPTIDE

<222> (1)..(42)

<223> GIP

<400> 4

Tyr Ala Glu Gly Thr Phe Ile Ser Asp Tyr Ser Ile Ala Met Asp Lys

1 5 10 15

Ile His Gln Gln Asp Phe Val Asn Trp Leu Leu Ala Gln Lys Gly Lys

20 25 30

Lys Asn Asp Trp Lys His Asn Ile Thr Gln

35 40

<210> 5

<211> 39

<212> PRT

<213> Artificial Sequence (Artificial Sequence)

<220>

<221> PEPTIDE

<222> (1)..(39)

<223> YELAN

<220>

<221> BINDING

<222> (39)..(39)

<223> OH modification

<400> 5

Tyr Ser Glu Gly Thr Phe Thr Ser Asp Tyr Ser Lys Tyr Leu Asp Ser

1 5 10 15

Gln Ala Ala Gln Asp Phe Val Gln Trp Leu Leu Ala Gly Gly Pro Ser

20 25 30

Ser Gly Ala Pro Pro Pro Ser

35

<210> 6

<211> 39

<212> PRT

<213> Artificial Sequence (Artificial Sequence)

<220>

<221> PEPTIDE

<222> (1)..(39)

<223> P3YELAN

<220>

<221> BINDING

<222> (10)..(10)

<223> palmitoyl-gamma E modification

<220>

<221> BINDING

<222> (39)..(39)

<223> NH2 modification

<400> 6

Tyr Ser Glu Gly Thr Phe Thr Ser Asp Lys Ser Lys Tyr Leu Asp Ser

1 5 10 15

Gln Ala Ala Gln Asp Phe Val Gln Trp Leu Leu Ala Gly Gly Pro Ser

20 25 30

Ser Gly Ala Pro Pro Pro Ser

35

<210> 7

<211> 39

<212> PRT

<213> Artificial Sequence (Artificial Sequence)

<220>

<221> PEPTIDE

<222> (1)..(39)

<223> P5YELAN

<220>

<221> BINDING

<222> (10)..(10)

<223> ((octadecanedioic acid monoacyl) -gamma AE) -2xOEG modification

<220>

<221> BINDING

<222> (39)..(39)

<223> NH2 modification

<400> 7

Tyr Ser Glu Gly Thr Phe Thr Ser Asp Lys Ser Lys Tyr Leu Asp Ser

1 5 10 15

Gln Ala Ala Gln Asp Phe Val Gln Trp Leu Leu Ala Gly Gly Pro Ser

20 25 30

Ser Gly Ala Pro Pro Pro Ser

35

<210> 8

<211> 39

<212> PRT

<213> Artificial Sequence (Artificial Sequence)

<220>

<221> PEPTIDE

<222> (1)..(39)

<223> P9YELAN

<220>

<221> BINDING

<222> (10)..(10)

<223> ((undecanedioic acid monoacyl) -gamma aE) -2xOEG modification

<220>

<221> BINDING

<222> (39)..(39)

<223> NH2 modification

<400> 8

Tyr Ser Glu Gly Thr Phe Thr Ser Asp Lys Ser Lys Tyr Leu Asp Ser

1 5 10 15

Gln Ala Ala Gln Asp Phe Val Gln Trp Leu Leu Ala Gly Gly Pro Ser

20 25 30

Ser Gly Ala Pro Pro Pro Ser

35

28页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:GIP-Exendin-4嵌合肽的分子改构及其二聚体在治疗糖尿病中的应用

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!