CA3085252A1 - Dual-function protein for lipid and blood glucose regulation - Google Patents

Dual-function protein for lipid and blood glucose regulation Download PDF

Info

Publication number
CA3085252A1
CA3085252A1 CA3085252A CA3085252A CA3085252A1 CA 3085252 A1 CA3085252 A1 CA 3085252A1 CA 3085252 A CA3085252 A CA 3085252A CA 3085252 A CA3085252 A CA 3085252A CA 3085252 A1 CA3085252 A1 CA 3085252A1
Authority
CA
Canada
Prior art keywords
dual
function protein
fgf21
protein
peptide
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
CA3085252A
Other languages
French (fr)
Inventor
Zhao DONG
Chi Zhou
Xiong FENG
Jiyu Zhang
Shixiang JIA
Qiang Li
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.)
Ampsource Biopharma Shanghai Inc
Original Assignee
Ampsource Biopharma Shanghai Inc
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 Ampsource Biopharma Shanghai Inc filed Critical Ampsource Biopharma Shanghai Inc
Priority to CA3085252A priority Critical patent/CA3085252A1/en
Publication of CA3085252A1 publication Critical patent/CA3085252A1/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • 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
    • C07K14/475Growth factors; Growth regulators
    • C07K14/50Fibroblast growth factors [FGF]
    • 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
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Abstract

The present disclosure relates to a dual-fonction protein for regulating blood glucose and lipid metabolism, wherein said dual-fonction protein comprises a human glucagon-like peptide 1 (GLP-1) analog and human fibroblast growth factor 21 (FGF21). In the present disclosure, provided is a method for preparing said dual fonction protein, and also provided is the use of said dual-fonction protein in the preparation of a biological substance for treating type 2 diabetes, obesity, dyslipidemia, fatty liver disease and/or metabolic syndrome. The dual-fonction protein provided in the present disclosure can synergistically regulate blood glucose and lipid levels in vivo, and satisfy multiple requirements for patients with type 2 diabetes such as lowering blood glucose, relieving hepatic steatosis, reducing body weight and improving metabolic disorders of circulating lipids.

Description

DUAL-FUNCTION PROTEIN FOR LIPID AND BLOOD GLUCOSE
REGULATION
The present disclosure relates to a GLP-1-FGF21 dual-function protein and related pharmaceutical combination, and also relates to the use of said dual-function protein for preparing a medicament for treating type 2 diabetes, obesity, hyperlipidaemia, fatty liver disease and/or metabolic syndrome, and treatment of these diseases using the medicament.
Glucagon-like peptide-1 (GLP-1) is an endocrine peptide consisting of 36 amino acids secreted by mammalian intestinal L cells, and stimulates insulin secretion from pancreatic beta cells in a glucose dependent manner by binding to and activating GLP-1 receptor (GLP-1R), inhibits glucagon release from pancreatic alpha cells to maintain normal glucose level.
In addition, it inhibits gastrointestinal movement and suppresses appetite (Knudsen LB, J
Med Chem, 2004, 4128-4134). Native human GLP-1 is easily inactivated in vivo by dipeptidyl peptidase IV (DDP-IV), and has a short circulating half-life.
Exendin-4 is isolated from the saliva of toxic lizards from South Africa and has 39 amino acids with 53%
homology to human GLP-1 and exerts a similar biological activity. Compared with the human GLP-1, Gly replaces Ala at the second position of N-telininus of Exendin-4 which enhances resistance of the peptide to the protcolytic degradation induced by DDP-IV and extends the circulating half-life in vivo. Exendin-4 has a special Trap-cage structure at the C-terminus, such that its binding affinity with GLP-1 receptor is significantly higher than that of human GLP-1 (Neidigh JW et al., Biochemistry, 2001, 40:13188-13200).
At the equimolar concentration, Exendin-4 exhibits a stronger effect on promoting insulin release from pancreatic beta cells. Although there is slight difference in structure and amino sequence, both the blood glucose metabolic regulators have been marketed, wherein most predominant ones are Liraglutide of Novo Nordisk and Exenatide ofAstraZeneca.
Treatment with Liraglutide or Exenatide 2-3 times daily can effectively control the blood glucose level Date Recue/Date Received 2020-07-02 in type 2 diabetes patients. However, the high injection frequency results in high cost and poor clinical compliance for patients. In order to prolong the in vivo half-life and bioavailability of GLP-1 and Exendin-4 analogs, Fc or HSA fusion technologies have been used for developing long-acting drugs. At present, marketed products are Dulaglutide of Eli Lilly and Albiglutide of GlaxoSmithKline. The most extensively used one in clinic is Dulaglutide, a GLP-1 hIgG4 Fc fusion protein (Dulaglutide), wherein its average half-life is up to 90 hours (Chinese patent CN 1802386 B), its clinical indication is type
2 diabetes, and the recommended dose regimen is subcutaneous injection once a week. Clinical study showed that Dulaglutide could effectively control postprandial blood glucose and glycosylated hemoglobin of diabetic patients, and lower the body weight of obese patients by inhibiting appetite. However, varying degrees of gastrointestinal adverse effects were observed. Epidemiological investigation showed that the large percentage of patients with type 2 diabetes accompanied with nonalcoholic fatty liver disease and lipid metabolism disorder (Radaelli MG et al., J Endocrinol Invest, 2017, s40618). However, no clinical study demonstrated that human GLP-1 or Exendin-4 analogs have the effect of treating fatty liver and hyperlipidaemia independent of weight loss (Petit JIM, Diabetes Metab, 2017, 43, 2S28-2S33). Therefore, GLP-1 products cannot completely satisfy all clinical needs for patients with type 2 diabetes.
The family of fibroblast growth factors (FGFs) has 22 members and 7 subfamilies, wherein the FGF19 subfamily exerts physiological activity in an endocrine manner, involves the regulation and control of energy and cholic acid homeostasis, glucose and lipid metabolism, and phosphate and vitamin D homeostasis (Moore DD et al., Science, 2007, 316:1436-1438 and Beenken et al., Nature Reviews Drug Discover, 2009, 8:235).
FGF21 is a member of FGF19 subfamily, and has 181 amino acids. The C-terminus of FGF21 binds first to a co-factor 13-Klotho transmembrane protein, induces FGFR binding to the N-Date Recue/Date Received 2020-07-02 terminus of FGF21, then forms a stable FGF21/13-Klotho/FGFR complex, which trigger subsequent signaling pathway in vivo (Yie J et al., FEBS Lett, 2009, 583(1):19-24 and Micanovic R et al., J Cell Physiol, 2009, 219(2): 227-234). Under physiological conditions, FGF21 is to promote glucose utilization independent of insulin (Kharitonenkov A et al., J
Clin Invest, 2005, 115(6): 1627-1635), to enhance insulin sensitization (Duthchak PA et al., Cell, 2012, 148, 387-393), to inhibit de novo lipogenesis and promote the fatty acid (3-oxidation in liver, to decrease serum triglyceride level (Xu J et al., Diabetes, 2009, 58, 250-259). In addition, FGF21 could decrease total cholesterol and low density lipoprotein-cholesterol contents in serum by inhibiting liver SREBP-2 synthesis to relieve hypercholesteremia (Lin Z et al., Circulation, 2015, 131, 1861-1871).
In conclusion, FGF21 exerts multiple regulatory functions on metabolic diseases, such as obesity, type 2 diabetes, nonalcoholic fatty liver and hyperlipidaemia.
Meanwhile, FGF21 is the only discovered member without any mitogenic effect in this superfamily, which greatly reduces potential carcinogenicity risk in clinical applications (Wu X
et al., Proc Natl Acad Sci USA, 2010, 170: 14158-14163). However, due to its unstable physicochemical property, native FGF21 does not possess druggability so far due to the following reasons: (1) native FGF21 protein has pool stability and is easily degraded by proteases in vivo; (2) FGF21 conformation is unstable with ease of aggregation, which increases difficulty in scale-up production; (3) native FGF21 has short circulating half-life, about 0.5-1 h in mice and 2-3 h in Cynomolgus monkeys (Kharitonenkov A et al., J Clin Invest, 2005, 115: 1627-1635). Various long-acting protein-engineering technologies are commonly used for prolonging the in vivo half-life of recombinant FGF21. For example, conjugation of FGF21 and PEG molecule increases the molecular weight, lowers the glomerular filtration rate, and prolongs the in vivo retention time (see WO 2005/091944, WO 2006/050247, WO
2008/121563 and WO 2012/066075); FGF21 fuses to long chain fatty acid (which can binds
3 Date Recue/Date Received 2020-07-02 to serum albumin) (see WO 2010/084169 and WO 2012/010553); preparation of an agonist antibody which specifically binds to FGFR or FGFR/13-klotho complex to mimic the mechanism of FGF21, and to activate FGF/FGFR signaling pathway (see WO
2011/071783, WO 2011/130417, WO 2012/158704 and WO 2012/170438); FGF21 fuses to Fc fragment to improve half-life (see WO 2004/110472, WO 2005/113606, WO 2009/149171, WO
2010/042747, WO 2010/129503, WO 2010/129600, WO 2013/049247, WO 2013/188181 and WO 2016/114633). At present, there is no marketed drug of long-acting FGF21 protein, but there are three long-acting FGF21 candidates in clinic trials, LY2405319 of Eli Lilly, PF-05231023 of Pfizer and BMS986036 of Bristol-Myers Squibb. In the clinical trials, for patients with type 2 diabetes, LY2405319 and PF-05231023 had weight loss effect and decreased serum TG level, but had no positive therapeutic effect on blood glucose (Gaich G
et al., Cell Metab, 2013, 18:333-340 and Dong JQ et al., Br J Clin Pharmacol, 2015, 80-1051-1063). BM5986036 exhibited a good therapeutic effect on nonalcoholic fatty liver, but there was no experimental study on blood glucose control for patients with type 2 diabetes.
Above-mentioned results showed that although the use of long-acting FGF21 protein alone can exert many pharmacodynamic activities such as in body weight, nonalcoholic fatty liver and hyperlipidaemia. However, it cannot satisfy the blood glucose control requirement which is crucial in the treatment of patients with type 2 diabetes.
Recently, some studies reported that the combination of GLP-1 and FGF21 has a synergistic effect on blood glucose control. For example, CN 102802657 A
disclosed that the combination of GLP-1 and FGF21 can synergistically lower the blood glucose level in db/db mice. However, the combined usage of drugs not only increases the administration frequency for patients and reduces the patient compliance, but also greatly increases treatment costs. In addition, a dual-function protein prepared by fusing GLP-1 and FGF21 was also reported. In order to solve the issue that FGF21 is easily degraded in vivo, scientists
4 Date Recue/Date Received 2020-07-02 generally introduce point mutations in native FGF21 molecule, but this inevitably increases the potential immunogenicity of the dual-function protein (WO 2017/074123 and CN
104024273 B). Furthermore, the reported synergistic effect of FGF21 and GLP-1 generally exhibited in terms of blood glucose control, but their synergistic effects in terms of other metabolic diseases, such as obesity, nonalcoholic fatty liver and lipid metabolism disorder are not investigated by comparing with marketed long-acting GLP-1 analogs. The reasons for lack of above-mentioned investigations may comprise the following: (1) neither native GLP-1 nor FGF21 is stable in vivo, and defection of structural integrity and stability in any molecule will eliminate the synergistic effect; (2) in the process of fusion of GLP-1 and FGF21 into a single protein, their respective three-dimensional conformation needs to be maintained to the maximum extent for preventing mutual interference, such that the functional synergy will be achieved, and this should be carefully considered when designing the molecule; (3) the functions of both GLP-1 and FGF21 depend on binding to their respective receptors, and it needs to be confirmed by a number of in vitro and in vivo experiments to clarify the conditions under which the dynamic equilibrium can be achieved among them. So far, there is no such report in published patents or other non-patent documents.
In conclusion, if a GLP-1-FGF21 dual-function protein drug that has enhanced stability, prolonged half-life and low immunogenicity can be developed in the art, then the multiple requirements of patients with type 2 diabetes for reducing blood glucose, relieving hepatic steatosis, reducing body weight and improving metabolic disorders of circulating lipids can be met.
Summary of the Disclosure
5 Date Recue/Date Received 2020-07-02 The present disclosure provides a dual-function protein having a synergistic effect in terms of blood glucose and lipid regulations and comprising human GLP-1 analog and human FGF21, the preparation method therefor and the use thereof. The present disclosure solves the issues such as the defects relating to unstable structure and short in vivo half-life of native GLP-1 or FGF21, retains the strong hypoglycemic effect of GLP-1 and the physiological effects of FGF21 on insulin sensitization, weight loss, fatty liver and hypercholesteremia treatment, and relieves gastrointestinal adverse effects caused by GLP-1 to some extent.
In one embodiment of the present disclosure, a dual-function protein can synergistically regulate blood glucose and lipids, wherein said dual-function protein comprises human glucagon-like peptide-1 analog (abbreviated as GLP-1 analog hereafter), linker peptide 1 (abbreviated as L1 hereafter), human fibroblast growth factor 21 (abbreviated as FGF21 hereafter), linker peptide 2 (abbreviated as L2 hereafter) and human immunoglobulin Fc fragment (abbreviated as Fc fragment hereafter) sequentially from the N-terminus to the C-terminus; wherein the linker peptide 1 comprises a flexible peptide; the linker peptide 2 comprises a flexible peptide and rigid peptide, the rigid peptide comprises at least 1 rigid unit, and the rigid unit comprises carboxyl terminal peptide of human chorionic gonadotropin 13-subunit or a truncated sequence thereof In one embodiment of the present disclosure, a dual-function protein can synergistically regulate blood glucose and lipids, wherein said dual-function protein consists of sequentially human glucagon-like peptide-1 analog (abbreviated as GLP-1 analog hereafter), linker peptide 1 (abbreviated as Li hereafter), human fibroblast growth factor 21 (abbreviated as FGF21 hereafter), linker peptide 2 (abbreviated as L2 hereafter) and human immunoglobulin Fc fragment (abbreviated as Fc fragment hereafter) from the N-terminus to the C-terminus;
wherein the linker peptide 1 consists of a flexible peptide; the linker peptide 2 consists of a
6 Date Recue/Date Received 2020-07-02 flexible peptide and rigid peptide, the rigid peptide consists of at least 1 rigid unit, and the rigid unit comprises carboxyl terminal peptide of human chorionic gonadotropin 13-subunit or a truncated sequence thereof.
In the present disclosure, said "human GLP-1 analog" refers to an analog, fusion peptide, or derivative which are obtained by substituting, deleting or adding one or more amino acid residues on the amino acid sequence of human GLP-1 (as shown in SEQ
ID NO:
1) and maintains human GLP-1 activity. For example, said GLP-1 analog comprises but is not limited to the amino acid sequences as shown in SEQ ID NO: 2, 3, 4 or 5 in the sequence listing. In at least one embodiment of the present disclosure, said GLP-1 analog is shown in SEQ ID NO: 2, and in another embodiment, said GLP-1 analog is shown in SEQ ID
NO: 5.
In the present disclosure, said "linker peptide 1 (L1)" is a short peptide between GLP-1 analog and FGF21 and has connecting function. In at least one embodiment, said linker peptide 1 is non-immunogenic, and generates enough distal distance between GLP-1 analog and FGF21, such that minimal steric hindrance effect is present, which does not affect or not affect severely correct folding and spatial conformation of GLP-1 analog and FGF21. A
person skilled in the art can design linker peptides according to conventional methods in the art. In at least one embodiment, a flexible peptide comprising 2 or more amino acids is used, and the amino acids are selected from the following amino acids: Gly(G), Ser(S), Ala(A) and Thr(T); in at least one embodiment, said linker peptide 1 comprises G and S residues.
The length of the linker peptide is very important for the activity of the dual-function protein, and in at least one embodiment, the linker peptide consists of 5-30 amino acids. In a at least one embodiment of the present disclosure, the amino acid sequence of said linker peptide 1 is GGGGGGGSGGGGSGGGGS.
7 Date recue / Date received 202 1-1 1-25 In the present disclosure, said "FGF21" comprises the sequence as shown in SEQ
ID
NO: 6 in which the secreting leader signal of amino acid position 1-28 is deleted; or comprises the isoform sequence of SEQ ID NO: 6 in which the secreting leader signal of amino acid position 1-28 is deleted and which has G141S or L174P substitution.
In a at least one embodiment of the present disclosure, said FGF21 comprises the amino acid sequence as shown in SEQ ID NO: 6 in which the secreting leader signal of amino acid position 1-28 is deleted and has L174P substitution.
In the present disclosure, said "linker peptide 2 (L2)" is a short peptide between FGF21 and Fc fragment and having connect function. Said linker peptide consists of a flexible peptide and a rigid peptide, wherein said flexible peptide comprises 2 or more amino acid residues which are selected from Gly(G), Ser(S), Ala(A) and Thr(T). In at least one embodiment, said flexible peptide comprises G and S residues. With regard to the present disclosure, preferably, the general structural formula of the amino acid composition of said flexible peptide is (GS)a(GGS)b(GGGS)c(GGGGS)d, wherein a, b, c and d are integers greater than or equal to 0, and a+b+c+d> 1.
In some embodiments of the present disclosure, said flexible peptide comprised in said L2 is selected from the following sequences:
(i) GGGGS;
(ii) GSGGGSGGGGSGGGGS;
(iii) GSGGGGSGGGGSGGGGSGGGGSGGGGS;
(iv) GGGGSGGGGSGGGGSGGGGSGGGGSGGGGSGGGGS;
(v) GGGSGGGSGGGSGGGSGGGS;
(vi) GGSGGSGGSGGS.
In the present disclosure, said rigid peptide constituting said linker peptide 2 (L2) consists of one or more rigid units, and said rigid units are selected from a full-length or
8 Date Recue/Date Received 2020-07-02 truncated sequence consisting of carboxyl terminal amino acids 113 to 145 of human chorionic gonadotropin I3-subunit (known as CTP rigid unit hereafter);
specifically, said CTP
rigid unit comprises the amino acid sequence as shown in SEQ ID NO: 7 or the truncated sequences thereof.
In at least one embodiment, said CTP rigid unit comprises at least 2 glycosylation sites;
for example, in one at least one embodiment of the present disclosure, said CTP rigid unit comprises 2 glycosylation sites, for example, said CTP rigid unit comprises 10 amino acids of N-terminus of SEQ ID NO: 7, i.e. SSSS*KAPPPS*; or said CTP rigid unit comprises 14 amino acids of C-terminus of SEQ ID NO: 7, i.e. S*RLPGPS*DTPILPQ; for another example, in another embodiment, said CTP rigid unit comprises 3 glycosylation sites, for example, said CTP rigid unit comprises 16 amino acids of N-terminus of SEQ ID
NO: 7, i.e.
SSSS*KAPPPS*LPSPS*R; for another example, in another embodiment, said CTP
rigid unit comprises 4 glycosylation sites, for example, said CTP rigid unit comprises 28, 29, 30, 31,32 or 33 amino acids and starts at positions 113, 114, 115, 116, 117 or 118 and terminates at position 145 of human chorionic gonadotropin 13-subunit. Specifically, said CTP rigid unit comprises 28 amino acids of N-terminus of SEQ ID NO: 7, i.e.
SSSS*KAPPPS*LPSPS*RLPGPS*DTPILPQ. In the present disclosure, * represents glycosylation sites. All possibilities represent independent embodiments of the present disclosure.
In some embodiments, the CTP rigid unit comprised in L2 of the present disclosure can preferably comprise one of the following sequences:
(i) CTP': SSSSKAPPPSLPSPSRLPGPSDTPILPQ;
(ii) CTP2: PRFQDSSSSKAPPPSLPSPSRLPGPSDTPILPQ;
(iii) CTP3: SSSSKAPPPS;
(iv) CTP4: SRLPGPSDTPILPQ;
9 Date Recue/Date Received 2020-07-02 (v) CTP5: SSSSKAPPPSLPSPSR.
In some embodiments, the CTP rigid unit provided in the present disclosure has at least 70% homology to the native CTP amino acid sequence; In some embodiments, the CTP rigid unit provided in the present disclosure has at least 80% homology to the native CTP amino acid sequence; In some embodiments, the CTP rigid unit provided in the present disclosure has at least 90% homology to the native CTP amino acid sequence; In some embodiments, the CTP rigid unit provided in the present disclosure has at least 95%
homology to the native CTP amino acid sequence.
In some embodiments of the present disclosure, L2 comprises 2, 3, 4 or 5 above-mentioned CTP rigid units. In some embodiments of the present disclosure, L2 of said dual-function protein comprises 2 CTP3 rigid unit: SSSSKAPPPSSSSSKAPPPS (CTP3-CTP3, or represented as (CTP3)2).
In the present disclosure, said "Fe fragment" is selected from the Fc fragments of human immunoglobulins IgG, IgM, IgA and variants thereof; in at least one embodiment, is selected from the Fc fragments of human IgGl, IgG2, IgG3 or IgG4 and variants thereof, wherein said human IgG Fc fragment (represented as vFc) comprises at least one amino acid modification located in wild type human IgG Fc, and the Fc variants have non-lytic characteristics, and show an extremely minimal Fc-mediated effector functions (ADCC and CDC functions) and/or enhanced binding affinity with FcRn receptor; most preferably, human IgG Fc variant is selected from the group of:
(i) vFcyl: hinge. CH2 and CH3 regions of human IgG1 containing Leu234Va1, Leu235Ala and Pro331Ser mutations (the amino acid sequence as shown in SEQ ID
NO: 8);
(ii) vFcy2-1: hinge. CH2 and CH3 regions of human IgG2 containing Pro331Ser mutation (the amino acid sequence as shown in SEQ ID NO: 9);
Date Recue/Date Received 2020-07-02 (iii) vFcy2-2: hinge, CH2 and CH3 regions of human IgG2 containing Thr250Gln and Met428Leu mutations (the amino acid sequence as shown in SEQ ID NO: 10);
(iv) vFcy2-3: hinge, CH2 and CH3 regions of human IgG2 containing Pro331Ser, Thr250Gln and Met428Leu mutations (the amino acid sequence as shown in SEQ ID
NO:
11).
(v) vFcy4: hinge, CH2 and CH3 regions of human IgG4 containing Ser228Pro and Leu235Ala mutations (the amino acid sequence as shown in SEQ ID NO: 12).
The Fc variants provided by the present disclosure comprises, but is not limited to above 5 variants of (i) to (v), and also can be the combination or overlap of functional variants among same IgG subtypes, for example, the variant of above-mentioned (iv) is a new combination variant of IgG2 Fc obtained by overlapping the mutation sites in (ii) and (iii).
The Fc variant (vFc) in the dual-function protein of the present disclosure contains human IgG, such as the hinge region and CH2 and CH3 regions of human IgGl, IgG2 and IgG4. Such CH2 region contains amino acid mutations at positions 228, 234, 235 and 331 (determined by EU numbering system). It is believed that these amino acid mutations can reduce Fc effector function. Human IgG2 does not bind to FcyR, but shows a very weak complement activity. The complement activity of Fcy2 variant having Pro331Ser mutation should be lower than that of native Fcy2, and is still an FcyR non-binder.
IgG4 Fc has some defects in activation of complement cascade, and its binding affinity with FcyR is lower than that of IgG1 by about one order of magnitude. Compared with native Fcy4, the Fcy4 variant having Ser228Pro and Leu235Ala mutations should show the minimal effector function.
Compared with native Fcyl, Fcyl having Leu234Val, Leu235Ala and Pro331Ser mutations also shows a reduced effector function. These Fc variants are more suitable for preparing a dual-function protein of FGF21 and analogs thereof than native human IgG Fc.
However, positions 250 and 428 (positions determined by EU numbering system) contain amino acid Date Recue/Date Received 2020-07-02 substitutions, such that the binding affinity of Fe region with neonate receptor FcRn is increased, thus further prolonging the half-life (Paul R et al., J Biol Chem, 2004, 279:6213-6216); the combination or overlap of above-mentioned two types of functional variants obtains new variants which have reduced effector function and prolonged half-life. The Fc variants of the present disclosure comprises, but is not limited to above-mentioned mutations;
the substitutions at other sites can also be introduced, such that Fc has a reduced effector function and/or enhanced affinity with FcRn receptor, at the same time, without causing reduced functions/activities of Fc variants or adverse conformational changes, and see Shields RL et al., J Biol Chem, 2001, 276(9):6591-604 for common mutation sites.
In one at least one embodiment of the present disclosure, the amino acid sequence of said dual-function protein is shown in SEQ ID NO: 13. In another at least one embodiment of the present disclosure, the amino acid sequence of said dual-function protein is shown in SEQ ID NO: 15.
The dual-function protein of the present disclosure is glycosylated;
preferably, said dual-function protein is glycosylated by being expressed in mammalian cells;
in at least one embodiment, said dual-function protein is glycosylated by being expressed in Chinese hamster ovary cells.
According to another embodiment of the present disclosure, provided is a DNA
encoding the above-mentioned dual function protein. In one at least one embodiment of the present disclosure, the DNA sequence encoding said dual-function protein is shown in SEQ
ID NO: 14.
According to still another embodiment of the present disclosure, provided is a vector.
The vector comprises the above-mentioned DNA.

Date Recue/Date Received 2020-07-02 According to still another embodiment of the present disclosure, provided is a host cell.
The host cell comprises the above-mentioned vector, or is transfected with the above-mentioned vector.
In a particular embodiment of the present disclosure, the host cell is a CHO-derived cell strain DXB-11.
According to still another embodiment of the present disclosure, provided is a pharmaceutical composition. The pharmaceutical composition comprises a pharmaceutically acceptable carrier, excipient or diluent, and an effective amount of the above-mentioned synergistic dual function protein.
According to another embodiment of the present disclosure, provided is a method for preparing or producing said dual-function protein from mammalian cell lines (such as a CHO-derived cell line), comprising following steps:
(a) introducing a DNA encoding the above-mentioned dual-function protein into a mammalian cell;
(b) screening a high-yield cell strain expressing more than 20 jig/b6 cells within a period of every 24 hours in the growth medium thereof from step (a);
(c) culturing the screened cell strain in step (b);
(d) harvesting the fermentation broth obtained from step (c), and purifying the dual function protein.
In at least one embodiment, said mammalian cell in step (a) is CHO cell; in at least one embodiment, said mammalian cell in step (a) is CHO-derived cell line DXB-11.
According to still another embodiment of the present disclosure, provided is the use of said dual-function protein in the preparation of a drug for treating FGF21 related diseases and GLP-1 related diseases, and other metabolic, endocrinic and cardiovascular diseases, comprising obesity, types 1 and 2 diabetes, pancreatitis, dyslipidemia, nonalcoholic fatty Date Recue/Date Received 2020-07-02 liver disease, nonalcoholic steatohepatitis, insulin tolerance, hyperinsulinemia, glucose intolerance, hyperglycemia, metabolic syndrome, acute myocardial infarction, high blood pressure, cardiovascular disease, atherosclerosis, peripheral arterial disease, stroke, cardiac failure, coronary heart disease, nephropathy, diabetic complication, neuropathy, gastroparesis, and conditions associated with the severe inactivation mutations of insulin receptor; preferably, said diseases comprise obesity, types 1 and 2 diabetes, dyslipidemia, nonalcoholic fatty liver disease, nonalcoholic steatohepatitis and metabolic syndrome.
Compared with existing products, the dual-function protein of the present disclosure has many advantages, which are demonstrated in detail by using, e.g., dual-function protein FP4I-2 of the present disclosure:
1. The half-life in vivo is prolonged, and blood glucose-lowering activity in vivo is maintained for a longer period of time. The glucose tolerance test performed on C57BL/6 mice shows that at 144 h after a single dose of FP4I-2, FP4I-2 still exhibits a good ability for promoting glucose utilization, which is better than the commercially available GLP-1 analog Liraglutide and Exenatide and native FGF21. Compared with GLP-1-Fc fusion protein Dulaglutide, FP4I-2 exhibits a more excellent blood glucose control effect in the type 2 diabetes mice.
2. Improved safety and tolerability. Dulaglutide induces severe gastrointestinal adverse effects after the first administration. In db/db mice, the initial 24 hour food intake of the mice after first administration of FP4I-2 is significantly elevated relative to that of mice administrated with Dulaglutide, which shows that the dual-function protein FP4I-2 can effectively relieve appetite inhibition induced by gastrointestinal adverse effects.
3. Improved therapeutic effects of dual-function protein on fatty liver.
Compared with Dulaglutide, FP4I-2 can significantly reduce the liver mass of db/db mice, improve liver function, and the mechanism does not completely depend on the appetite inhibition Date Recue/Date Received 2020-07-02 related to GLP-lanalog, demonstrating the physiological activity of FGF21.
Relative to native FGF21, the in vivo half-life of FP4I-2 is significantly prolonged. In the animal model, the dose frequency of FP4I-2 is twice per week, which will improve the clinical feasibility.
4. The C-terminal sequence of FGF21 is crucial to its activity, most dual-proteins reported in the prior art uses FGF21 N-terminal fusion, and free C-terminus is beneficial to maintain activity; however, the C-terminus of FGF21 also contains various protease hydrolysis sites, and is very easily degraded; exposed intact C-terminus is more easily attacked by protease and degraded. In order to overcome this problem, the prior art avoids using native FGF21, but introducing corresponding mutations to improve its stability, however, this inevitably increases the potential immunogenicity of dual-function proteins.
In contrast, the dual-function protein constructed in the present disclosure uses native FGF21, and has an Fc fragment connected at its C-terminus. The dual-function protein provided by the present disclosure not only has a significantly prolonged in vivo half-life in circulation, but also has a synergistic effect in terms of blood glucose and lipid regulations, which suggests that the constructed dual-function protein well maintains the properties of this two active molecules, and has good stability. This benefits result from the new type of linker peptide among FGF21 and Fc variants; the linker peptide consists of a flexible peptide and a rigid peptide, and the CTP rigid unit contains multiple 0-carbohydrate side chains, can forms a relatively stable three-dimensional conformation, which can effectively separate FGF21 and Fc, thereby lowers the steric hindrance effect caused by Fc fragment to the utmost extent and keep relatively good FGF21 biological activity. In addition, carbohydrate side chain of CTP rigid unit can mask the enzymolysis site of FGF21. The protective effect lowers the sensibility of enzymolysis to proteases and achieves the purpose for protein stability.
Date Recue/Date Received 2020-07-02 5. Mutations on Fc only retains the long half-life property in circulation of Fc, reducing or eliminating ADCC and CDC effects (such as P33 1S), thus increases the safety of drug use. In addition, Fc variants (such as T250Q/M428L) having an enhanced binding affinity with neonate receptor (FcRn) can further prolong the half-life of dual function protein.
DETAILED DESCRIPTION OF THE DISCLOSURE
Human GLP-1 analog The term "human GLP-1 analog" used herein refers to an analog, fusion peptide, and derivative which are obtained by substituting, deleting or adding one or more amino acid residues on the amino acid sequence of human GLP-1 (as shown in SEQ ID NO: 1) and maintain human GLP-1 activity. For example, said human GLP-1 analog comprises but are not limited to the amino acid sequences as shown in SEQ ID NO: 2, 3, 4 or 5 in the sequence listing.
Human FGF21 The term "human FGF21" used herein refers to a wild type human FGF21 polypeptide.
The sequence of the wild type FGF21 protein can be obtained from UNIPROT
database, and the accession number is Q9NSA1. The precursor protein consists of 209 amino acids, comprising a signal peptide (amino acids 1-28) and a mature protein (amino acids 29-209).
US 2001012628 Al teaches the isoform or allelic form of the wild type FGF21 having the substitution from Leu to Pro in the mature protein (in the present disclosure, as shown in positions 47-227 of SEQ ID NO: 13); another isoform of wild type FGF21 having the substitution from Gly to Ser (Gly at the position 141 of SEQ ID NO: 6 is substituted or replaced by Ser).

Date Recue/Date Received 2020-07-02 WO 2003/011213 teaches another isoform (see SEQ ID NO: 2 disclosed in WO
2003/011213, which has a signal peptide of 27 amino acid residues) having a relatively short signal peptide (in the present disclosure, Leu at position 23 of SEQ ID NO: 6 deleted).
In the present disclosure, the wild type FGF21 comprises SEQ ID NO: 6 and the sequence of the mature protein portion (amino acids 29-209) of the isoform having L 174P
or G141S substitutions after removing the leader peptide; in addition, also comprised is the full-length sequence of the precursor protein with the above-mentioned 27 or 28 amino acid signal peptide added before those above sequences.
hCG-I3 carboxyl terminal peptide (CTP) CTP is a short carboxyl terminal peptide of human chorionic gonadotropin (hCG) (3-subunit. Four reproduction-related polypeptide hormones follicle-stimulating hormone (FSH), luteinizing hotinone (LH), thyroid stimulating hormone (TSH) and chorionic gonadotropin (hCG) contain the same alpha subunit and the different specific beta subunit from each other. The in vivo half-life of hCG, compared with other three hormones, is prolonged, which is mainly due to the specific carboxyl terminal peptide (CTP) of its beta subunit (Fares FA et al., Proc Natl Acad Sci USA, 1992, 89: 4304-4308). Native CTP
contains 37 amino acid residues, has 4 0-glycosylation sites, and has sialic acid residue at the terminus. Negative charged, highly sialylated CTP can resist the clearance of kidneys on same, thus prolong the in vivo half-life of same. However, the inventors herein creatively connects a rigid peptide including at least one CTP rigid unit with a flexible peptide with a suitable length, collectively as linker peptide 2 for connecting FGF21 and Fc fragment.
N-terminal and C-terminal sequences of FGF21 are crucial to the functions of FGF21.
The spatial conformation of FGF21 is complex and fragile, such that FGF21 has a poor stability, is easily degraded and aggregated; if FGF21 is fused to a ligand, the steric hindrance effect will interfere with the correct folding of FGF21, making the activity of FGF21 Date Recue/Date Received 2020-07-02 significantly lowered or even lost, or more easily to generate a polymer.
Adding CTP rigid units between FGF21 and Fc variants is equivalent to adding a section of rigid linker peptide.
In one embodiment, it ensures that the FGF21 fused at N-terminus will not affect the binding site of Fc variants and FcRn, thereby not affecting the half-life; in addition, the Protein A
binding site of Fc is very important for the purification step in the preparation process, and connecting CTP rigid units ensures that N-terminus fused FGF21 also will not "mask" its binding site with protein A. In another embodiment, the addition of CTP rigid units also makes Fc fragments with about 25 KD not interfere with the correct folding of N-terminus fused FGF21, and not cause the biological activity/functions of FGF21 lowered or lost. This may be interpreted that a CTP rigid polypeptide with multiple carbohydrate side chains, which, relative to the random coil of (GGGGS)n of flexible linker peptides, can form a stable three-dimensional conformation, and this "barrier- effect promotes FGF21 and Fc fragment to fold and form a correct three-dimensional conformation independently, thus not affecting their respective biological activity. In another embodiment, the protective effect of carbohydrate side chain of CTP can lower the sensibility of linker peptides to proteases, such that the dual-function protein is not easily degraded in the linker region. In addition, CTP is derived from native human hCG, has no immunogenicity, therefore, relative to non-native encoded amino acid sequence, is more suitable for being used as a linker peptide.
IgG Fc variant Non-lytic Fc variants Fc elements are derived from the constant region Fc of immunoglobulin IgG, and have an important effect in immune defense for eliminating pathogens. The effector function of IgG mediated by Fc is exerted via two mechanisms: (1) binding to the Fc receptors (FcyRs) on the cell surface, digesting pathogens by phagocytosis or lysis, or by killer cells through antibody-dependent cytotoxic (ADCC) pathway, or (2) binding to Clq of the first Date Recue/Date Received 2020-07-02 complement component Cl, triggering the complement dependent cytotoxic (CDC) pathway, thereby lysing pathogens. Among four human IgG subtypes, IgG1 and IgG3 can effectively bind to FeyRs, the binding affinity of IgG4 with FcyRs is relatively low, and the binding of IgG2 with FeyRs is too low to be determined, and therefore, human IgG2 almost has no ADCC effect. In addition, human IgG1 and IgG3 also can effectively bind to C 1 q, thereby activating the complement cascade. The binding of human IgG2 with Clq is relatively weak, and IgG4 does not bind with C 1 q (Jefferis R et al., Immunol Rev, 1998, 163:
59-76), and therefore, the CDC effect of human IgG2 is also weak. There is no native IgG
subtype which is very suitable for constructing GLP-1-FGF21 dual function protein. In order to obtain a non-lytic Fc without effector function, the most effective method is performing mutation modification on the complement and receptor binding domain of Fc fragment, regulating the binding affinity of Fc with related receptors, reducing or eliminating ADCC
and CDC effects, only retaining the long half-life property of Fc in circulation, and not generating cytotoxicity.
For additional mutation sites comprised in non-lytic Fc variants, one can refer to RL et al., J
Biol Chem, 2001, 276(9):6591-604 or Chinese invention patent CN
201280031137.2.
Fc variants having an enhanced binding affinity with neonate receptor (FcRn) The plasma half-life of IgG depends on its binding with FcRn, and generally, they bind at pH 6.0, and dissociate at pH 7.4 (plasma pH). By the study of the binding site of the two, the binding site on IgG with FcRn is modified, such that the binding ability thereof is increased at pH 6Ø It is proven that the mutations of some residues in human Fey domain which is important for binding with FcRn can increase the serum half-life. It has been reported that the mutations of T250, M252, S254, T256, V308, E380, M428 and N434 can increase or reduce FcRn binding affinity (Roopenian et al., Nat Rview Immunology, 2007, 7:715-725). South Korea patent no. KR 10-1027427 discloses Trastuzumab (Herceptin, Genentech) variants with an increased FcRn binding affinity, and these variants comprise Date Recue/Date Received 2020-07-02 one or more amino acid modifications selected from 257C, 257M, 257L, 257N, 257Y, 279Q, 279Y, 308F and 308Y. South Korea patent no. KR 2010-0099179 provides bevacizumab (avastin, Genentech) variants, and these variants comprise amino acid modifications of N4345, M252Y/M428L, M252Y/N4345 and M428L/N4345, and show an increased half-life in vivo. In addition, Hinton et al. also find that 2 mutants of T250Q and M428L can increase the binding with FcRn by 3 and 7 times respectively. Mutating the 2 sites at the same time, the binding will be increased by 28 times. In rhesus monkeys, the mutants of M428L or T250QM/428L show the plasma half-life in vivo is increased by 2 times (Paul R.
Hinton et al., J Immunol, 2006, 176:346-356). For additional mutation sites comprised in Fc variants having an enhanced binding affinity with neonate receptor (FcRn), one can refer to Chinese invention patent CN 201280066663.2. In addition, in some studies performing T250Q/M428L mutations on the Fc fragment of five humanized antibodies, the interaction between Fc and FcRn is improved, and in the subsequent in vivo pharmacokinetic test, it finds that using subcutaneous injection administration, the pharmacokinetic parameters of Fc mutation antibodies are improved compared with wild type antibodies, such as an increased in vivo exposure, lowered clearance rate and improved subcutaneous bioavailability (Datta-Mannan A et al., MAbs. Taylor & Francis, 2012, 4(2):
267-273).
Terms "FGF21-related conditions" and "GLP-1-related conditions" comprise obesity, types 1 and 2 diabetes, pancreatitis, dyslipidemia, nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, insulin tolerance, hyperinsulinemia, glucose intolerance, hyperglycemia, metabolic syndrome, acute myocardial infarction, high blood pressure, cardiovascular disease, atherosclerosis, peripheral arterial disease, stroke, cardiac failure, coronary heart disease, nephropathy, diabetic complication, neuropathy, gastroparesis, and conditions associated with the severe inactivation mutations of insulin receptor.
Date Recue/Date Received 2020-07-02 "Conditions associated with the severe inactivation mutations of insulin receptor"
describe the conditions of subjects with insulin receptor (or a direct downstream possible protein thereof) mutation, wherein said mutation results in a severe insulin tolerance, but generally no obesity which is common in type 2 diabetes. In many embodiments, subjects with these conditions exhibit the symptom complex of types 1 and 2 diabetes.
Therefore, the involved subjects are divided into several types according to the severity, comprising: type A diabetes resistance, type C insulin resistance (AKA HAIR-AN syndrome), Rabson-Mendenhall syndrome, Donohue's syndrome or Leprechaunism. These conditions are associated with a very high endogenous insulin level, and results in an elevated blood glucose level. Therefore, in the involved subjects, there are many clinical features associated with "insulin toxicity", wherein the clinical features comprise androgen excess, polycystic ovarian syndrome (PCOS), hirsutism and acanthosis nigricans (overgrowth of wrinkly skin and pigmentation).
"Diabetic complications" is the dysfunction of other tissue/organs of the body induced by chronic hyperglycemia, such as diabetic nephropathy, diabetic neuropathy, diabetic feet (foot ulcers and low blood circulation) and eye lesions (retinopathy).
Diabetes also increases the risks of heart disease and osteoarticular diseases. The other long-term complications of diabetes comprise skin, digestive, sexual function, teeth and gums disease.
"Metabolic syndrome (MS)" is the morbidness caused by abnormal metabolic parameters, comprising: (1) abdominal obesity or overweight; (2) atherosclerosis and dyslipidemia, such as hypertriglyceride and reduction in high density lipoprotein cholesterol (HDL-C); (3) hypertension; (4) insulin resistance and/or abnormal glucose tolerance. In some criteria, microalbuminuria, hyperuricemia, pro-inflammatory state (C-reactive protein) and pro-thrombogenesis state (increase in Fibrinogen and Plasminogen inhibitor-1) are also comprised.

Date Recue/Date Received 2020-07-02 "Dyslipidemia" is a lipoprotein metabolic disorder, comprising the oversynthesis or defect of lipoprotein. Dyslipidemia can exhibit as the elevated concentration of total cholesterol, low density lipoprotein (LDL) cholesterol and triglycerides, and the reduced concentration of high density lipoprotein (HDL) cholesterol.
"Nonalcoholic fatty liver disease (NAFLD)" is a liver disease which is not associated with abused alcohol consumption and is characterized in hepatocellular steatosis.
"Nonalcoholic steatohepatitis (NASH)" is a liver disease which is not associated with abused alcohol consumption and is characterized in hepatocellular steatosis accompanied by lobular inflammation and fibrosis.
"Atherosclerosis" is an angiopathy and is characterized in lipid deposits irregularly distributed on endangium of large and medium-sized arteries, which results in hemadostenosis, and eventually develops into fibrosis and calcification.
Brief Description of the Drawinks Fig. 1. shows the nucleotide sequence of dual-function protein FP4I-2 of SpeI/EcoRI
fragment in PCDNA3.1 expression vector according to the embodiments of the present disclosure and the deduced amino acid sequence, consisting of alpha 1 microglobulin leader peptide (1-19), GLP-1 analog (20-47), Li (48-65), FGF21 mature protein (66-246), L2 (247-301) and IgG2 Fc (302-524).
Fig. 2a. Reduced SDS-PAGE electrophoretogram of GLP-1-FGF21 dual-function protein FP41-2.
Fig. 2b. SEC-HPLC spectrogram of GLP-1-FGF21 dual-function protein FP4I-2.
Fig. 3a. Glucose tolerance test curve of GLP-1-FGF21 dual-function proteins and FP4I-2 16 h after a single injection (means SEM, n = 8).

Date Recue/Date Received 2020-07-02 Fig. 3b. Glucose tolerance test iAUC of GLP-1-FGF21 dual-function proteins and FP4I-2 16 h after a single injection (means SEM, n = 8); statistical difference symbols:
compared with the control group, *P < 0.05, and **P < 0.01.
Fig. 4a. Glucose tolerance test curve of GLP-1-FGF21 dual-function proteins and FP4I-2 96 h after a single injection (means SEM, n = 8).
Fig. 4b. Glucose tolerance test iAUC of GLP-1-FGF21 dual-function proteins and FP4I-2 96 h after a single injection (means SEM, n = 8); statistical difference symbols:
compared with the control group, *P < 0.05, and **P < 0.01.
Fig. 5a. Glucose tolerance test curve of GLP-1-FGF21 dual-function proteins and FP4I-2 144 h after a single injection (means SEM, n = 8).
Fig. 5b. Glucose tolerance test iAUC of GLP-1-FGF21 dual-function proteins and FP41-2 144 h after a single injection (means SEM, n = 8); statistical difference symbols:
compared with the control group, *P < 0.05, and **P < 0.01.
Fig. 6a. Glucose tolerance test curve of Exendin4-FGF21 dual-function proteins 3 16 h after a single injection (means SEM, n = 8).
Fig. 6b. Glucose tolerance test iAUC of Exendin4-FGF21 dual-function proteins 3 16 h after a single injection (means SEM, n = 8); statistical difference symbols: compared with the control group, *P < 0.05, and **P < 0.01; compared with the Dulaglutide group, #P
<0.05, and "P < 0.01.
Fig. 7a. Glucose tolerance test curve of Exendin4-FGF21 dual-function proteins 3 96 h after a single injection (means SEM, n = 8).
Fig. 7b. Glucose tolerance test iAUC of Exendin4-FGF21 dual-function proteins 3 96 h after a single injection (means SEM, n = 8); statistical difference symbols: compared with the control group, *P < 0.05, and **P < 0.01; compared with the Dulaglutide group, #P
<0.05, and "P < 0.01.

Date Recue/Date Received 2020-07-02 Fig. 8a. Glucose tolerance test curve of Exendin4-FGF21 dual-function proteins 3 144 h after a single injection (means SEM, n = 8).
Fig. 8b. Glucose tolerance test iAUC of Exendin4-FGF21 dual-function proteins 3 144 h after a single injection (means SEM, n = 8); statistical difference symbols:
compared with the control group, *P < 0.05, and **P < 0.01; compared with the Dulaglutide group, #P < 0.05, and 44P < 0.01.
Fig. 9. Effect of GLP-1-FGF21 dual-function proteins FP4I-1 and FP4I-2 on 24 h food intake in db/db mice after first administration (means SEM, n = 6);
statistical difference symbols: compared with the control group, *P < 0.05, and **P < 0.01; compared with the Dulaglutide group, #P < 0.05, and "P < 0.01.
Fig. 10. Effect of multiple administrations of GLP-1-FGF21 dual-function proteins FP4I-1 and FP4I-2 on glycated hemoglobin in db/db mice (means SEM, n = 6);
statistical difference symbols: compared with the control group, *P < 0.05, and **P <
0.01; compared with the Dulaglutide group, #P < 0.05, and "P < 0.01.
Fig. 11. Effect of multiple administrations of GLP-1-FGF21 dual-function proteins FP4I-1 and FP4I-2 on accumulative food intake in db/db mice (means SEM, n =
6);
statistical difference symbols: compared with the control group, *P < 0.05, and **P < 0.01;
compared with the Dulaglutide group, #P < 0.05, and 44P < 0.01.
Fig. 12. Effect of GLP-1-FGF21 dual-function proteins FP4I-1 and FP4I-2 on body weight in obese mice induced by high-fat diet (means SEM, n = 7);
statistical difference symbols: compared with the obesity control group, *P < 0.05, and **P < 0.01;
compared with the Dulaglutide group, #P < 0.05, and 44/' < 0.0/; compared with the FP4I-1 group, &P
<0.05, and "P < 0.01.
Fig. 13. Effect of GLP-1-FGF21 dual-function proteins FP4I-1 and FP4I-2 on liver mass in obese mice induced by high-fat diet (means SEM, n = 7); statistical difference Date Recue/Date Received 2020-07-02 symbols: compared with the obesity control group, *P < 0.05, and **P < 0.01;
compared with the Dulaglutide group, #P < 0.05, and "P < 0.01.
Fig. 14. Effect of GLP-1-FGF21 dual-function proteins FP4I-1 and FP4I-2 on liver triglyceride content in obese mice induced by high-fat diet (means SEM, n =
7); statistical difference symbols: compared with the obesity control group, *P < 0.05, and **P < 0.01;
compared with the Dulaglutide group, #P < 0.05, and 44P < 0.01.
Fig. 15. Effect of GLP-1-FGF21 dual-function proteins FP4I-1 and FP4I-2 on serum triglyceride in obese mice induced by high-fat diet (means SEM, n = 7);
statistical difference symbols: compared with the obesity control group, *P < 0.05, and **P < 0.01;
compared with the Dulaglutide group, #P < 0.05, and "P < 0.01; compared with the FP4I-1 group, &P < 0.05, and "P < 0.01.
Fig. 16. Effect of GLP-1-FGF21 dual-function proteins FP4I-1 and FP4I-2 on serum total cholesterol content in obese mice induced by high-fat diet (means SEM, n = 7);
statistical difference symbols: compared with the obesity control group, *P <
0.05, and **P
<0.01; compared with the Dulaglutide group, #P < 0.05, and "P < 0.01.
Fig. 17. Effect of GLP-1-FGF21 dual-function proteins FP4I-1 and FP4I-2 on serum low density lipoprotein cholesterol content in obese mice induced by high-fat diet (means SEM, n = 7); statistical difference symbols: compared with the obesity control group, *P <
0.05, and **P < 0.01; compared with the Dulaglutide group, 4/' < 0.05, and "P
< 0.01.
The present disclosure is further described below in combination with specific embodiments. It is to be understood that these embodiments serve only to illustrate the present disclosure and are not limiting the scope of the present disclosure.
In the following embodiments, experimental methods without specifying specific conditions are generally performed under conventional conditions, for example, those described in Sambrook et al., Date Recue/Date Received 2020-07-02 Molecular Cloning: A Laboratory Manual (New York: Cold Spring Harbor Laboratory Press, 1989), or the conditions recommended by the manufacturer.
Generally, the dual-function protein of the present disclosure is prepared synthetically.
The nucleotide sequence according to the present disclosure, a person skilled in the art can conveniently use various known methods to prepare the encoding nucleic acid of the present disclosure. These methods are for example but not limited to: PCR, and DNA
artificial synthesis etc., the specific methods can refer to J. Sambrook, "Molecular Cloning: A
Laboratory Manual". As an embodiment of the present disclosure, a method comprising fragment synthesis of nucleotide sequences, followed by overlap extension of PCR can be used for constructing the encoding nucleic acid sequence of the present disclosure.
Also provided in the present disclosure is an expression vector comprising a sequence encoding the dual-function protein of the present disclosure and a regulatory element transcriptionally linked thereto. Said "transcriptionally linked" or "transcriptionally linked to" refer to such a condition that some parts of a linear DNA sequence can regulate or control the activity of other parts in the same linear DNA sequence. For example, if a promoter controls the transcription of a sequence, then the promoter is transcriptionally linked to the encoding sequence.
The expression vector can use commercially available ones, for example but not limited to: vectors pcDNA3, pIRES, pDR and pUC18 which can be used for expression in an eukaryotic system. A person skilled in the art can select a suitable expression vector according to the host cell.
According to the restriction map of the known expression vector, a person skilled in the art can insert the sequence encoding the dual-function protein of the present disclosure into a suitable restriction site to prepare the recombinant expression vector of the present disclosure following conventional methods via restriction digestion and ligation.

Date Recue/Date Received 2020-07-02 Also provided in the present disclosure is a host cell expressing the dual-function protein of the present disclosure, wherein said host cell comprises the sequence encoding the dual-function protein of the present disclosure. In at least one embodiment, said host cell is a eukaryotic cell, for example but not limited to CHO, a COS cell, a 293 cell and a RSF cell etc. As a at least one embodiment of the present disclosure, said cell is a CHO cell, which can well express the dual-function protein of the present disclosure, and the dual-function protein with a good binding activity and stability can be obtained.
Also provided in the present disclosure is a method for preparing the dual-function protein of the present disclosure using recombinant DNA, the steps thereof comprise:
1) Providing a nucleic acid sequence encoding the synergistic dual function protein;
2) Inserting the nucleic acid sequence of 1) into a suitable expression vector, and obtaining a recombinant expression plasmid;
3) Introducing the recombinant expression plasmid of 2) into a suitable host cell;
4) Culturing the transformed host cell under a condition suitable for expression;
5) Collecting the supernatant, and purifying the dual-function protein product.
To introduce said encoding sequence into the host cell one can use multiple known technologies in the art, for example but not limited to: calcium phosphate precipitation, protoplast fusion, liposome transfection, electroporation, microinjection, reverse transcription method, phage transduction method, and alkali metal ion method.
With respect to the culture of and expression in the host cell can refer to Olander RM
Dev Biol Stand, 1996, 86:338. Cells and debris in the suspension can be removed by centrifugation, and the supernatant is collected. Agarose gel electrophoresis technique can be used for identification.
The dual-function protein prepared as described herein can be purified to have a substantially homogeneous property, such as has a single band on SDS-PAGE

Date Recue/Date Received 2020-07-02 electrophoresis. For example, when the recombinant protein is expressed for secretion, a commercially available ultrafiltration membrane (such as products of Millipore and Pellicon etc.) can be used to separate said protein, wherein firstly, the expression supernatant is concentrated. The concentrate can be purified by the method of gel chromatography, or by the method of ion exchange chromatography, for example, by anion exchange chromatography (DEAE etc.) or cation exchange chromatography. The gel matrix can be common matrices for protein purification, such as agarose, glucan, and polyamide etc. Q- or SP-groups is a relatively ideal ion exchange group. Finally, the above-mentioned purified product can be further refined and purified by the methods of hydroxyapatite adsorption chromatography, metal chelate chromatography, hydrophobic interaction chromatography and reversed high performance liquid chromatography (RP-HPLC). All the above-mentioned purification steps can be used in different combination in order to make the protein purity substantially homogeneous.
The expressed dual-function protein can be purified using an affinity column containing a specific antibody, receptor or ligand of said dual function protein.
According to the properties of the affinity column, conventional methods, such as high salt buffer and changing pH etc. can used to elute the fusion polypeptide binding to the affinity column.
Optionally, at the amino terminus or carboxyl terminus of said dual function protein, one or more polypeptide fragments also can be contained as protein tags. Any suitable tags can be used in the present disclosure. For example, said tags can be FLAG, HA, HAL c-Myc, 6-His or 8-His etc. These tags can be used for purifying the dual function protein.
Non-limitipe Exemplary Embodiments Date Recue/Date Received 2020-07-02 EXAMPLES
Example 1: Construction of an expression plasmid of the synergistic dual function protein All gene sequences encoding alpha 1 microglobulin secretion leader signal, GLP-analog, Li, FGF21 mature protein, L2 (comprising a flexible linker unit and rigid linker unit) and human IgG Fc variants were optimized using CHO preferred codons and the full-length gene sequences were synthesized. There are a SpeI at the 5' and a EcoRI at the 3' for subcloning the target gene encoding the fusion protein into the expression vector PXY1A1 modified from PCDNA3.1 (Fig. 1 exemplarily set forth the nucleotide sequence of the dual-function protein FP4I-2 and the translated amino acid sequence). The expression plasmid contained the early promoter of cytomegalovirus, leading to high expression of exogenous genes in mammalian cells. The plasmid also contained a selective marker conferring kanamycin resistance in bacteria, and G418 resistance in mammalian cells.
Furthermore, the host cell carrying DHFR- mutant, PXY1A1 expression vector contained the gene of mouse dihydrofolate reductase (DHFR) could amplify the fusion gene and DHFR gene in the absence of methotrexate (MTX) (see U.S. Patent No. 4,399,216).
Various dual-function proteins comprising GLP-1 and FGF21 were constructed.
Here, three are exemplified: FP4I-1, FP4I-2 and FP4I-3. The amino acid composition is shown in Table 1 (L1 and L2 were underlined, and mutated amino acids in Fc variants were boxed).
Table 1. Amino acid composition of each synergistic dual function protein PI PDS S PLLQFGGQVRQRYLYT DDAQQTEAHLE I REDGTVGGAADQ
S PE SLLQLKALKPGVIQ I LGVKT SRFLCQRPDGALYGSLHFDPEAC
SFRELLLEDGYNVYQSEAHGLPLHLPGNKSPHRDPAPRGPARFLPL
PGLPPAPPEPPGILAPQPPDVGSSDPLSMVGPSQGRS PSYASGGGG
SGGGGSGGGGSGGGGSGGGGSSSSSKAPPPSSSSSKAPPPSESKYG
PPC P PC PAPE FAGGP SVFLF PPKPKDTLMI SRTPEVTCVVVDVSQ
EDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWL
NGKEYKCKVSNKGLPSS I EKT I SKAKGQPREPQVYTLPPSQEEMTK
NQVSLTCLVKGFY PSDIAVEWE SNGQPENNYKTT PPVLDSDGS EEL
YSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK

Date Recue/Date Received 2020-07-02 HGEGTFTSDVSSYLEEQAAKEFIAWLVKGGGGGGGSGGGGSGGGGS
HPIPDSSPLLQFGGQVRQRYLYTDDAQQTEAHLEIREDGTVGGAAD
QSPESLLQLKALKPGVIQILGVKTSRFLCQRPDGALYGSLHFDPEA
CSFRELLLEDGYNVYQSEAHGLPLHLPGNKSPHRDPAPRGPARFLP
LPGLPPAPPEPPGILAPQPPDVGSSDPLSMVGPSQGRSPSYASGSG

TPILPOVECPPCPAPPVAGPSVFLFPPKPKDQLMISRTPEVICVV
VDVSHEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTFRVVSVLIVV
HQDWLNGKEYKCKVSNKGLPAIIIEKTISKTKGQPREPQVYTLPPS
REEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDS
DGSFFLYSKLTVDKSRWQQGNVFSCSVIIHEALHNHYTQKSLSLSP
GK
HGEGTFTSDLSKQMEEEAVRLFIEWLKNGGPSSGAPPPSGGGGGGG
SGGGGSGGGGSHPIPDSSPLLQFGGQVRQRYLYTDDAQQTEAHLEI
REDGTVGGAADQSPESLLQLKALKPGVIQILGVKTSRFLCQRPDGA
LYGSLHFDPEACSFRELLLEDGYNVYQSEAHGLPLHLPGNKSPHRD
PAPRGPARFLPLPGLPPAPPEPPGILAPQPPDVGSSDPLSMVGPSQ
GRSPSYASGSGGGGSGGGGSGGGGSGGGGSGGGGSSSSSKAPPPSL

PSPSRLPGPSDTPILPQVECPPCPAPPVAGPSVFLFPPKPKDQLM
ISRTPEVICVVVDVSHEDPEVQFNWYVDGVEVHNAKTKPREEQFNS
TFRVVSVLTVVHQDWLNGKEYKCKVSNKGLPADEKTISKTKGQP
REPQVYTLPPSREEMTKNQVSLICLVKGFYPSDIAVEWESNGQPEN
NYKTTPPMLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVLHEALHNH
YTQKSLSLSPGKVDKSRWQQGNVFSCSVIIHEALHNHYTQKSLSLS
PGK
Example 2: Expression of the dual-function protein in a transfected cell line A recombinant expression vector plasmid was transfected into a mammalian host cell line to express the synergistic dual function protein. In order to stabilize the high expression, a preferred host cell line was DHFR defective CHO-cell (U.S. Patent No.
4,818,679). In the present example, the host cell was selected from CHO-derived cell line DXB11.
A preferred transfection method was electroporation, but other methods such as calcium phosphate and liposome-induced transfection also can be used. A Gene PulserTM
electroporation apparatus (Bio-Rad Laboratories, Hercules, CA) setting at 300 V of electric field and 1500 pF'd of capacitance was used in the present experiment and 50 pg pure expression plasmid was mixed with 5 x 107 CHO cells in the cuvette. Two days after the transfection, a selection Date recue / Date received 202 1-1 1-25 medium containing 0.6 mg/mL G418 was used. Quantitative ELISA using anti-human IgG
Fc was applied to screen the transfectants with the resistance to G418. Anti-human FGF21 or anti-human GLP-1 by ELISA was used to quantify expression of the dual-function protein.
A 96 well culture plate was subjected to the limiting dilution, the well generating a high level of the dual-function protein was subcloned.
In order to achieve a relatively high expression of the dual-function protein, it was appropriate to use the DHFR gene inhibited by MTX for co-amplification. In another selection medium containing incremental concentrations of MTX, the gene of the dual-function protein was co-amplified with the DHFR gene. The subclone with a positive DHFR
expression was subjected to the limiting dilution, the selection pressure was gradually increased and the transfectant which can grow in a medium with up to 6 p,M MTX
was selected. The secreting rate of transfectant was determined and the cell line with a high expression of exogenous protein was screened out. The cell lines with secretory rate higher than about 10 (preferably about 20) g/106 (i.e. a million) cells/24 hours was subjected to an adaptive suspension culture in serum-free medium, then the dual-function protein was purified by a specified medium.
Example 3: Purification and qualification of the dual-function protein This example describes the exemplary purification and qualification methods of 2. The cell culture supernatant was subjected to clarifying treatments, such as high speed refrigerated centrifugation and 0.22 pm sterile filtration etc., then purified by three chromatograph steps including protein A, anion exchange and hydrophobic chromatography, the specific method was as follows: In the first step, protein A was used for capture, wherein the equilibrium solution was PBS buffer, the eluant was a citrate buffer at pH
3.5, then the eluted protein was neutralized by 1 M Tris solution. In the intermediate purification process, Date Recue/Date Received 2020-07-02 high resolution anion exchange packing material Q SepharoseTM HP (GE company) was selected to remove residual impurity proteins. A combined mode was used, that is, 20 mM
Tris-HC1, 0.2 M NaCl, pH 7.5 solution was used for rinsing, and 20 mM Tris-HC1, 0.3 M
NaCl, pH 7.5 solution was used for elution. In the fine purification step, Butyl Sepharose FF
(GE) was selected to remove polymers; due to different hydrophobic properties of FP4I-2 monomer and polymer, the monomer with weak hydrophobic property flowed through directly, but the polymer with high hydrophobic property bound to the medium;
hydrophobic chromatography was selected as flow through mode, and the equilibrium solution was PBS
buffer.
The qualitative analysis result is shown in Figs. 2a and 2b. The theoretical molecular weight of single stranded FP4I-2 was about 53 KD, due to the absence of glycosylation sites, under the reducing condition, SDS-PAGE electrophoresis showed that the actual mass of the single stranded FP4I-2 molecule was about 70 KB. FP4I-1 and -3 were prepared by the same method.
Example 4: Effect of a single injection of the dual-function protein on glucose utilization in C57BL/6 mice 8 weeks aged male C57BL/6J mice at SPF grade (purchased from Beijing HFK
Bioscience Ltd.) were selected. Housing conditions: temperature 22-25 C, relative humidity 45-65%, and 12h-light/dark cycle. After acclimation for 1 week, mice were randomly divided into control group, Dulaglutide 120 nmol/kg group, FP4I-2 120 nmol/kg group and FP4I-1 120 nmol/kg group (n = 7) according to body weight. The mice in the treatment groups were injected subcutaneously with corresponding drug solutions, while the mice in the control group were injected subcutaneously with PBS buffer. After the injection, mice in each group were fasted for 16 h, and then glucose tolerance test was performed. The fasting Date recue / Date received 202 1-1 1-25 blood glucose values of the mice were determined followed by an intraperitoneal injection of a 2g/kg glucose solution, the blood glucose values were determined at 15 min, 30 min, 60 min, 90 min and 120 min after glucose injection, and the increased area below the curve and above the baseline (iAUC) was calculated by the trapezoidal method. The glucose tolerance test was further performed on the mice of each group at 96 h and 144 h after the administration, and the method was the same as above. The data were represented as means SEM, and analyzed using SPSS18. 0 statistical software. For the Gaussian distribution data, statistical comparison of the means among the groups was performed using one-way ANOVA, followed by LSD test for the homogeneity of variance or Dunnet T3 test for the heterogeneity of variance; non-parametric test was used for the Non-Gaussian distribution data. P < 0.05 represented a significant statistical difference.
As shown in Figs.3a and 3b, FP4I-1 and FP41-2 significantly improved glucose utilization in the mice at 16 h after administration when compared with the control group (P
<0.01). It can be known from Figs.4a and 4b that FP4I-1 and FP4I-2 also can significantly improve the glucose utilization level in mice at 96 h after administration as well (P < 0.01).
It can be known from Figs.5a and 5b that FP4I-1 and FP4I-2 significantly improved the glucose utilization level in mice even at 144 h after administration (P <
0.05). The results showed that in the case of a suddenly increased glucose level in vivo, the GLP-dual-function protein had a rapid response to the glucose level and normalized it to the physiological level by promoting release and secretion of insulin with a long-acting activity, and therefore it could be used for treating diabetes and the complications induced by the absolute or relative deficiency of insulin. When the mice were fasted for 16 h after FP4I-1 or FP4I-2 administration, no shock or death due to hypoglycemia were noted in any mouse, indicating that the dual-function protein would not result in hypoglycemic symptoms as insulin.

Date Recue/Date Received 2020-07-02 In addition, the activity of Exendin4-FGF21 dual-function protein FP4I-3 on the glucose utilization was determined by above-mentioned method as well. C57BL/6 mice were divided into the control group, Dulaglutide group and FP4I-3 group.
Corresponding drug solutions (120nmol/kg) were administrated subcutaneously to the mice in Dulaglutide group and FP4I-3 group, respectively, and PBS buffer was administrated to the mice in the control group. The glucose tolerance test was performed at 16 h, 96 h, and 144 h after the injection.
As shown in Figs. 6a and 6b, 16 h after the administration, compared with the control group, FP4I-3 significantly improved the glucose utilization (P < 0.01), but the activity was significantly weaker than that of Dulaglutide (P < 0.01). As shown in Figs. 7a and 7b, 96 h after the administration, FP4I-3 significantly improved the glucose utilization in mice (P <
0.01) though the efficacy was significantly lower than Dulaglutide as well (P
< 0.01). As shown in Figs. 8a and 8b, 144 h after the administration, the ability of FP41-3 to improve glucose utilization in mice was not observed (P> 0.05).
The glucose tolerance test of FP4I-3 in animals demonstrated that Exendin-4 did not display a synergistic effect with FGF21. The hypoglycemic effect of FP4I-3 was significantly weaker than that of Dulaglutide indicated that the circulating half-life of FP4I-3 was shorter than Dulaglutide. In contrast, the preferred GLP-1-FGF21 dual-function proteins FP4I-2 and FP4I-1 had a relatively strong stability in vivo, and were not easily degraded and inactivated, and maintained a longer in vivo pharmacodynamic activity relative to Exendin4-FGF21 dual-function protein FP4I-3. Above results indicated that the combination modes of three functional components, GLP-1 analogs, FGF21 and Fc fragment in the dual-function protein were not random and arbitrarily, wherein the selection of GLP-1 analogs, the structure of linker peptide, the fusion sequence, even the difference of glycosylation pattern would affect accuracy and stability of the dual-function protein Date Recue/Date Received 2020-07-02 conformation to varying degrees, and it determined whether the active molecules were functionally synergetic and the half-life was prolonged or not.
Example 5: Hypoglycemic effect of dual-function protein in db/db mice 8 weeks aged male db/db mice were purchased from Shanghai SLAC Laboratory Animal Ltd. Housing conditions: temperature 22-25 C, relative humidity 45-65%, and 12 h-light/dark cycle. After housed individually for 1 week as acclimation, the mice were divided into 4 groups according to body weight, blood glucose and food intake: control group, Dulaglutide group, FP4I-1 group and FP4I-2 group (n = 7). Mice in the control group were injected subcutaneously with PBS buffer, and mice in other groups were injected subcutaneously with 120 nmol/kg corresponding drug solutions (twice per week, totally 8 times). Daily food intake of each mouse was recorded. At the end of the dosing period, mice were fasted for 16 hours, 5 pL whole blood sample was collected from the eye socket to measure glycosylated hemoglobin. The data were represented as means standard error ( s), and were analyzed using SPSS 18.0 statistical software. For the data follow Gaussian distribution, one-way analysis of variance was used for comparing mean difference among groups, followed by LSD test for the homogeneity of variance or Dunnet T3 test for the heterogeneity of variance; non-parametric test was used for the data follow non-normal distribution. P < 0.05 represented a significant statistical difference.
As shown in Fig. 9, compared with Dulaglutide group, after the first administration, the food intakes within 24 hour of the mice in FP4I-1 and FP41-2 groups were significantly elevated (P < 0.01). The results showed that GLP-1-FGF21 dual-function protein could significantly relieve symptoms of severe gastrointestinal adverse effects induced by the first administration of long-acting GLP-1 receptor agonist drugs. As shown in Fig.
10, FP4I-1, FP4I-2, Dulaglutide group can significantly lower the glycosylated hemoglobin values of Date Recue/Date Received 2020-07-02 db/db mice (P < 0.01), and the glycosylated hemoglobin values of mice in FP4I-1 and FP4I-2 groups were significantly lower than that in Dulaglutide group (P < 0.05).
db/db mouse is a spontaneously hyperglycemic animal model with a severe insulin tolerance.
The GLP-1-FGF21 dual-function protein exhibited a better property than Dulaglutide in the long-term glycemic control. Based on the data in Example 4, the insulinotropic activity of GLP-1-FGF21 dual-function protein was not significantly better than Dulaglutide.
Wild type FGF21 exhibited a good insulin sensitization effect in the hypeinsulinemic-euglycemic clamp test (Xu J et al., Diabetes, 2009, 58:250-259), but there was no direct evidence showing that Dulaglutide had an insulin sensitization effect in vivo. In conclusion, the superiority in blood glucose control exhibited by GLP-1-FGF21 dual-function protein should be the result of synergistic effect of GLP-1 analog promoting release and secretion of insulin and FGF21 enhancing insulin sensitivity. As shown in Fig. 11, in the experimental period, the cumulative food intake of mice in FP4I-1 and FP4I-2 groups was significantly higher than that in Dulaglutide group (P < 0.05), the results showed that in the condition of excluding factors intervening food intake, the blood glucose control activity of FP4I-1 and FP4I-2 groups on type 2 diabetes should be higher than that of Dulaglutide.
Example 6. Therapeutic effects of the dual-function protein on weight loss, hepatic steatosis and lipid metabolism disorder in obese mice induced by high-fat diet 8 weeks aged C57BL/6 mice were purchased from Shanghai SLAC Laboratory Animal Ltd. Housing conditions: temperature 22-25 C, relative humidity 45-65%, and lighting time 12 h/d. After acclimation for 1 week, 7 mice were selected and fed with low-fat diet (D12450B, Research Diets), and other mice were fed with high-fat diet (D12451, Research Diets). 40 weeks later, obese mice were subjected to adaptive feeding with single animal/cage for 1 week, then the obese mice were divided into five groups according to body Date Recue/Date Received 2020-07-02 weight and weekly food intake: obese control group, Dulaglutide group, high fat diet pair-fed group, FP4I-1 group and FP4I-2 group (n = 7). In the experiment, the amounts of daily diet per mouse in high fat diet pair-fed, FP4I-land FP4I-2 groups were consistent with daily food intake per mouse in Dulaglutide group. Mice in the obese control group and high fat diet pair-fed group were injected subcutaneously with PBS buffer solution, and mice in other groups were injected subcutaneously with 120 nmol/kg corresponding drug solutions, once every 6 days, and totally 2 times. The body weight of each mouse was recorded before and after the dosing period. At the end of the dosing period, mice in each group were fasted for 16 hours, whole blood was collected from the eye socket, and centrifuged at 2000 xg for 15 min to obtain serum. Serum lipid profiles were determined by an automatic biochemical analyzer. Liver tissue was excised, washed with normal saline, then removed residual liquid with filter paper and weighed. About 50 mg liver tissue at the same part of each live was taken, and the triglyceride content was determined using the Folch method. The results were represented in the form of triglyceride content per mg liver tissue. The data were represented as means SEM, and analyzed using SPSS18.0 statistical software. For the Gaussian distribution data, statistical comparison of the means among the groups was performed using one-way ANOVA, followed by LSD test for the homogeneity of variance or Dunnet T3 test for the heterogeneity of variance; non-parametric test was used for the Non-Gaussian distribution data. P < 0.05 represented a significant statistical difference.
As shown in Figs. 12 to 17, after administrated with Dulaglutide, body weight, liver mass, liver triglyceride, triglycerides, total cholesterol and low density lipoprotein-cholesterol contents in serum were significantly lowered (P<0.01) in the obese mice induced by high-fat diet. Dulaglutide could cause severe gastrointestinal adverse effects and suppressed appetite by regulating central nervous system, resulted in reduction in food intake.
In this example, daily supplied the same amount of diet to the mice in the high-fat diet pair-Date Recue/Date Received 2020-07-02 fed group as the corresponding mice in Dulaglutide group, despite the parameters mentioned above were significantly lowered when compared with that in the obese control group, there was no significant statistical difference from that of Dulaglutide group (P >
0.05). The results showed that the effects of Dulaglutide on weight loss, hepatic steatosis and lipid metabolic disorder substantially depended on inhibition of appetite without any other mechanisms. The obese mice in FP4I-1 group and FP4I-2 group were given the same amount of diet as the corresponding mice in Dulaglutide group, compared with Dulaglutide group, body weight and serum triglyceride level of the mice in FP4I-2 group were significantly decreased (P < 0.01), which demonstrated that FP4I-2 had additional functions of reducing fatty acid synthesis and promoting fatty acid metabolism and utilization in vivo. The results indicated that FP4I-2 could be used for treating obesity and obesity-induced metabolic syndrome.
Compared with Dulaglutide group, liver mass and liver triglyceride content of mice in FP4I-2 group were significantly decreased (P < 0.01 or P < 0.05), which demonstrated that FP4I-2 effectively reduced the excessive accumulation of triglyceride in liver, improved liver function. The results indicated that FP4I-2 could be used for treating various liver diseases induced by hepatic steatosis, such as nonalcoholic fatty liver, nonalcoholic steatohepatitis, liver fibrosis and liver cirrhosis.
Compared with Dulaglutide group, both total serum cholesterol and low density lipoprotein-cholesterol content of mice in FP4I-2 group were significantly reduced (P < 0.01 or P < 0.05), indicating that FP4I-2 can be used for treating hypercholesteremia and relevant cardiovascular and cerebrovascular diseases, such as hypertension, coronary heart disease, chronic heart failure, cerebral infarction and atherosclerosis. Compared with Dulaglutide group, the body weight, liver mass, liver triglyceride content, serum triglycerides, total Date Recue/Date Received 2020-07-02 cholesterol and low density lipoprotein cholesterol levels in FP4I-1 group were mildly decreased but no significant differences were observed.
The present study demonstrated that FP4I-1 and FP4I-2 could treat obesity, fatty liver disease and lipid metabolic disorder via the physiological activity of FGF21, and was not completely dependent on the food intake regulation effect of GLP-1 analogs;
the therapeutic effect of FP4I-2 in the obese mice was superior to Dulaglutide, which indicated that it could compensate for the deficiency of Dulaglutide in the clinic. In conclusion, the therapeutic mechanisms of FP4I-2 are more abundant than that of Dulaglutide, which is more suitable for the requirement of diversified clinical therapy.
This disclosure provides merely exemplary embodiments of the disclosure. One skilled in the art will readily recognize from the disclosure and claims, that various changes, modifications and variations can be made therein without departing from the spirit and scope of the disclosure as defined in the following claims.

Date recue / Date received 202 1-1 1-25

Claims (33)

1. A dual-function protein comprising, sequentially, human glucagon-like peptide 1 (GLP-1) analog, linker peptide 1, human fibroblast growth factor 21 (FGF21), linker peptide 2 and human immunoglobulin Fc fragment from the N to C-terminus;
wherein:
the linker peptide 1 comprises a flexible peptide;
the linker peptide 2 comprises a flexible peptide and a rigid peptide;
the rigid peptide comprises at least 1 rigid unit; and the rigid unit comprises carboxyl terminal amino acids 113 to 145 of human chorionic gonadotropin (3-subunit or a truncated sequence thereof; and vvherein the GLP-1 analog comprises the amino acid sequence defined by SEQ ID NO: 2 or 5.
2. The dual-function protein of claim 1, wherein said dual-function protein is glycosylated.
3. The dual-function protein of claim 1, wherein said linker peptide 1 comprises a flexible peptide consisting of 2 or more amino acids.
4. The dual-function protein of claim 3, wherein the amino acids are selected from G, S, A and T.
5. The dual-function protein of claim 4, wherein the amino acid sequence of the flexible peptide is GGGGGGGSGGGGSGGGGS.
6. The dual-function protein of claim 1, wherein said human FGF21 comprises the sequence defined by SEQ ID NO: 6 wherein the leader peptide of amino acid position 1-28 is deleted.
7. The dual-function protein of claim 1, wherein said human FGF21 comprises the sequence defined by SEQ ID NO: 6 wherein the leader peptide of amino acid position 1-28 is deleted and which has G1415 or L174P substitution.
8. The dual-function protein of claim 1, wherein the flexible peptide constituting said linker peptide 2 comprises 2 or more amino acids selected from G, S, A and T.
Date recue / Date received 2021-11-25
9. The dual-function protein of claim 8, wherein the general structural formula of the amino acid composition of said flexible peptide is (GS)a(GGS)b(GGGS)e(GGGGS)d, vvherein a, b, c and d are integers greater than or equal to 0, and a+b+c+d>
1.
10. The dual-function protein of claim8, wherein the amino acid composition of said flexible peptide is selected from:
(i) GGGGS;
(ii) GSGGGSGGGGSGGGGS;
(iii) GSGGGGSGGGGSGGGGSGGGGSGGGGS;
(iv) GGGGSGGGGSGGGGSGGGGSGGGGSGGGGSGGGGS;
(v) GGGSGGGSGGGSGGGSGGGS; and (vi) GGSGGSGGSGGS.
11. The dual-function protein of claim 1, wherein the rigid units constituting said linker peptide 2 are selected from SEQ ID NO: 7 and a truncated amino acid sequence thereof;
wherein said truncated amino acid sequence comprises at least 2 glycosylation sites.
12. The dual-function protein of claim 11, wherein the rigid units comprise one of the following amino acid sequences:
(i) SS SSKAPPPSLPSPSRLPGPSDTPILPQ;
(ii) PRFQDSSSSKAPPPSLPSPSRLPGPSDTPILPQ;
(iii)SSSSKAPPPS;
(iv) SRLPGPSDTPILPQ; or (v) SSSSKAPPPSLPSPSR.
13. The dual-function protein of claim 11, wherein said rigid units comprise an amino acid sequence that has at least 90% or 95% amino acid identity with SEQ ID NO: 7 .
14. The dual-function protein of claim 11, wherein said rigid units comprise the truncated amino sequence of SEQ ID NO: 7.
15. The dual-function protein of claim 1, wherein said rigid peptide comprises 1, 2, 3, 4 or rigid units.

Date recue / Date received 2021-11-25
16. The dual-function protein of claim 1, wherein said human immunoglobulin Fc fragment is a variant having a reduced ADCC effect and/or CDC effect and/or enhanced binding affinity with FcRn receptor.
17. The dual-function protein of claim 16, wherein said Fc variant is selected from:
(i) hinge, CH2 and CH3 regions of human IgG1 containing Leu234Va1, Leu235A1a and Pro331Ser mutations;
(ii) hinge, CH2 and CH3 regions of human IgG2 containing Pro331Ser mutation;
(iii) hinge, CH2 and CH3 regions of human IgG2 containing Thr250G1n and Met428Leu mutations;
(iv) hinge, CH2 and CH3 regions of human IgG2 containing Pro331Ser, Thr250G1n and Met428Leu mutations; and (v) hinge, CH2 and CH3 regions of human IgG4 containing Ser228Pro and Leu235A1a mutations.
18. The dual-function protein of claim 1, comprising the amino acid sequence defined by SEQ ID NO: 13 or 15.
19. A DNA molecule encoding the dual-function protein of claim 1.
20. A DNA molecule comprising the sequence defined by SEQ ID NO: 14.
21. A vector comprising the DNA molecule of claim 19.
22. A host cell comprising the DNA molecule of claim 19 or 20, or the vector of claim 21.
23. A pharmaceutical composition, wherein the pharmaceutical composition comprises a pharmaceutically acceptable carrier, excipient or diluent, and an effective dose of the dual-function protein of claim 1.
24. A method for preparing a dual-function protein, said method comprising:
(a) introducing the DNA sequence encoding the dual-function protein of claim 19 into a mammalian cell;
(b) screening a high-yield cell strain expressing more than 20 ng/106 (million) cells within a period of every 24 hours in the growth medium thereof from step (a);
(c) culturing the screened cell strain in step (b), and expressing the dual-function protein;

Date recue / Date received 2021-11-25 (d) harvesting fermentation supernatant obtained from step (c), and purifying the dual-function protein.
25. The method of claim 24, wherein said mammalian cell in step (a) is a CHO cell.
26. The method of claim 25, wherein said mammalian cell is CHO-derived cell line DXB-11.
27. Use of the dual-function protein of any one of claims 1-18 in the manufacture of a medicament for treating one or more FGF21 related diseases and GLP-1 related diseases.
28. Use of the dual-function protein of any one of claims 1-18 for treating one or more FGF21 related diseases and GLP-1 related diseases.
29. Use of the pharmaceutical composition of claim 23 in the manufacture of a medicament for treating one or more FGF21 related diseases and GLP-1 related diseases.
30. Use of the pharmaceutical composition of claim 23 for treating one or more related diseases and GLP-1 related diseases.
31. The dual-function protein of any one of claims 1-18 for use in treating one or more FGF21 related diseases and GLP-1 related diseases.
32. The pharmaceutical composition of claim 23 for use in treating one or more FGF21 related diseases and GLP-1 related diseases.
33. The use of any one of claims 27-30, or the for use of claim 31 or 32, wherein the disease is obesity, type 1 diabetes, type 2 diabetes, pancreatitis, dyslipidemia, nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, insulin resistance, hyperinsulinemia, glucose intolerance, hyperglycemia, metabolic syndrome, acute myocardial infarction, hypertension, cardiovascular disease, atherosclerosis, peripheral arterial disease, stroke, heart failure, coronary heart disease, nephropathy, diabetic complication, neuropathy, gastroparesis, or symptoms associated with the severe inactivation mutations of insulin receptor.

Date recue / Date received 2021-11-25
CA3085252A 2020-07-02 2020-07-02 Dual-function protein for lipid and blood glucose regulation Pending CA3085252A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CA3085252A CA3085252A1 (en) 2020-07-02 2020-07-02 Dual-function protein for lipid and blood glucose regulation

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CA3085252A CA3085252A1 (en) 2020-07-02 2020-07-02 Dual-function protein for lipid and blood glucose regulation

Publications (1)

Publication Number Publication Date
CA3085252A1 true CA3085252A1 (en) 2022-01-02

Family

ID=79296015

Family Applications (1)

Application Number Title Priority Date Filing Date
CA3085252A Pending CA3085252A1 (en) 2020-07-02 2020-07-02 Dual-function protein for lipid and blood glucose regulation

Country Status (1)

Country Link
CA (1) CA3085252A1 (en)

Similar Documents

Publication Publication Date Title
CN113603794B (en) Synergistic bifunctional proteins for regulating blood glucose and lipids
CN110229238B (en) Human fibroblast growth factor 21 fusion protein and preparation method and application thereof
US20220002368A1 (en) Dual function proteins comprising fgf21 mutant protein and pharmaceutical composition comprising same
RU2741087C2 (en) Fgf21 fused with long-acting proteins and pharmaceutical composition containing same
US8809499B2 (en) Fusion protein of human fibroblast growth factor-21 and exendin-4
TWI784914B (en) Composition for treating diabetes comprising long-acting insulin analogue conjugate and long-acting insulinotropic peptide conjugate
JP2020079301A (en) Compositions comprising oxyntomodulin analogs for treating diabetes or diabesity
PT1831252E (en) Glp-1 analog fusion protein formulations
CN110121355B (en) Pharmaceutical composition comprising fusion protein for preventing or treating hepatitis, liver fibrosis and liver cirrhosis
US20210371488A1 (en) Dual-function protein for lipid and blood glucose regulation
CA3085252A1 (en) Dual-function protein for lipid and blood glucose regulation
AU2020204470B1 (en) Dual-function protein for lipid and blood glucose regulation
WO2022143515A1 (en) Human glp-1 polypeptide variant and application thereof
WO2022143516A1 (en) Human glp-1 polypeptide variant and use thereof
KR20220157910A (en) Composition For Combi-Therapy Comprising Growth Differentiation Factor-15 Variant and Glucagon-Like Peptide-1 Receptor Agonist
WO2023278786A1 (en) Chimeric proteins for targeted delivery of growth factors to the glomerulus