Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
DUAL AGONIST FUSION PROTEINS
Document Type and Number:
WIPO Patent Application WO/2020/028806
Kind Code:
A1
Abstract:
Disclosed herein are fusion proteins including an elastin-like peptide domain, a GLP-1 receptor agonist domain attached to a N-terminal end of the ELP domain, and a FGF21 receptor agonist domain attached to the C-terminal end of the ELP domain. Also disclosed are methods of making the fusion proteins, compositions including a plurality of fusion proteins, and uses of the fusion proteins and compositions.

Inventors:
CHILKOTI ASHUTOSH (US)
GILROY CASLIN (US)
Application Number:
PCT/US2019/044911
Publication Date:
February 06, 2020
Filing Date:
August 02, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV DUKE (US)
International Classes:
A61K38/18; A61K38/26; C07K14/50; C07K14/605
Domestic Patent References:
WO2017192449A12017-11-09
Foreign References:
US20110082283A12011-04-07
Other References:
See also references of EP 3829622A4
Attorney, Agent or Firm:
ZERN, Blaine, J. et al. (US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A fusion protein comprising:

an elastin-like polypeptide (ELP) domain;

a GLP-1 receptor agonist domain attached to a N-terminal end of the ELP domain; and a FGF21 receptor agonist d omai n attached to a C-terminal end of the ELP d omai n

2. The fusion protein of claim 1 , wherein the ELP domain comprises an ammo acid sequence of (VPGXG)n (SEQ ID NO: !), wherein X is any amino acid except proline and n is 2 to 200.

The fusion protein of claim 2, wherein n is 80 to 160.

4. The fusion protein of claim 2, wherein X is valine, alanine, leucine, or a combination thereof.

5. The fusion protein of claim 4, wherein X is a ratio of valine: alanine of 1 :0 to 10: 1.

6. The fusion protein of claim 5, wherein n is 120 and X is a ratio of valine: alanine of 4: 1.

7. The fusion protein of claim 1, wherein the GLP-1 receptor agonist domain comprises an ammo acid sequence selected from the group consisting of (SEQ ID NO: 3), (SEQ ID NQ:4), (SEQ ID NO: 5), and variants thereof.

8. The fusion protein of claim 1, wherein the FGF21 receptor agonist domain comprises an ammo acid sequence selected from the group consisting of (SEQ ID NO: 6), (SEQ ID NO: 7), (SEQ ID NO:8), (SEQ ID NO:9), and variants thereof.

9. The fusion protein of claim 1 , wherein the fusion protein has a transition temperature (Tt) of about 25 °C to about 37 °C.

10. The fusion protein of claim 1, wherein the fusion protein has a molecular weight of about 50 kDa to about 100 kDa.

11. A composition comprising:

a plurality of fusion proteins according to claim 1, wherein the plurality of fusion proteins assemble into an aggregate above the Tt of the fusion protein.

12. The composition of claim 11, further comprising a biologically active agent.

13. A method of treating a metabolic disease in a subject in need thereof, the method comprising administering to the subject an effective amount of the composition of claim 1 1.

14. The method of claim 13, wherein the metabolic disease is selected from the group consisting of obesity, type 2 diabetes mellitus, pancreatitis, dyslipidemia, nonalcoholic fatty liver disease (NAFLD), nonalcoholic steatohepatitis (NASH), insulin resistance, hyperinsulinemia, glucose intolerance, hyperglycemia, and a combination thereof.

15. The method of claim 13, wherein the composition releases the fusion protein following administration for greater than 3 days.

16. The method of claim 13, wherein administration of the composition results in the subject having at least one of decreased blood glucose level, decreased body fat, increased insulin production, decreased hemoglobin Ale values, decreased circulating faty acids, decreased liver fat content, decreased liver inflammation, and decreased liver fibrosis compared to a subject not receiving the administration of the composition.

17. A method of synthesizing a dual agonist fusion protein, the method comprising: - transforming a bacteria with a recombinant expression vector comprising a first polynucleotide encoding the fusion protein of claim 1 ; and

- culturing the transformed bacteria to express the fusion protein.

18. The method of claim 17, wherein the expression vector further comprises a second polynucleotide encoding a translation initiation domain attached to the N-termina! end of the fusion protein.

19. The method of claim 18, wherein the translation initiation domain comprises a leader sequence and a protease cleavage site, the protease cleavage site located between the leader sequence and the fusion protein.

20. The method of claim 17, wherein culturing is done at less than 37 °C.

Description:
DIJAL AGONIST FUSION PROTEINS

CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] This application claims priority to U.S. Provisional Application No. 62/713,752 filed on August 2, 2018, which is incorporated fully herein by reference.

SEQUENCE LISTING

[0002] The sequence listing is filed with the application m electronic format only and is incorporated by reference herein. The sequence listing text file "028193-9331-

WO01 As Filed Sequence Listing.txt" was created on August 2, 2019, and is 81,600 bytes in size.

BACKGROUND

[0003] Over 30 million people in the United States have diabetes, with the number of diagnosed individuals growing rapidly, and obesity serving as a major risk factor. Type 2 diabetes mellitus (T2D) accounts for most new cases and is characterized by a state of insulin resistance and impaired ability to maintain glucose homeostasis. Treatment beyond lifestyle changes generally begins with oral anti-diabetic agents, however these medicines have only transitory benefit as the progressive nature of T2D requires therapeutic intensification ultimately including insulin within 5-10 years for many. Moreover, many current treatments, including insulin, are frequently accompanied by weight gam. Thus, there is a pressing need for the development of drugs or drug combinations that maximize glycemic control while promoting weight loss.

SUMMARY

[0004] In one aspect, disclosed are fusion proteins comprising an elastm-like polypeptide (ELP) domain; a GLP-1 receptor agonist domain attached to a N -terminal end of the ELP domain; and a FGF21 receptor agonist domain attached to a C-terminal end of the ELP domain. [0005] in another aspect, disclosed are compositions comprising a plurality of fusion proteins as disclosed herein, wherein the plurality of fusion proteins assemble into an aggregate above the T t of the fusion protein.

[0006] In another aspect, disclosed are methods of treating a metabolic disease in a subject in need thereof, the method comprising administering to the subject an effective amount of the composition as disclosed herein.

[0007] In another aspect, disclosed are methods of synthesizing a dual agonist fusion protein, the method comprising transforming a bacteria with a recombinant expression vector comprising a first polynucleotide encoding the fusion protein as disclosed herein; and culturing the transformed bacteria to express the fusion protein.

BRIEF DESCRIPTION OF THE DRAWINGS

[0008] This patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.

[0009] FIG. 1 is a schematic of a dual agonist fusion protein.

[0010] FIG. 2 is a schematic for improving bacterial expression of dual agonist fusion proteins.

[0011] FIG. 3 is a series of plots showing that recombinant GLP1-ELP-FGF21 fusion proteins have dual agonism and lower critical solution temperature (LCST) phase behavior (FIG. 3A and FIG. 3B) In vitro GLP-1 and FGF21 activity assays for GLP1 -ELP-FGF21. GLP-1R agonism (FIG. 3 A) was measured by quantifying cAMP production in HEK293 cells stably expressing the GLP-1R and a cAMP-inducible luciferase reporter. Cells were stimulated 5 h with GLP1-ELP- FGF21, the GLP1-ELP single agonist control, or native GLP-1. FGF21 receptor agonism (FIG. 3B) was measured by quantifying ERK1/2 phosphorylation in HEK293 cells stably expressing FGFR1 and pKlotho, and normalizing phospho-ERKl/2 to total ERKl/2 Cells were stimulated 5 min with GLP1-ELP-FGF21, the ELP-FGF21 single agonist control, or native FGF21. Data are presented as mean ± SEM, n=3. (FIG. 3C, FIG 3D, and FIG. 3E) LCST phase transition behavior of GLP1 -ELP-FGF21. (FIG. 3C) The optical density at 350 nm of GLP1-ELP-FGF21 at the indicated concentration m PBS, was measured as a function of temperature, with temperature ramped at a rate of l°C/min. (FIG. 3D) Turbidity vs. temperature scans were repeated as in (FIG. 3C) for the indicated concentrations (n=3). T t s were defined as the temperature corresponding to the 50% maximum optical density and are plotted as a function of fusion protein concentration. The horizontal dashed line indicates the approximate temperature of the subcutaneous (s.c.) space in a mouse. (FIG. 3E) A turbidity scan was repeated for GLP1 - ELP-FGF21 at an injection-relevant concentration (150 mM), with the temperature ramped up to 37°C, and then down to 20°C.

[0012] FIG. 4 is a set of plots showing that GLP-1 and FGF21 co-treatment can augment the blood giucose-low/ering and weight gain-inhibiting effects of single drug treatment. 6-week-old db/db mice (n=6-7) were injected s.c. with 1000 nmol/kg ELP-FGF21, 1000 nmol/kg GLP1- ELP, or 1000 nmol/kg each of GLP1-ELP and ELP-FGF21. Ambient blood glucose levels (FIG. 4A) and body weights (FIG. 4B) were measured 48 h post- injection and reported as a magnitude change from pre- treatment baseline and a % change from pre-injection weight. Data are presented as mean ± SEM; * = treatment compared to vehicle; L = comparisons between treatments; */ L = p<005; **/ /n = p<0 01; ***/ LLL = p<0.001; *** # / LLLL = pO.0001; NS = not significant.

[0013] FIG. 5 is a set of plots showing that GLP1-ELP-FGF21 dual agonist fusion proteins have potent and sustained effects on glycemia and body weight. (FIG. 5A, FIG. 5B, FIG. 5C, and FIG. 5D) 6-week-oid db/db mice (n=3-4) received a single s.c. injection of GLP1-ELP-FGF21 at the indicated dose or vehicle. Ambient blood glucose levels were measured every' 24 h until animals returned to baseline levels and are reported as blood glucose vs. time AUC or raw values (FIG. 5 A and FIG. 5B). Body weights were recorded daily and are reported as a % change from pre-injection weight over time (FIG. 5C) or on day 7 post-injection (FIG. 5D). (FIG. 5E) 1000 nmol/kg GLP1 -ELP-FGF21 was administered s.c to 6- week-old db/db mice (n=4) as a radiolabeled protein, blood samples were collected at indicated time points following injection, and plasma gamma counts were correlated to fusion protein concentration. Regression curves were fit to the terminal portion of the data set, and data could be described by either a first-order (dotted) or a zero-order (solid) elimination model. Data are presented as mean ± SEM, * =

P 0.05. ** 0.01 .

[0014] FIG. 6 is a series of plots showing production and in vitro characterization of single agonist ELP fusion controls. (FIG. 6A) In vitro activity assay evaluating the ECso of GLP1-ELP fusion protein at the GLP-1 R. GLP-1R ago sm was measured by quantifying cAMP production following 5 h stimulation of HEK293 cells stably expressing the GLP-lR and a cAMP-inducible luciferase reporter. Data are presented as mean ± SEM, n=3. (FIG. 6B, FIG. 6C, FIG. 6D, FIG. 6E, FIG. 6F, and FIG. 6G) LCST phase transition behavior of single agonist ELP fusion proteins. (FIG. 6B and FIG. 6C) The optical density at 350 nm measured as a function of temperature for GLP1-ELP (FIG. 6B) or a 1 : 1 mixture of GLPl-ELP and ELP-FGF21 (FIG. 6C). Dilutions were prepared in PBS, with the 1 : 1 mixture consisting of the indicated concentration of each respective fusion protein. Temperature was ramped at a rate of l°C/min. (FIG. 6D and FIG. 6E) Turbidity' vs. temperature scans were repeated as in (FIG. 6B and FIG. 6C) for the indicated concentrations (n=3). T t s were measured as the temperature corresponding to the 50% maximum optical density and plotted as a function of concentration. The horizontal dashed line indicates the approximate temperature of the s.c. space m a mouse. (FIG. 6F and FIG. 6G) Turbidity scans were repeated for GLPl-ELP and the 1 : 1 mixture at injection-relevant concentrations (200 mM and 100 mM, respectively), ramping up to 37°C, then down to 20°C.

[0015] FIG. 7 is a set of plots showing single agonist ELP fusion control pharmacokinetic profiles. 6-week-old db/dh mice (n=4~5) received a single s.c. injection of GLPl-ELP (FIG. 7A), or a 1 : 1 mixture of GLPl-ELP and ELP-FGF21 (FIG 7B). GLPl -ELP was radiolabeled in (FIG. 7A), while the L I mixture in (FIG 7B) was tested once when GLPl-ELP was radiolabeled and once when ELP-FGF2I was radiolabeled. All fusions were injected at 200 mM and dosed at 1000 nmol/kg, with the 1: 1 mixture consisting of 1000 nmol/kg each GLPl-ELP and ELP-FGF21. Blood samples were collected at indicated time points following injection, and plasma gamma counts were measured and correlated to fusion protein concentration. Lines represent regression curves fit to the terminal portion of each data set. Data can be described by both a first-order (dotted) or a zero-order (solid) elimination model. Data are presented as mean ± SEM.

[0016] FIG. 8 is a series of plots showing raw body weight and %HbAlc values for chronic dual agonist treatment study. 6-week-old dh/db mice (n=6-7) were treated weekly for 4 weeks with GLP 1 -ELP-F GF21 , GLP1 -ELP, ELP-FGF21 , a 1 : 1 mixture of GLP1 -ELP and ELP-FGF21 , or PBS vehicle. Drugs were administered s.c. at 1000 nmol/kg (or 1000 nmol/kg of each in the drug mixture). (FIG. 8A) Body weights are consistent between treatment groups at the glucose tolerance test (GTT) performed 3 days after the first injection (Day 3). (FIG. 8B) Chronic treatment with GLP-1/FGF21 combination therapy protects from %HbAlc elevation. %HbAlc was measured prior to the first treatment (Day 0) and 6 days following the final treatment (Day 27). (FIG. 8C) Body weights are consistent between mice treated chronically with either a 1 : 1 mixture of GLP1-ELP and ELP-FGF21 or the GLP1 -ELP-FGF21 dual agonist drug. Data are presented as mean ± SEM, ** = p<0.01, NS : = not significant.

[0017] FIG. 9 is a series of plots sho wing acute and chronic GLP1-ELP-FGF21 dual agonist performance. 6-week-old db/db mice (n=6-7) were treated weekly for 4 weeks with GLP1-ELP- FGF21 , GLP 1 -ELP, ELP-FGF21, a 1 : 1 mixture of GLP 1 -ELP and ELP-FGF21 , or PB S vehicle. Fusion proteins w r ere administered s.c. at 1000 nmol/kg (or 1000 nmol/kg of each in the drug mixture). (FIG. 9A, FIG. 9B, and FIG. 9C) Glucose challenge. 72 h after the first treatment cycle, the dual agonist and single agonist control cohorts were fasted 5 h, baseline blood glucose and plasma insulin levels were measured (FIG. 9A), and mice were injected i.p. with 0.75 g/kg glucose. Blood glucose levels were measured at indicated time points (FIG. 9B), and blood glucose vs. time AUC values were calculated (FIG. 9C). (FIG. 9D) Prior to the first treatment (Day 0) and 6 days following the final treatment (Day 27), %HbAlc was measured in all cohorts, and reported as a magnitude change from pre-study values. (FIG. 9E and FIG. 9F) Body weights and food consumption were measured every 1-2 days and are reported as % change from pre- injection weights (FIG. 9D) and cumulative food intake per mouse (FIG. 9E). (FIG. 9G, FIG.

9H, and FIG. 91) 72 h following the final treatment cycle, a GTT was repeated m the dual agonist and 1 : 1 mixture treatment cohort. Data are presented as mean ± SEM; * = p<0.05; *** = p<0.001 ; **** = p<0.0001; NS = not significant.

[0018] FIG. 10 is a set of plots showing that GLP1 -ELP-FGF21 treatment does not predispose mice to nocturnal hypoglycemia or excessive glucose-stimulated insulin secretion. (FIG. 10A) 8- week-old db/db mice (n= :: 6) were injected s.c. with 1000 nmol/kg GLP1 -ELP-FGF21 or vehicle, and ad libitum- fed blood glucose levels were measured 48 h following treatment administration (“Fed”). Mice were then subjected to an overnight 16 h fast, after which blood glucose measurements were repeated (“Fasted”). The horizontal dashed line indicates the traditionally defined threshold of hypoglycemia, 55 mg/dL (FIG. 10B) 6-week-old db/db mice (n=6-7) were injected s.c. with 1000 nmol/kg dual agonist or vehicle, and subjected to a glucose tolerance test 72 h post-treatment. Animals were fasted 5 h, after which mice were injected i.p. with 0.75 g/kg glucose. Pasting plasma insulin levels w¾re measured immediately before the glucose bolus, and glucose-stimulated insulin levels were measured 10 nun following the bolus. Data are presented as mean ± SEM, * = p < 0.05. NS = not significant. [0019] FIG. 11 is a plot of in vitro activity of GLP1-ELP single agonist control at the GLP1R GLPl-ELP2o%Aia ,i 2o incorporates an ELP of 120 VPGXaaG pentapeptide repeats with a 2:8 ratio of alanine:va!ine at the X aa residue, which matches the ELP employed in the GLP1 -ELP-FGF21 dual agonist. GLP1 -ELP 40%Aia,i60 i ncorporates an ELP of 160 VPGXa a G repeats with a 4:6 ratio of alanmewalme at the X residue. GLP1R agonism was measured by quantifying cAMP production following 5 h stimulation of HEK293 cells stably expressing the GLP1R and a cAMP-inducible luciferase reporter. Data are presented as means ± SEM, n=3. ECsos were determined by fitting a three-parameter dose-response curve.

[0020] FIG. 12 is a set of plots showing GLP1-ELP effects on giycemia and body weight. 6- week-old db/db mice (n=3-4) received a single s.c. injection of 1000 nmol/kg GLP1- ELP 2 o%Aia , 120 or PBS vehicle. Ambient blood glucose levels (FIG. 12A) were measured eve 24 h for 10 days, and body weights (FIG. 12B) were recorded daily and reported as a % change from pre- injection weight over time. Data are presented as means ± SEM.

DETAILED DESCRIPTION

[0021] It has been found that GLP-1 and FGF21 can each (1) promote insulin production in the b-cell; and (2) can exert cytoprotective effects on b-cells through pathways that appear to converge. Disclosed herein are fusion proteins that incorporate GLP-1 and FGF21 into a unimolecular drug that can provide synergistic effects at the cellular level, and can translate to superior efficacy in vivo when compared to a GLP1/FGF21 drug mixture.

1. Definitions

[0022] Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. In case of conflict, the present document, including definitions, will control. Preferred methods and materials are described below; although methods and materials similar or equivalent to those described herein can be used in practice or testing of the present invention. All publications, patent applications, patents and other references mentioned herein are incorporated by reference in their entirety.

The materials, methods, and examples disclosed herein are illustrative only and not intended to be limiting.

[0023] The terms“comprise(s),”“include(s),”“having,”“has, ”“can, “contain(s),” and variants thereof, as used herein, are intended to be open-ended transitional phrases, terms, or words that do not preclude the possibility of additional acts or structures. The singular forms “a,”“and” and“the” include plural references unless the context clearly dictates otherwise. The present disclosure also contemplates other embodiments“comprising,”“consisting of’ and “consisting essentially of,” the embodiments or elements presented herein, whether explicitly set forth or not.

[0024] The modifier“about” used in connection with a quantity is inclusive of the stated value and has the meaning dictated by the context (for example, it includes at least the degree of error associated with the measurement of the particular quantity). The modifier“about” should also be considered as disclosing the range defined by the absolute values of the two endpoints. For example, the expression“from about 2 to about 4” also discloses the range“from 2 to 4.” The term“about” may refer to plus or minus 10% of the indicated number. For example,“about 10%” may indicate a range of 9% to 11 %, and“about 1” may mean from 0.9-1.1. Other meanings of“about” may be apparent from the context, such as rounding off, so, for example “about 1” may also mean from 0.5 to 1.4.

[0025] For the recitation of numeric ranges herein, each intervening number there between with the same degree of precision is explicitly contemplated. For example, for the range of 6-9, the numbers 7 and 8 are contemplated in addition to 6 and 9, and for the range 6.0-7.0, the number 6.0, 6.1 , 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, and 7.0 are explicitly contemplated.

[0026] The term“agonist,” as used herein refers to a polypeptide-based entity that binds to a receptor and activates the receptor to produce a biological response. An“antagonist” blocks or inhibits the action or signaling of the agonist. An“inverse agonist” causes an action opposite to that of the agonist. The activities of agonists, antagonists, and inverse agonists may be determined in vitro , in situ, in vivo, or a combination thereof.

[0027] The term“amino acid” as used herein refers to naturally occurring and non-natural synthetic amino acids, as well as amino acid analogs and amino acid mimeties that function in a manner similar to the naturally occurring amino acids. Naturally occurring ammo acids are those encoded by the genetic code. Amino acids can be referred to herein by either their commonly known three-letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission.

[0028] The term“biologically active agent,” as used herein, refers to a substance that can act on a cell, virus, tissue, organ, organism, or the like, to create a change in the functioning of the cell, virus, tissue, organ, or organism. Examples of a biologically active agent include, but are not limited to, drugs, pharmaceuticals, anti-microbial agents, cells, proteins, and nucleic acids. A biologically active agent is capable of treating and/or ameliorating a condition or disease, or one or more symptoms thereof, in a subject. Biologically active agents of the present disclosure also include prodrug forms of the agent.

[0029] The term“C-termina! end,” as used herein refers to a fragment of a polypeptide that begins at any amino acid in the C-termmal half of the polypeptide and ends at the last amino acid of the polypeptide. For example, the C-terminai end of SEQ ID NO: 19 begins at any amino acid from about amino acid 300 to about amino acid 590 of SEQ ID NO: 19 and ends at ammo acid 600 of SEQ ID NO: 19.

[0030] The term“C-terminus,” as used herein refers to the last amino acid of a polypeptide.

[0031] The term "effective amount" or“therapeutically effective amount,” as used herein refers to an amount sufficient to effect beneficial or desirable biological and/or clinical results.

[0032] The term“expression vector,” as used herein indicates a plasmid, a virus or another medium, known in the art, into which a nucleic acid sequence for encoding a desired protein can be inserted or introduced.

[0033] The term“fusion,” as used herein refers to a single protein or polypeptide that is produced by joining two or more originally separate genes into a single gene.

[0034] The term“host cell,” as used herein is a cell that is susceptible to transformation, transfection, transduction, conjugation, and the like with a nucleic acid construct or expression vector. Host cells can be derived from plants, bacteria, yeast, fungi, insects, animals, etc. In some embodiments, the host cell includes Escherichia coii.

[0035] The term“metabolic disease,” as used herein refers to a cluster of conditions, such as increased blood pressure, high blood sugar, excess body fat, and abnormal cholesterol or triglyceride levels --- that can occur together or individually, increasing the risk of, e.g , heart disease, stroke and diabetes. Examples include, but are not limited to, type 1 diabetes mellitus, type 2 diabetes mellitus, postprandial hyperglycemia, overweight, obesity, metabolic syndrome, gestational diabetes, pancreatitis, dyslipidemia, nonalcoholic fatty liver disease (NAFLD), nonalcoholic steatohepatitis (NASH), insulin resistance, hypermsulinemia, glucose intolerance, and hyperglycemia. [0036] The term“N-terminal end,” as used herein refers to a fragment of a polypeptide that begins at the first amino acid of the polypeptide and ends at any ammo acid in the N-terminal half of the polypeptide. For example, the N-terminal end of SEQ ID NO: 19 is from ainino acid 1 of SEQ ID NO: 19 to any amino acid from about amino acid 10 to amino acid 300 of SEQ ID NO: 19.

[0037] The term“N-temnnus,” as used herein refers to the first amino acid of a polypeptide.

[0038] The term“peptide,” as used herein, refers to a linked sequence of two or more amino acids linked by peptide bonds.

[0039] The term“subject,”“patient,” or“organism,” as used herein, includes humans and mammals (e.g., mice, rats, pigs, cats, dogs, and horses). Typical subjects of the present disclosure may include mammals, particularly primates, and especially humans. For veterinary applications, suitable subjects may include, for example, livestock such as cattle, sheep, goats, cows, swine, and the like; poultry such as chickens, ducks, geese, turkeys, and the like, as well as domesticated animals particularly pets such as dogs and cats. For diagnostic or research applications, suitable subjects may include mammals, such as rodents (e.g., mice, rats, hamsters), rabbits, primates, and swine such as inbred pigs and the like.

[0040] The term“treatment” or“treating,” as used herein when referring to protection of a subject from a disease, means preventing, suppressing, repressing, ameliorating, or completely eliminating the disease. Preventing the disease involves administering a composition of the present disclosure to a subject prior to onset of the disease. Suppressing the disease involves administering a composition of the present disclosure to a subject after induction of the disease but before its clinical appearance. Repressing or ameliorating the disease involves administering a composition of the present disclosure to a subject after clinical appearance of the disease.

[0041] The term“variant,” as used herein refers to a peptide or polypeptide that differs in amino acid sequence by the insertion, deletion, or conservative substitution of amino acids, hut retain at least one biological activity. Representative examples of“biological activity” include the ability to be bound by a specific antibody or polypeptide or to bind to its specific receptor. Variant can mean a substantially identical sequence. Variant can mean a functional fragment thereof. Variant can also mean a polypeptide with an amino acid sequence that is substantially identical to a referenced polypeptide with an ammo acid sequence that retains at least one biological activity. A conservative substitution of an ammo acid, i.e., replacing an amino acid with a different ammo acid of similar properties (e.g., hydrophificity, degree and distribution of charged regions) is recognized in the art as typically involving a minor change.

[0042] A variant can be a polynucleotide sequence that is substantially identical over the full length of the full gene sequence or a fragment thereof. The polynucleotide sequence can be

80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical over the full length of the gene sequence or a fragment thereof. A variant can be an amino acid sequence that is substantially identical over the full length of the amino acid sequence or fragment thereof The amino acid sequence can be 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical over the full length of the amino acid sequence or a fragment thereof

2. Dual Agonist Fusion Proteins

[0043] Disclosed herein are dual agonist fusion proteins that comprise an elastin-like polypeptide (ELP) domain, a glucagon-like peptide- 1 (GLP-1) receptor agonist domain, and a fibroblast growth factor 21 (FGF21) receptor agonist domain. The dual agonist fusion protein (also referred to as fusion protein herein) has a linear architecture where the GLP-1 receptor agonist domain is attached to a N-termina! end of the ELP domain, and the FGF21 receptor agonist domain is attached to a C-terminal end of the ELP domain. Accordingly, the ELP domain is positioned in between the GLP-1 receptor agonist domain and the FGF21 receptor agonist domain. The linear architecture of the fusion protein can allow each of the receptor agonist domains to interact with their target receptors.

[0044] The disclosed fusion proteins have a phase transition at a transition temperature (T t ) due to the presence of the ELP domain.“Phase transition” or“transition” refers to the aggregation of fusion proteins, which occurs sharply and in some instances reversibly at a specific temperature. The phase transitioning ability and T t of the fusion protein can be analyzed via a UV-Vis spectrophotometer and other techniques known within the art. Below the T t , for example, the fusion protein may be highly soluble. Upon heating above the transition temperature, for example, the fusion protein may hydrophobically collapse and aggregate, forming a separate, phase. The T t of the fusion protein may be dependent on the T t of the ELP domain alone (e.g., unattached to the receptor agonist domains). An aggregate of fusion proteins may have a varying size. The aggregate may be, for example, nanoscale aggregates, micron- sized aggregates, or macroscale aggregates. In some embodiments, at a temperature above the T t , the aggregate has a diameter or length of about 100 nm to about 1 cm.

[0045] The fusion protein may have a T t of about 20 °C to about 40 °C, such as about 25 °C to about 37 °C, about 26 °C to about 35 °C, or about 27 °C to about 32 °C. The fusion protein may have a T t greater than 20 °C, greater than 21 °C, greater than 22 °C, greater than 23 °C, greater than 24 °C, or greater than 25 °C. The fusion protein may have a T t less than 40 °C, less than 39 °C, less than 38 °C, less than 37 °C, less than 36 °C, or less than 35 °C.

[0046] The fusion protein may undergo phase transition at varying concentrations. For example, the fusion protein may phase transition at a concentration of about 5 mM to about 1 M, such as about 10 mM to about 500 mM, about 15 mM to about 250 mM, about 20 mM to about 150 mM, or about 25 mM to about 100 mM. In some embodiments, the fusion protein phase transitions at a concentration that is suitable for administration to a subject.

[0047] In addition, phase transition behavior may enable purification of the fusion protein using inverse transition cycling, thereby eliminating the need for chromatography. “Inverse transition cycling” refers to a protein purification method for polypeptides having phase transition behavior, and the method may involve the use of the fusion protein’s reversible phase transition behavior to cycle the solution through soluble and insoluble phases, thereby removing contaminants and eliminating the need for chromatography.

[0048] The dual agonist fusion protein can have a varying molecular weight. The molecular weight of the fusion protein can be measured by SDS-PAGE analysis, or other techniques known within the art. The fusion protein may have a molecular weight of about 50 kDa to about 100 kDa, such as about 55 kDa to about 95 kDa, about 60 kDa to about 85 kDa, about 65 kDa to about 80 kDa, or about 65 kDa to about 100 kDa. In some embodiments, the fusion protein has a molecular weight of greater than 50 kDa, greater than 55 kDa, greater than 60 kDa, or greater than 65 kDa. In some embodiments, the fusion protein has a molecular weight of less than 100 kDa, less than 95 kDa, less than 90 kDa, less than 85 kDa, or less than 80 kDa.

[0049] In some embodiments, the fusion protein comprises an amino acid sequence selected from the group consisting of (SEQ ID NO: 15), (SEQ ID NO: 17), (SEQ ID NO: 18), (SEQ ID NO:23), (SEQ ID NO:24) and variants thereof. In some embodiments, the fusion protein compnses an amino acid sequence selected from the group consisting of (SEQ ID NO: 15), (SEQ ID NO: 17), (SEQ ID NO: 18), (SEQ ID NO:23), and (SEQ ID NO:24).

[0050] In some embodiments, the fusion protein comprises an amino acid sequence selected from the group consisting of (SEQ ID NO: 15), (SEQ ID NO: 17), (SEQ ID NO: 18), and variants thereof. In some embodiments, the fusion protein comprises an amino acid sequence selected from the group consisting of (SEQ ID NO: 15), (SEQ ID NO: 17), and (SEQ ID NO: 18). In some embodiments, the fusion protein is (SEQ ID NO: 15), (SEQ ID NO: 17), or (SEQ ID NO: 18).

A. £lastin-like Polypeptide Domain

[0051] The ELP domain is a thermally responsive polypeptide that can instill phase transition properties to the fusion proteins. For example, the ELP domain has its own independent T t , which can be the same or different from that of the fusion protein. The T t of the ELP domain, the fusion protein, or both can be adjusted by varying the ammo acid sequence of the ELP domain, by varying the length of the ELP domain, or a combination thereof. In addition, the ELP domain is an unstructured polypeptide lacking secondary structure, which can provide a flexible linker between the two different receptor agonist domains.

[0052] The unattached ELP domain (e.g., not attached to either receptor agonist domain) may have a T t of about 20 °C to about 40 °C, such as about 25 °C to about 37 °C, about 26 °C to about 35 °C, or about 27 °C to about 32 °C. The unattached ELP domain may have a T t greater than 20 °C, greater than 21 °C, greater than 22 °C, greater than 23 °C, greater than 24 °C, or greater than 25 °C. The unattached ELP domain may have a T t less than 40 °C, less than 39 °C, less than 38 °C, less than 37 °C, less than 36 °C, or less than 35 °C.

[0053] The ELP domain can comprise a pentapeptide repeat sequence (VPGXG) n (SEQ ID NO: l), wherein X is any ammo acid except proline and n is an integer greater than or equal to 1 . In some embodiments, n is 2 to 200, 80 to 160, or 100 to 140. In some embodiments, n is 60,

120, or 180. In some embodiments, n is 120. In some embodiments, X is valine, alanine, leucine, or a combination thereof. Embodiments that include a combination of valine, alanine, and/or leucine can include these ammo acids at vary ing ratios. For example, X may he a ratio of vaime: alanine of 1 :0 to 10: 1. In some embodiments, X is a ratio of valine: alanine of 4: 1. In some embodiments, the ELP domain comprises (SEQ ID NO: 19). In some embodiments, the ELP domain is (SEQ ID NO: 19).

B. GLP-1 Receptor Agonist Domain [0054] GLP-1 receptor agonists refer to a class of agonists based on an endogenous ligand- receptor system that can mediate action of GLP-1. GLP1 is a 31 -ammo acid endogenous peptide released from the intestines post-prandially that can enhance glucose-stimulated insulin secretion from the pancreas. When administered as a drug, GLPl receptor (GLP1R) agonists can improve long-term glycenuc control as measured by glycated hemoglobin (HbAlc), can promote satiety and weight loss, and can improve b-ceil function. Furthermore, the msulmotropic effects of GLP1R agonists may decrease when glucose levels drop below a certain level, which can reduce the risk of hypoglycemia. Accordingly, the GLP-1 receptor agonist domain can be used to target multiple aspects of metabolic diseases, such as type 2 diabetes.

[0055] In some embodiments, the GLP-1 receptor agonist domain comprises an ammo acid sequence selected from the group consisting of:

AAHGEGTFTSDVSSYLEEQAAKEFIAWLVKGA (SEQ ID NOG);

GAHGEGTFTSD V S S YLEEQ AAKEFI AWLVKGA (SEQ ID NOG);

HGEGTFTSDVSSYLEEQAAKEFIAWLVKGA (SEQ ID NO: 4). and variants thereof.

[0056] In some embodiments, the GLP-1 receptor agonist domain comprises an amino acid sequence selected from the group consisting of: (SEQ ID NOG); (SEQ ID NOG); and (SEQ ID NO: I ).

[0057] The GLP-1 receptor agonist domain is located at the N-termina! end of the ELP domain in order to allow the N-terminal end of the GLP-1 receptor agonist domain to interact with its target. In some embodiments, the GLP-1 receptor agonist domain is attached to the N- terminus of the ELP domain.

C, FGF21 Receptor Agonist Domain

[0058] FGF21 refers to a metabolic hormone that can target the liver, pancreas, and/or adipose tissues to regulate insulin resistance and lipid metabolism. Accordingly, the FGF21 receptor agonist domain can be used to target multiple aspects of metabolic diseases, such as type 2 diabetes. In some embodiments, the FGF21 receptor agonist domain comprises an amino acid sequence selected from the group consisting of:

AYPIPDSSPLLQFGGQVRQRYLYTDDDQDTEAHLEIREDGTVVGAAHRSPESLLELKAL

KPGVIQILGVKASRFLCQQPDGALYGSPHFDPEACSFRERLLEDGYNVYQSEAHGLP LRL

PQKDSPNQDATSWGPVRFLPMPGLLHEPQDQAGFLPPEPPDVGSSDPLSMVEGSQGR SP

SYAS (SEQ ID NOG); AYPIPDSSPLLQFGGQVRQRYLYTDDDQDTEAHLEIREDGTVVGAAHRSPESLLELKAL

KPGVIQILGVKASRFLCQQPDGALYGSPHFDPEACSFRELLLEDGYNVYQSEAHGLP LRL

PQKDSPNQDATSWGPVRFLPMPGLLHEPQDQAGFLPPEPPDVGSSDPLSMVEPLQGR SP

SYAS (SEQ ID NO:6);

HPIPDSSPLLQFGGQVRQRYLYTDDAQQTEAHLEIREDGTVGGAADQSPESLLQLKALK

PGVIQ1LGVKTSRFLCQRPDGALYGSLHFDPEACSFRERLLEDGYNVYQSEAHGLPL HLP

GNKSPHRDPAPRGPARFLPLPGLPPALPEPPGILAPQPPDVGSSDPLSMVGGSQGRS PSYA

S (SEQ ID NO: 7);

HPIPDSSPLLQFGGQVRQRYLYTDDAQQTEAHLEIREDGTVGGAADQSPESLLQLKALK

PGVIQILGVKTSRFLCQRPDGALYGSLHFDPEACSFRELLLEDGYNVYQSEAHGLPL HLP

GNKSPHRDPAPRGPARFLPLPGLPPALPEPPGILAPQPPDVGSSDPLSMVGPSQGRS PSYA

S (SEQ ID NO: 8), and variants thereof.

[0059] In some embodiments, the FGF21 receptor agonist domain comprises an amino acid sequence selected from the group consisting of: (SEQ ID NO: 5), (SEQ ID NO:6), (SEQ ID NO: 7), and (SEQ ID NO: 8).

[006Q] The FGE21 receptor agonist domain is located at the C-terminal end of the ELP domain in order to allow the C-terminal end of the FGF2I receptor agonist domain to interact with its target. In some embodiments, the FGF21 receptor agonist domain is attached to the C~ terminus of the ELP domain.

3, Methods of Making the Dual Agonist Fusion Protein

[0061] Also disclosed are methods of making the dual agonist fusion proteins. The method can include transforming a bacteria with a recombinant expression vector comprising a first polynucleotide encoding disclosed fusion proteins, and culturing the transformed bacteria to express the disclosed fusion proteins. The culturing temperature of the bacteria can affect the yield of the fusion protein. In some embodiments, culturing is done at less than 37 °C In some embodiments, culturing is done at about 10 °C to less than 37 °C.

[0062] The first polynucleotide may also encode an alanine-alanine or a glycine-alanine leader sequence attached to the N-terminal end of the fusion protein, which can be cleaved off by, e.g., DPP4 during culturing and/or purification. The polynucleotide encoding the fusion protein can also be adapted to provide higher yields of the fusion protein. For example, the expression vector can further comprise a second polynucleotide encoding a translation initiation domain attached to the N -terminal end of the fusion protein. In some embodiments, the translation initiation domain is attached to the N-termmus of the fusion protein. The translation initiation domain can include a leader sequence and a protease cleavage site, where the protease cleavage site is located between the leader sequence and the fusion protein. In some

embodiments, the translation initiation domain comprises MSKGPGENLYFQGA (SEQ ID NO:20). In some embodiments, the translation initiation domain is (SEQ ID NQ:20).

A. Polynucleotides

[0063] Further provided are polynucleotides encoding the fusion proteins detailed herein. A vector may include the polynucleotide (e.g., first polynucleotide) encoding the fusion proteins detailed herein. To obtain expression of a polypeptide, one may subclone the polynucleotide encoding the polypeptide into an expression vector that contains a promoter to direct transcription, a transcription/translation terminator, and if for a nucleic acid encoding a protein, a ribosome binding site for translational initiation. An example of a vector is pet24. Suitable bacterial promoters are well known in the art. Further provided is a host cell transformed or transfected with an expression vector comprising a polynucleotide encoding a fusion protein as detailed herein. Bacterial expression systems for expressing the protein are available in, e.g., E coli, Bacillus sp , and Salmonella (Paiva et al, Gene 1983, 22, 229-235; Mosbach et al., Nature 1983, 302, 543-545), which is incorporated by reference herein in its entirety. Kits for such expression systems are commercially available. Eukaryotic expression systems for mammalian cells, yeast, and insect cells are well known in the art and are also commercially available.

Retroviral expression systems can also be used in the present disclosure.

4. Uses of the Dual Agonist Fusion Proteins

A. Compositions

[0064] Also disclosed are uses of the dual agonist fusion proteins. As mentioned above, the fusion protein has temperature dependent phase transition behavior. This can be used advantageously for drug delivery applications. For example, phase transition behavior may be used to form drug depots within a tissue of a subject for controlled release of the fusion protein. Accordingly , disclosed herein are compositions comprising a plurality of fusion proteins, wherein the plurality of fusion proteins assemble into an aggregate above the T t of the fusion protein. In addition, the composition can further comprise a biologically active agent that can be encapsulated upon the plurality of fusion proteins forming an aggregate. Examples of biologically active agents include, but are not limited to, msulm, sulfony lurea,

thiazolidinediones DPP-4 inhibitors, SGLT2 inhibitors, metformin, PPAR agonists, farnesoid X receptor agonists, and glucose-dependent insuiinotropic polypeptide in some embodiments the biologically active agent is selected from the group consisting of insulin, sulfonylurea, thiazolidinediones, DPP-4 inhibitors, SGLT2 inhibitors, metformin, PPAR agonists, farnesoid X receptor agonists, glucose-dependent insuiinotropic polypeptide, and a combination thereof

i. Administration

[0065] The composition comprising the plurality' of fusion proteins can be formulated for administration in accordance with standard techniques known to those skilled in the

pharmaceutical art. The composition may be prepared for administration to a subject and used in, e.g., methods of treating metabolic diseases as discussed below. Such compositions comprising the fusion protein can be administered in dosages and by techniques well known to those skilled in the medical arts taking into consideration such factors as the age, sex, weight, and condition of the particular subject, and the route of administration.

[0066] The composition comprising the plurality' · of fusion proteins can be administered prophylactical!y or therapeutically. In prophylactic administration, the compositions can be administered m an amount sufficient to induce a response. In therapeutic applications, the compositions can be administered to a subject in need thereof in an amount sufficient to elicit a therapeutic effect. An amount adequate to accomplish this is referred to as“therapeutically effective dose.” Amounts effective for this use will depend on, e.g., the particular composition of the fusion protein regimen administered, the manner of administration, the stage and severity of the disease, the general state of health of the patient, and the judgment of the prescribing physician

[0067] An effective amount of the composition comprising the plurality' of fusion proteins as described herein may be given in one dose, but is not restricted to one dose. The administration can be two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty, or more, administrations of the composition. Where there is more than one administration in the disclosed methods, the administrations can be spaced by time intervals of one minute, two minutes, three, four, five, six, seven, eight, nine, ten, or more minutes, by intervals of about one hour, two hours, three, four, five, six, seven, eight, nine, ten, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 hours, and so on. The

administrations can also be spaced by time intervals of one day, two days, three days, four days, five days, six days, seven days, eight days, nine days, ten days, 1 1 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, or combinations thereof.

The administration of the composition is not limited to dosing intervals that are spaced equally m time, but encompass doses at non-equal intervals, such as a priming schedule of administration at 1 day, 4 days, 7 days, or 25 days.

[0068] A dosing schedule of, for example, once/week, twice/week, three times/week, four times/week, five times/week, six times/week, seven times/week, once every two weeks, once every- three weeks, once every four weeks, once every five weeks, and the like, is available for the composition. The dosing schedules can encompass dosing for a total period of time, for example, one week, two weeks, three weeks, four weeks, five weeks, six weeks, two months, three months, four months, five months, six months, seven months, eight months, nine months, ten months, eleven months, or twelve months.

[0069] Provided are cycles of the above dosing schedules. The cycle can be repeated about, e.g., every seven days; every' 14 days; every' 21 days; every 28 days; every 35 days; every' 42 days; every 49 days; every 56 days; every' 63 days; every' 70 days; and the like. An interval of non-dosing can occur between a cycle, where the interval can be about, e.g., seven days; 14 days; 21 days; 28 days; 35 days; 42 days; 49 days; 56 days; 63 days; 70 days; and the like.

[0070] The composition comprising the plurality of fusion proteins may further include a pharmaceutically acceptable carrier. As used herein, "pharmaceutical acceptable carrier" refers to a physiologically acceptable diluent including, but not limited to water, phosphate buffered saline, or saline. Acceptable carriers, excipients, or stabilizers are nontoxic to subjects at the dosages and concentrations employed, and can include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid, BHA, and BHT; low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin or immunoglobulins; hydrophilic polymers, such as polyvinylpyrrolidone, ammo acids such as glycine, glutamme, asparagine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrms; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming counter-ions such as sodium; and/or nonionic surfactants such as Tween, Pluronics, or PEG. The compositions including a pharmaceutically acceptable carrier optionally may be sterile. The compositions may be frozen or lyophilized for storage and reconstituted in a suitable sterile carrier prior to use. The compositions having a pharmaceutically acceptable carrier can be generated in accordance with conventional techniques described in, e.g., Remington : The Science and Practice of Pharmacy, 21st Edition, Lippincott Williams & Wilkins, Philadelphia, PA (2001), which is incorporated by reference herein in its entirety.

[0071] The composition comprising the plurality' of fusion proteins can be administered by methods known in the art as described in Donnelly' et al. (Ann. Rev. Immunol. 1997, 15, 617- 648); Feigner et al. (U.S. Patent No. 5,580,859, issued Dec. 3, 1996); Feigner (U.S. Patent No. 5,703,055, issued Dec. 30, 1997); and Carson et al. (U.S. Patent No. 5,679,647, issued Oct. 21, 1997), ail of which are incorporated by reference herein in their entirety. The composition comprising the plurality of fusion proteins can be delivered via a variety of routes. Typical delivery routes include parenteral administration, e.g., intradermal, intramuscular or

subcutaneous delivery'. Other routes include oral administration, intranasal, intravaginal, transderma!, intravenous, intraarterial, intratumoral, intraperitoneal, and epidermal routes. In some embodiments, the composition is administered intraperitoneal! y or subcutaneously to the subject.

[0072] The composition comprising the plurality' of fusion proteins according to the present disclosure may also be administered with one or more additional drugs/compounds. Methods for co-administration with an additional drug/compound are well known in the art (Hardman, et al. (eds.) (2001 ) Goodman and Gilman’s The Pharmacological Basis of Therapeutics, 10th ed , McGraw-Hill, New York, N. Y.; Poole and Peterson (eds.) (2001) Pharmacotherapeutics for Advanced Practice: A Practical Approach, Lippincott, Williams & Wilkins, Phi la., Pa.; Chabner and Longo (eds.) (2001) Cancer Chemotherapy and Biotherapy, Lippincott, Williams & Wilkins, Phila. , Pa.), all of which are incorporated by reference herein in their entirety.

B. Methods of Treating Metabolic Diseases

[0073] Also disclosed herein are methods of treating metabolic diseases using the compositions and fusion proteins. The method of treating a metabolic disease in a subject in need thereof can include administering to the subject an effective amount of the composition comprismg the plurality of fusion proteins. The above description of the fusion protein and compositions thereof can also be applied to the methods of treating metabolic diseases.

[0074] Examples of metabolic diseases include, but are not limited to, obesity, type 2 diabetes mellitus, pancreatitis, dyslipidemia, nonalcoholic fatty liver disease (NAFLD), nonalcoholic steatohepatitis (NASH), insulin resistance, hyperinsulinemia, glucose intolerance,

hyperglycemia. In some embodiments, the metabolic disease is selected from the group consisting of obesity, type 2 diabetes mellitus, pancreatitis, dyslipidemia, nonalcoholic fatty liver disease (NAFLD), nonalcoholic steatohepatitis (NASH), insulin resistance, hyperinsulinemia, glucose intolerance, hyperglycemia, and a combination thereof. In some embodiments, the metabolic disease is type 2 diabetes mellitus.

[0075] As mentioned above, the composition can comprise a plurality of fusion proteins that can form an aggregate above the T t of the fusion proteins. For example, when the fusion protein has a T t below body temperature, it can form an aggregate upon administration to the subject.

The aggregate formation can allow the fusion protein to be released over an extended period of time, such as for about 12 hours to about 1 year following administration. In some embodiments, the composition releases the fusion protein following administration for greater than 1 day, greater than 3 days, greater than 10 days, greater than 1 month, or greater than 6 months. In some embodiments, the composition releases the fusion protein following administration for less than 1 year, less than 9 months, less than 6 months, less than 1 month, or less than 2 weeks. In some embodiments, the composition releases the fusion protein following administration for greater than 3 days.

[0076] The fusion protein and compositions thereof can be used advantageously m the treatment of metabolic diseases, such as type 2 diabetes mellitus. For example, administration of the composition can result in the subject having at least one of decreased blood glucose level, decreased body fat, increased insulin production, decreased hemoglobin Al e values, decreased circulating fatty acids, decreased liver fat content, decreased liver inflammation, and/or decreased liver fibrosis compared to a subject not receiving the administration of the

composition. In some embodiments, the administration of the composition can result in the subject having an about 10% to about 90% decrease in blood glucose levels compared to a subject not receiving the administration of the composition. The decrease in blood glucose levels can be over a course of 1 to 7 days following administration. In some embodiments, the administration of the composition can result in the subject having an about 1% to about 25% decrease in body fat compared to a subject not receiving the administration of the composition. The decrease m body fat can be measured within 1 to 7 days following administration.

[0077] The above description for administration of the fusion proteins, aggregates thereof, and compositions thereof can also be applied to the methods of treating metabolic diseases.

Expression of Dual Agonist Fusion Proteins

Materials & Methods

[0078] Expression Vector Synthesis: The nucleotide sequence encoding the 182 amino acid murine wild type FGF21 protein, minus the signal peptide, was codon optimized for E. coli expression and ligated into a pET~24a+ vector modified for seamless fusion of genes. Point mutations for amino acid substitutions L99R, P172G, and L173S were introduced to enhance protein stability, and the mutated Fgf21 gene was fused at the 5’ end to a gene encoding an ELP, following a previously reported seamless cloning strategy. The ELP of 120 repeats of a (Val- Pro-Gly-Xaa-Gly) pentapeptide - where X aa signifies a 4: 1 ratio of VabAla - and the final vector encoded the polypeptide fusion“ELP-FGF21

[0079] The analog included GLP-1 (7-37) with A8G, G22E, R36A amino acid substitutions, as well as an AA leader at the N terminus to enable activation through cleavage by Dipeptidyl- Peptidase 4 (DPP4). Following a similar process as for Fgf21, the nucleotide sequence encoding the resulting 32 ammo acid GLP-1 peptide was codon optimized for E. coli expression, ligated into the modified pET-24a+ vector, and fused at the 3’ end to the gene encoding the ELP described above. The final vector encoded the polypeptide fusion“GLP1-ELP”

[0080] For synthesis of the vector encoding the GLP-l /FGF21 dual agonist drug, the gene encoding GLP1-ELP was fused at the 3’ end to the mutated Fgf21 gene following a seamless cloning strategy (McDaniel, J.R., J.A. MacKay, F.G. Quiroz, and A. Chilkoti, Recursive Directional Ligation by Plasmid Reconstruction Allows Rapid and Seamless Cloning of

Oligomeric Genes. Biomacromolecules, 2010. 1 1 (4): p. 944-952, which is incorporated by reference in its entirety), and the final vector encoded the polypeptide fusion referred to as “GLP 1 -ELP-F GF21.”

[0081] Protein Expression & Purification: The GLP1 analog includes GLP1 (7-37) with A8G, G22E, R36A amino acid substitutions, as well as an AA leader at the N terminus to enable activation through cleavage by DPP4. Following a similar process as described for assembling the ELP-FGF21 -encoding vector, the nucleotide sequence encoding the 32 amino acid GLP1 peptide was codon optimized for E. coli expression, ligated into the modified pET-24a+ vector, and fused at the 3’ end to a gene encoding an ELP. The resulting vector encoded a GLP1-ELP polypeptide fusion, with the ELP varying depending on (VPGX aa G) pentapeptide repeats and the X aa guest residue composition. The gene encoding each GLP 1 -ELP construct was then fused at the 3’ end to the mutated Fg£21 gene. The final vector encoded the polypeptide fusions referred to as“GLP1-ELP-FGF21.”

[0082] GLP 1-ELP-FGF21 -encoding expression vectors were transformed into either SI I u file cells or Ultra BL21 (DE3) cells. Expression and purification were carried out as described for ELPioo % vai ,60 -FGF21 with the following exception: pre~ and post-induction temperatures were experimentally varied. The final purified fusion products were visualized on Coomassie-stained SDS-PAGE gels. For production of the GLP I~ELp4o %Aia .i 60 control, the ELP-encoding expression vector was transformed into Ultra BL21 (DE3) cells and expression and ITC purification were carried out.

[0083] A starter culture containing 50 niL of 55 g/L, terrific broth (TB) plus 250 mM kanamyein was inoculated and grown overnight at 37°C with orbital shaking at 250 rpm. The starter culture was centrifuged, resuspended in TB, and used to inoculate three l-L volumes of TB plus kanamyein in 6 L Erienmeyer flasks. The flasks were cultured at 30°C with orbital shaking at 200 rpm until they reached an ODeoo of 2.0. Protein expression was then induced by addition of 250 mM IPTG. The culturing temperature was reduced to 16°C, and growth was allowed to proceed for an additional 1 8 h.

[0084] Bacterial cultures were centrifuged at 4°C for 10 min at 3365 ref and resuspended in cold PBS. Cell membranes were disrupted via sonication (Q500 somcator, QSonica, Newtown, CT), and pulsed at 10 s on and 40 s off for a total sonication tune of 90 s. DNA was precipitated by addition of 10% polyethylenimme, and cell lysates were separated into soluble and insoluble fractions by centrifugation at 4°C for 10 mm at 23,645 ref. The soluble fraction was brought to room temperature, and the ELP fusion protein was purified from solution by ITC. In this process, the phase transition of the ELP fusion protein was triggered by addition of 0.2 M (NIL^SCL, producing a turbid suspension due to coacervation of the ELP fusion. The suspension was centrifuged at 25°C for 15 min at 23,426 ref; this step is referred to as a“hot spin.” The supernatant was discarded and the pellet was resolubilized in PBS at 4°C with 25 rpm gentle rotation (R4045 Roto-Bot Programmable Rotator, Benchmark Scientific, Sayreville, NJ). The resulting solution was centrifuged at 4°C for 5 min at 18,407 ref to pellet insoluble contaminants, and the supernatant was reserved; this step is referred to as a“cold spin.”

[0085] The ITC process was repeated by warming the solution to room temperature, adding (MLi^SCL to trigger the phase transition, centrifuging at 25°C for 8 min at 18,407 ref to pellet the ELP fusion protein, resolubilizing the pellet in PBS at 4°C with gentle rotation, and centrifuging at 4°C for 5 mm at 18,407 ref. Three total rounds of ITC were necessary to isolate the fusion from contaminants, and final products were visualized on a Coomassie- or CuCl 2 - stained SDS-PAGE gel.

Results

[0086] The GLP1/FGF21 dual agonist was designed as a head-to-taii polypeptide fusion protein, with GLP1 located at the N terminus and FGF21 at the C terminus (FIG. 1 ). This orientation provided GLP1 a solvent-exposed N terminus and FGF21 an exposed C terminus, both important for activating their respective receptors, while the linear architecture enabled facile synthesis and scale-up in a bacterial expression system. Between GLP1 and FGF21 was fused an ELP to serve a two-fold role as both a linker and a circulation-enhancing scaffold. GLP1-ELP-FGF21 fusions employed mutations in FGF21 to promote protein stability and amino acid substitutions in GLP1 to stabilize the alpha helix and protect from proteolytic cleavage. GLP1 also incorporated a di-alanine leader to facilitate recombinant expression before endogenous removal by DPP4 to expose an active N terminus. A library of dual agonist constructs was assembled in which the ELP varied in length and composition (Table 1) - with the goal of producing fusion proteins of variable Ls to allow for identification of constructs with optimal properties for in vivo depot formation.

[0087] Expression yields of GLP1-ELP-FGF21 fusions were poor (Table 1). We have observed that recombinant expression of fusion proteins with GLP1 at the N terminus have low'

_99_ yields at reduced culturing temperatures (< 37°C). However, reduced temperatures are important when expressing fusion proteins incorporating FGF21 to promote proper protein folding. Thus, we were left with the option of varying the ELP composition and the bacterial strain and testing resulting expression levels. The best yields were observed for the GLP1 -ELP-FGF21 fusion incorporating ELP 20%Ai a ,i 2o. GLPl -ELP 2 o%Aia ,i2 o-FGF21 was produced in SHuffle cells at reduced culturing temperatures and purified by the non-chromatographic ITC purification method. A 72 kDa band associated with the full-length fusion product was visible by SDS-PAGE throughout the purification process, and ITC purification alone was sufficient to isolate the dual agonist from contaminants.

Table 1. GLP1-ELP-FGF21 construct compositions, culturing conditions, and associated expression yields.

[0088] GLP1 expresses on a wide variety of ELPs, however expression yields of GLP1 -ELP are universally lower compared to ELP-FGF21. GLP1-ELP yields are further diminished at the reduced culturing temperatures necessary for proper FGF21 folding. Meanwhile, ELP-FGF21 expresses in large quantities, but only with a selective number of ELPs. Thus, GLP1-ELP-FGF21 expressed best on one ELP and in ver small quantities. Nevertheless, a full-length dual agonist fusion protein was produced recombmantly and purified by ITC. GLPI -ELP 2 o%Aia.i 2 o-FGF21 was capable of potently activating both the GLPI and FGF21 receptors, therefore exhibiting dual agonism in vitro.

Increasing GLP1-ELP-FGF21 expression yields

Materials & Methods

[0089] Expression vector synthesis: The GLP1 analog consisted of GLP1 (7-37) with A8G, G22E, R36A amino acid substitutions. A leader was added to the N terminus of GLP1 comprised of an expression-enhancing sequence (MSKGPG) (SEQ ID NO:2l) followed by a TEV protease cleavage sequence (ENLYFQG) (SEQ ID NO:22), which was separated from GLPI by a single alanine. Cleavage by TEV protease should thereby leave a GA leader, which will then serve as a substrate for cleavage by DPP4. Oligonucleotides encoding the GLPI with the N-terminal leader were codon optimized for E. coli expression, annealed, and ligated into the modified pET-24a+ vector. The gene encoding Leader-GLPl was fused at the 3’ end to the gene encoding

ELP20%Aiaj 20 -FGF21 following the previously reported seamless cloning strategy, and the final vector encoded the polypeptide fusion“Leader-GLPl ~ELP 2 o %At a ,i20 -FGF21” - or simply “Leader-GLP-ELP-FGF2l”

[0090] Protein expression and purification: Leader-GLPl -ELP-FGF21 was expressed and purified as described for leaderless GLPI -ELP-FGF21 , however Leader-GLPl -ELP-FGF21 was resuspended after its final round of ITC in TEV protease reaction buffer, substituting 3 mM glutathione for dithiothreitol. The final fusion product was visualized on a CuCl 2 -stained SDS- PAGE gel. Conventional GLPI -ELP-FGF21 (expressed without a translation-enhancing leader) and GLP1 -ELP were produced as described above.

[0091] Removal of the translation-enhancing sequence: Leader-GLPl -ELP-FGF21 was incubated for 16 h at 4°C with TEV protease (Sigma) at a 1 : 100 proteaseLeader-GLPl-ELP- FGF21 mass ratio. After the reaction, the phase transition of the ELP was triggered by addition of 0.2 M (NELhSCL and the resulting suspension containing immiscible GLPI -ELP-FGF21 , as well as miscible cleaved Leader and TEV protease was centrifuged 15 min at 25°C. The pellet containing GLP1-ELP-FGF21 was resuspended in PBS.

[0092] cAMP production assay: Following removal of the Leader from GLP1 -ELP-FGF21, GLP1 in vitro activity was measured through the cell-based assay described herein. As with fusions employing the previous GLPl analog version, the dual agonist was first incubated overnight at 4°C with DPP4 at a 1 :500 DPP4:GLP1 molar ratio to expose an active N terminus. This time, however, the intended cleavage substrate was a GA leader instead of the AA leader. Cells were treated with serial dilutions of GLP1-ELP-FGF21 (after removal of the expression enhancing sequence), conventional GLP1-ELP-FGF21 (expressed without a translation- enhancing leader), GLPl -ELP, or native human GLPl (7-37) (ProSpec) and resulting luminescence was measured as previously described. Data were fit to a three-parameter dose- response curve to determine ECsos.

Results

[0093] Design and production of expression-enhanced GLP1-ELP-FGF21 : A vector was assembled that encoded the dual agonist along with a leader sequence that served three purposes: 1) enhance expression, 2) allow for removal of the expression enhancer by protease cleavage, and 3) allow for removal of the protease cleavage scar by endogenous DPP4.

[0094] Expression of the dual agonist was robustly enhanced by addition of the MSKGPG leader sequence. A 73 kDa band associated with Leader-GLPl -ELP-FGF21 was visible in the bacterial cell lysate and persisted in the soluble fractions, with final protein yields surpassing 50 rng/L following ITC purification.

[0095] Expression-enhanced GLP1-ELP-FGF21 is partially active: Approximately 4 rng purified Leader-GLPl -ELP-FGF21 was reacted with TEV protease overnight for removal of all leader sequence residues except for glycine-alanine (FIG. 2). The ELP phase change was then triggered by addition of salt, and the dual agonist was spun out of the reaction and resuspended. Leader-GLPl -ELP-FGF21 was then incubated overnight with DPP4 for removal of the last two residues to expose the N-terminal histidine on GLP1, and GLP1R agonism was assayed in vitro. As observed previously, fusion of GLP1 to an ELP or to ELP-FGF21 increased the EC so -10- fold (3.1 ± 0.5 pM compared to 29.5 ± 5.0 pM for GLPl -ELP 2 o %Aia,i 2o and 23.9 ± 5.7 pM for conventionally expressed dual agonist). Unfortunately, the incorporation and removal of an expression-enhancing leader increased the dual agonist ECso yet an additional order of magnitude (212 ± 46 pM), suggesting incomplete leader removal.

[0096] Described here was a proof-of-concept study to investigate the possibility of improving dual agonist yields. We were successful in robustly increasing expression, with product yields increasing from 5 to 50 mg/L, which is a quite meaningful difference in the field of recombinant protein production. With optimization, we should be able to increase the efficiency of leader sequence removal to restore full GLP1 receptor binding capability. One disadvantage of this design is that both cleaved and uncleaved Leader-GLP1-ELP-FGF21 are spun out of solution and retained after the reaction. Thus, any dual agonist that failed to successfully react with TEV protease - and is therefore incapable of binding the GLP1R - will be carried downstream to the DPP4 reaction and the GLP1 activity assay where it will manifest as an increase in EC so. The TEV protease reaction must therefore be optimized for near 100% cleavage efficiency, which will require experimenting with different ratios of protease:Leader- GLP1-ELP-FGF21. It should be noted that the TEV protease employed in this pilot study may have been only partially functional.

In vitro Characterization of Dual Agonist Fusion Proteins

Materials & Methods

[0097] Endotoxin purification and testing: All proteins were endotoxin-purified using Acrodise Units (Pali Corporation, Port Washington, NY), and resulting endotoxin levels were tested using the Endosafe Nexgen-PTS spectrophotometer (Charles River Laboratories,

Wilmington, MA).

[0098] ERK phosphorylation assay: For quantitative evaluation of FGF21 in vitro activity, a HEK293 cell line was previously generated that stably expresses murine b-Klotho and FGF receptor 1 and thereby enables FGF2l-mediated ERK 1/2 phosphorylation. Cells were seeded at 5 x 10 4 cells/cm 2 and adhered overnight. After serum starvation for 6 h, cells were treated with serial dilutions of FGF21 -containing fusion proteins or native mouse FGF21 (ProSpec-Tany,

East Brunswick, NJ) for 5 min. Cells were lysed and assessed for phospho-ERKl/2 and total ERK 1/2 content using the AlphaLlSA Surefire Ultra Assay Kits (PerkinElmer) and the EnSpire Alpha Plate Reader (PerkinElmer). Phospho-ERKl/2 was normalized to total ERK1/2 and fit to a three-parameter dose-response curve to determine ECsos using GraphPad Prism 8 software (La Jolla, CA).

[0099] cAMP production assay: GLP-1 in vitro activity was quantified by a cell-based assay that utilizes a PIEK293 ceil line that stably expresses the GLP-1 R and a cAMP-inducible luciferase reporter. Cells were seeded at 1 x 10 5 cells/cm 2 into 96 well plates and adhered overnight. Concurrently, GLP-1 -containing fusions were incubated overnight at 4°C with DPP4 (ProSpec) at a 1 :500 DPP4: GLP-1 molar ratio to cleave the AA leader and expose the active N terminus of GLP-1. In the morning, cell media was replaced with induction buffer (129 mM NaCl, 4.8 mM KCl, 1.2 mM MgSOy 1.2 mM KFLPGy 2.5 mM CaCb, 5 mM NaHCOs, 10 mM HEPES, 0.5% BSA, and 50 mM 3 -isobutyl- 1-methylxanthine). Cells were treated with serial dilutions of GLP- 1 -containing fusion proteins or native human GLP-1 (7-37) (ProSpec) m induction buffer for 5 h, at which point supernatants were removed and replaced with Bright-Glo (Promega, Madison, WI). Luminescence of the supernatant samples were measured on a Victor X3 Plate Reader (Perkin Elmer) and normalized to zero drug treatment control wells. Data were fit to a three-parameter dose-response curve to determine ECsos using GraphPad Prism 8 software.

[00100] Phase behavior characterization: The lower critical solution temperature (LCST) phase transiti on behavior of ELP fusion proteins was evaluated by monitoring the OD 350 of solutions in PBS as a function of temperature on a Cary 300 UV-visible spectrophotometer equipped with a multicell thermoelectric temperature controller (Agilent Technologies, Santa Clara, CA). Heating and cooling were set to a rate of l°C/min. The T t was defined as the temperature at which the optical density reached 50% of its maximal value.

Results

[00101] GLP1-ELP-FGF21 has dual agonism at the GLP-1 and FGF21 receptors: The half maximal effective concentration (ECso) for the FGF21 and GLP-1 dual agonist components were measured using in vitro activity assays in cells stably expressing either the GLP-1 R or the FGF21 receptor complex. Fusion of GLP-1 to an ELP increased GLP-1 R ECso approximately ten-fold (FIG. 3 A), while the GLP-1 R ECso of GLP1-ELP-FGF21 (23.9 ± 5.7 pM) was not different from that of GLP1-ELP (29.5 ± 5.0 pM). Fission of FGF21 to an ELP increased the FGF21 receptor ECso approximately twenty-fold (FIG. 3B). The dual agonist had a marginally greater FGF21 receptor EC50 (43.2 ± 8.4 nM) compared to ELP-FGF21 (18.8 ± 4.5 nM), but this difference was not statistically significant (p > 0.05). Together, these data demonstrate that simultaneous presentation of GLP-1 and FGF21 on an ELP does not significantly impact the activity of each drug. See also FIG 1 1 for agonist activity.

[00102] Phase transition behavior of GLP1-ELP-FGF21 is suitable for depot formation: The LCST phase transition behavior of the GLP1-ELP-FGF21 fusion was evaluated by monitoring the optical density of a fusion protein solution as a function of temperature, defining the Ί\ as the temperature at which the solution becomes turbid. A target T t range between 27°C and 32°C was chosen to be suitable for depot formation - triggered by body heat - with ELP-drug release kinetics appropriate for once weekly dosing. An ELP fusion with a T t < 27°C forms an excessively stable coacervate that exhibits poor drug absorption, while a fusion with a T t near 35°C (the temperature of the s.c. space) exhibits a bolus-like release profile. GLP1-ELP-FGF21 was confirmed to have LCST phase change behavior, with a T t between 27 and 29°C at the injection-relevant concentration range of 100-200 mM (FIG. 3C). The T t was concentration- dependent (FIG. 3D), and the dual agonist phase change behavior was reversible (FIG. 3E). The reversibility and inverse dependence of T t on fusion protein concentration are attributes to the controlled release capabilities of ELP-based drug depots: as fusion unimers at the depot margin are diluted, their T t rises above body temperature, thereby reversing the LCST phase transition and allowing release of ELP-drug molecules from the coacervate.

in vivo Characterization of fJoal Agonist r us son Frotems

Materials & Methods

[00103] Animals: In vivo studies were conducted in accordance with the A A AL AC-accredited Duke Institutional Animal Care and Use Committee. 5-week-old B6.BKS(D)-Lepr db /J (“db/db”) male mice were purchased from Jackson Laboratory (Bar Harbor, ME) and maintained on a 12h/12h light/dark cycle with ad libitum access to food (LabDiet 5053) and water. Animals were group-housed and allowed 1-week acclimation before stratification into control and treatment groups based on baseline ambient blood glucose levels and body weights.

[00104] ELP fusion treatments: ELP fusion proteins were administered at 150-200 mM concentration via injection into the subcutaneous (s.c.) space on the hmd flank. Animals received either a single s.c. injection for short-term studies or weekly s.c. injections for chronic studies. The“1 : 1 mixture” treatment group received a single injection containing an equimolar mixture of GLP1-ELP and ELP-FGF21, with the indicated dose referring to the dose of each respective fusion in the mixture. Vehicle control refers to PBS.

[00105] Ambient blood glucose measurements: Blood glucose levels were measured from a tail vein puncture with an AlphaTRAK 2 Blood Glucose Meter (Zoetis, Parsippany-Troy Hills, NJ) every- 24 h, 1-2 h following onset of the light cycle. Data are presented as either raw values, or as a magnitude change from a mean baseline established from three independent measurements collected prior to treatment. Areas under the curve (AUCs) were calculated with GraphPad Prism 8 software using the trapezoidal rule and setting a Y=0 baseline. Treatment group AUCs were normalized to the vehicle-treated group where indicated.

[00106] Glucose tolerance test: Animals were fasted at the onset of the light cycle for 5 h with ad libitum access to water, then injected via mtraperitoneal (i.p.) with 0.75 g/kg D-glucose.

Blood glucose levels were measured from a tail snip with a Contour Blood Glucose Meter (Bayer, Leverkusen, Germany) at t=0 (before glucose injection), 10, 20, 30, 60, 90, and 120 min. AUCs were calculated as described for ambient blood glucose measurements.

[00107] Extended fast blood glucose measurements: Animals were fasted for 16 h starting at 4:00 PM with ad libitum access to water, after which blood glucose levels were measured from the tail vein as described for ambient blood glucose measurements.

[00108] Blood parameters: 50 JIL blood samples were collected from the tail veins following a 6 h fast using Mierovette EDTA coated tubes (Sarstedt), which were placed on ice until centrifugation at 5000 ref at 4°C for 15 min. Plasma supernatants were stored at -80°C until time of assay. Plasma insulin levels were quantified using the Mercodia Mouse Insulin ELISA, according to the manufacturers’ instructions. Percent glycosylated hemoglobin (%HbAlc) was measured from whole blood using a DCA Vantage Analyzer (Siemens).

[00109] Food Intake: For food intake studies, animals were group-housed 4 per cage, and food pellets were weighed every 1 -2 days. Total cage food intake was calculated for each time increment, averaged per animal, and summed over the course of the study to yield a cumulative food intake per mouse. N=8 treatment groups were divided into 2 cages, allowing for an n=2 food intake standard error calculation. 90 [00110] Pharmacokinetic studies: Tyrosine residues on GLP1-ELP, ELP-FGF21, or GLP1 - ELP-FGF21 fusion proteins were reacted with Xa l25 i radionuclide (Perkin Elmer) using Pierce Pre-Coated lodination Tubes (Thermo Fisher Scientific) and the indirect method for iodmation. Radiolabeled protein was purified from unreacted radionuclide with Zeha Spin Desalting Columns (Thermo Fisher Scientific). Activities of radiolabeled constructs were measured with an Atomlab 400 Dose Calibrator (Biodex, Shirley, NY) and correlated to protein concentration.

Mice received a single s.c. injection of radiolabeled fusion, and 10 pL blood samples were collected at frequent time points from the tail vein and stored at room temperature until radioactivity quantification. Sample counts were measured at the end of the study on a Wallac Wizard 1480 Automatic Gamma Counter (Perkin Elmer). An activity vs. count standard curve was used to convert sample counts to activities, and subsequently to moles of drug.

[00111] Pharmacokinetic analysis: The maximum serum concentration (C max ) was recorded as observed, as well as time to reach C max (t max )· AUC was estimated utilizing a serum

concentration of 0 nM at time zero and extrapolated to 16 days post-administration based on a linear regression curve fit to the terminal portion of the log serum concentration vs. time curve. Absorption half-life (ti / 2. ; * s ) was estimated from the slope of the linear regression curve. When a drug administered at an extravascular site yields a terminal half-life greater than that resulting from an i.v bolus, the terminal half-life reflects the absorption half-life.

[00112] Statistical analyses (used throughout the Examples]: Data are presented as means ± SEM. Data were analyzed by one-way ANOVA followed by Dunnett’s multiple comparisons tests, or by two-way repeated measures ANOVA followed by Dunnett’s tests. In vitro ECsos were compared by one-way ANOVA followed by Tukey’s tests.

Results

[00113] GLP1-ELP and ELP-FGF21 co-treatment has potent weight-lowering effects: To test our hypothesis that GLP-l and FGF21 act at least additively to control giycerma and inhibit weight gain, we carried out a short-term pilot study comparing a combination treatment of long- acting GLP-l and FGF21 analogs to each respective single-drug treatment. ELPs are repetitive peptide polymers characterized by a (VPGX aa G) n sequence, where“X aa ” is any amino acid besides proline, and“n” is the number of repeats. A notable feature of ELPs is their reversible lower critical solution temperature (LCST) phase behavior in aqueous media. ELPs have a distinct and tunable“transition temperature” (T t ); below the T t they are miscible m water, and above the T t they form a water-immiscible coaeervate, and their thermal responsiveness is retained when genetically fused to a peptide or protein drug. By manipulating the T t - via choice of the X aa residue and the molecular weight - an ELP-drug fusion can be designed to form an immiscible depot under the skm that steadily releases ELP fusions into systemic circulation. GLP1-ELP and ELP-FGF21 fusions are active in vitro and form subcutaneous (s.c.) depots capable of blood glucose-lowering effects for at least 5 days in diabetic mice following a single injection.

[00114] Db/db mice w r ere injected s.c. with GLP1-ELP, ELP-FGF21, an equimolar mixture of the two drugs, or vehicle control. Ambient blood glucose levels and body weights were measured 48 h post-injection and reported as a change from pre-injection baseline. Ail treatments significantly reduced blood glucose levels compared to vehicle, while combination treatment resulted in blood glucose levels that trended even lower than each respective single drug (FIG. 4A). Treatment with ELP-FGF21 or GLP1-ELP effectively inhibited weight gain (-0.9 ± 0.3% and -0.2 ± 0.6%) compared to vehicle-treated mice, who gained 2 7 ± 0.7% body weight in 48 h (FIG. 4B). In contrast, mice treated with the ELP-FGF21 + GLP1-ELP drug combination exhibited a robust 5.6 ± 0.6% reduction in body weight (FIG. 4B). Together these data suggest that GLP-l and FGF21 act at least additively to induce weight loss - and possibly to improve glyeemic control - in diabetic mice.

[00115] A GLP1 -ELP-FGF21 dual agonist fusion protein has sustained dose-dependent effects on body weight and glycemia: The dual agonist drug was tested for efficacy in diabetic mice.

The db/db mouse model was selected due to its extreme degree of hyperglycemia paired with obesity, as high baseline body weight and glyeemic levels would provide a larger window to identify potentially additive effects of dual agonism. Mice were treated with either a single GLP1-ELP-FGF21 s.c. injection at the indicated dose, or vehicle. Ambient blood glucose levels were measured daily, until all cohorts returned to baseline levels. Significant reductions in blood glucose vs. tune AUC were observed at the two highest doses tested (750 nmol/kg and 1000 nmol/kg) (FIG 5A), however, raw blood glucose vs. time data revealed that it was not effect size but rather effect duration that increased in a dose-dependent manner (FIG 5B). All doses reduced blood glucose levels from >300 mg/'dL to <150 mg/'dL (FIG 5B), with effects persisting 4 days at the lowest dose (250 nmol/kg), and 8 days at the highest dose tested (1000 nmol/kg) (FIG. 5B). Compared to vehicle control, all tested doses of GLP1-ELP-FGF21 had an inhibitory effect on weight gam, while the two highest doses tested induced weight loss - reducing body weights by up to 7.2 ± 2.3% (FIG. 5C). On day 7 post-treatment, a net weight loss effect persisted in the 750 and 1000 nmol/kg groups (-2.6 ± 3.3% and -2.7 ± 3.7%, respectively) (FIG. 5D).

[00116] To confirm that sustained efficacy was a result of prolonged drug in circulation - as is observed when an ELP depot forms at the site of injection - plasma drag levels w/ere measured over time following a single s.c. injection of GLP1-ELP-FGF21 to db/db mice. Aside from a modest burst release in the first 24 h, plasma drug levels remained steady near 100 nM out to day 10 (FIG. 5E), at which point drag levels decreased at a rate consistent with first-order elimination. When a linear regression curve was fit to the terminal portion of the data (Table 2), the absorption half-life was calculated to be 7.6 ± 1.1 days (Table 3). Interestingly, the data were described nearly as well by a zero-order elimination model (R 2 = 0.81) as by a first-order model (R 2 = 0.85) (Table 2). When pharmacokinetic data were analyzed alongside the 1000 nmol/kg blood glucose vs. time efficacy data (FIG. 5B), 100 nM appeared to be the minimal therapeutic concentration, as blood glucose levels returned to baseline in the same time frame that serum drug levels dropped below 100 nM (on or after day 10).

[00117] In summary, treatment of obese and hyperglycemic mice with a GLP-1/FGF21 dual agonist drug had potent and sustained effects on body weights and ambient blood glucose levels. A single injection was sufficient to maintain therapeutic drug levels and protect mice from hyperglycemia and weight gam for >7 days, demonstrating the suitability of a GLP1-ELP- FGF21 depot for a onee-weekly dosing scheme. A dose of 1000 nmol/kg was identified as yielding a maximal therapeutic effect and selected for further evaluation.

[00118] A GLP1 -ELP-FGF21 dual agonist confers greater giycernic control compared to a. long-acting GLP-1 RA: To elucidate the relative contribution of GLP-1 and FGF21 to the potent in vivo effects of the dual agonist, we next compared GLP1 -ELP-FGF21 with the GLP1-ELP and ELP-FGF21 single agonist monotherapies. An equimolar mixture of GLP1-ELP and ELP-FGF21 was included to understand the impact of incorporating both drugs into a single molecule.

[00119] To ensure consistency across treatments, the same ELP sequence was employed in each drug formulation. To do so, a vector encoding GLP-1 fused at its C terminus to the ELP of interest ((VPGXG) l2 o with a 4: 1 valine: alanine ratio at the X residue position) was expressed in E. coli and purified by 1TC. A 52 kDa band associated with the GLP1-ELP fusion was visible by SDS-PAGE following purification. The GLP1-ELP fusion was tested for GLP-1R agonism and exhibited a ten-fold increase in ECso compared to native GLP-l (FIG 6A). LCST phase transition behavior was evaluated for GLP1-ELP and an equimolar mixture of GLP1-ELP and ELP-FGF21, revealing T t s of 27.5°C and 28°C, respectively, at the injection-relevant concentration of 200 mM (FIG 6B and FIG. 6C). The I s w¾re concentration-dependent (FIG 6D and FIG. 6E), and the phase change behavior of each fusion/fusion mixture was reversible (FIG. 6F and FIG. 6G)). Note that the LCST phase transition behavior of ELP-FGF21 has been characterized previously (Gilroy, C.A., S. Roberts, and A. Chilkoti, Fusion of fibroblast growth factor 21 to a thermally responsive biopolymer forms an injectable depot with sustained anti- diabetic action. Journal of Controlled Release, 2018. 277: p. 154-164, which is incorporated by reference herein in its entirety).

[00120] Next, pharmacokinetic profiles were evaluated for the GLP1-ELP fusion, as well as the equimolar mixture of GLP1-ELP and ELP-FGF21. Both GLP1-ELP and the mixture exhibited steady plasma drug levels consistent with sustained release from an s.c. depot. The GLPl-ELP monotherapy depot behaved similarly to that of the dual agonist, releasing fusion molecules into circulation at a rate that could be described by a zero-order elimination model (Table 2), and resulting m plasma drug levels hovering near 100 nM for at least 10 days (FIG 7A). Interestingly, the 1 : 1 mixed depot released GLP1 -ELP and ELP-FGF21 fusion unimers at different rates (FIG. 7B). The ELP-FGF21 component reached a higher C um than the GLPl -ELP component (Table 3), and ELP-FGF21 plasma levels dropped below 100 nM by day 4, while GLPl-ELP plasma levels hovered steadily around 100 nM for 7 days. Though absorption of each component of the mixture fit a zero-order release model (Table 2), neither GLPl -ELP nor ELP- FGF21 maintained steady plasma levels for as long as GLPl -ELP-FGF21 , suggesting an advantage to delivering the two drugs as a single molecule.

[00121] As a further analysis of the GLPl-ELP control, ambient blood glucose levels were measured daily for 10 days, and treatment wath the GLPl-ELP fusion resulted in significant and sustained effects on giycemia, reducing levels from >300 mg/dL to <200 mg/dL for the duration of the study (FIG. 12A). Body weights were recorded daily and reported as a % change from pre- injection weight, and GLPl -ELP 20%Aia, 120 treatment significantly inhibited weight gam compared to control mice (FIG. 12B). Thus, GLP1-ELP fusion is fully functional - as should be the associated GLP1-ELP/ELP-FGF21 mixture.

[00122] After establishing the optimal dosing parameters and appropriate controls, the dual agonist was assessed for its effects on glycemic control. Dh/db mice were treated weekly for four weeks with 1000 nmol/kg GLP 1 -ELP-FGF21 , GLP1-ELP monotherapy, ELP-FGF21 monotherapy, or vehicle. Acute glycemic effects were evaluated through an intraperitoneal (i.p. ) glucose tolerance test (GTT) (0.75 mg/kg) performed 3 days after the first dosing cycle, when body weights were consistent across treatment groups (FIG. 8A). Fasting blood glucose and plasma insulin levels were measured prior to the glucose challenge (FIG. 9A). ELP-FGF21 monotherapy treatment significantly decreased GTT AUG compared to vehicle (FIG. 9B and FIG. 9C) with equivalent levels of fasting plasma insulin (FIG. 9A, right panel), suggesting increased insulin sensitivity upon FGF21 treatment. GLP1-ELP monotherapy outperformed ELP-FGF21 in improving glucose tolerance (FIG 9B and FIG. 9C), and the GLP1-ELP-FGF21 treatment was superior to both single agonist monotherapies. Fasting blood glucose levels were lowest in the dual agonist cohort (125 ± 6 0 mg/dL compared to 167 ± I I mg/dL for GLP1-ELP and 237 ± 21 mg/dL for ELP-FGF2I) (FIG. 9A, left panel), and GLP1 -ELP-FGF21 treatment significantly decreased GTT AUC compared to GLP1-ELP or ELP-FGF21 (FIG. 9B and FIG 9C). Whereas GLP1-ELP achieved its effects on glyeemia with a concomitant significant increase in fasting plasma insulin, the dual agonist achieved robust glycemic control without a significant elevation in insulin, indicating increased insulin sensitivity mediated specifically by the FGF21 component. Insulin sensitization is believed to be the primary mechanism by which FGF2I alters glyeemia, however other mechanisms may include increasing glucose uptake in adipose and muscle tissues, decreasing hepatic glucose output and inhibiting glucagon secretion.

Table 2 Regression fits for pharmacokinetic data.

Labeled construct Regression model

[00123] To assess long-term glycemic control, percent glycated hemoglobin Ale (%HbAlc) was measured prior to initiation of treatment (day 0) and at the termination (day 27) of the chronic dosing study. Both ELP-FGF21 and GLP1 -ELP monotherapies significantly reduced %HbA1 c elevation relative to control over the four-week study, though chronic GLP1-ELP treatment induced a +1.0 ± 0.2% rise from days 0 to 27 (FIG. 9D and FIG. 8B). The dual agonist cohort exhibited the greatest degree of long-term glycemic control, with a minimal +0.3 ± 0.2% change in %HbAlc (p < 0.07 compared to GLP1-ELP). Together these data demonstrate that treatment with the GLP1-ELP-FGF21 dual agonist drug affords superior glycemic control compared to equimolar dosing of a long-acting GLP-IRA. Furthermore, GLP-1 and FGF21 appear to act additively in improving glucose homeostasis through enhanced insulin secretion and insulin sensitivity associated with each respective component of the dual agonist.

Table 3. Pharmacokinetic parameters for ELP fusion protein depots following s.c. administration to mice.

Data are reported as means ± SEM. C max , observed maximum serum concentration; ax, time to Cmax; AUC, area under the curve; ti / 2 , abs , absorption half-life.

[00124] GLP-l and FGF21 act in combination to inhibit weight gain: Chronic dosing with the GLP1-ELP monotherapy significantly inhibited weight gain over the four- week treatment period compared to vehicle. The reduced rate of weight gain was likely due m part to a significant reduction in food intake by the GLP1-ELP cohort (FIG. 9F) - which is consistent with the anorectic effect observed upon GLP-l RA therapy. The dual agonist treatment also inhibited weight gain, and each treatment cycle resulted in a potent weight loss effect, with negative net weight gam values persisting through day 12 (FIG. 9E). Chronic dual agonist treatment resulted in significantly reduced weight gain compared to GLPI -ELP monotherapy (14.1 ± 1.6% vs. to 25.3 ± 1.9%), despite GLP1 -ELP-FGF21 -treated mice consuming chow at an equivalent rate to GLPI-ELP. Thus, the weight reducing effects of the dual agonist cannot be attributed to an enhanced anorectic effect, but instead point to a separate mechanism likely involving thermogenic action exclusive to the FGF21 component. It should he noted that ELP-FGF21 monotherapy did not induce a significant effect on body weight, indicating that cooperative action between FGF21 and GLP-l may be necessary for realizing the full weight loss effect.

[00125] Metabolic effects observed between the GLP1-ELP-FGF21 dual agonist and a GLP- 1/FGF21 single agonist mixture: To elucidate whether the metabolic effects of GLP-l /FGF21 combination therapy w¾re impacted by the drug format, a mixture of GLPI-ELP and ELP- FGF21 was included in the chronic dosing study for direct comparison to GLP1-ELP-FGF21. Changes in %HbAlc were consistent between cohorts, however the mixture group trended slightly higher than the dual agonist (+0.4 ± 0.2% compared to +0.3 ± 0.2%). Glyeemic control was further evaluated by a GTT performed 3 days following the final treatment. Fasting blood glucose levels were potently reduced upon GLP1-ELP-FGF21 treatment compared to vehicle (92.7 ± 8.3 mg/dL vs. 384.8 ± 33.2 mg/dL), while fasting levels in the co-therapy cohort trended slightly higher (1 11.4 ± 15.5 mg/dL) (FIG. 9G, left panel). Both combination therapy formats resulted in greatly improved glucose tolerance (FIG. 9H and FIG. 91), though the dual agonist cohort trended towards lower glucose peaks and faster recoveries to pre-injection baselines compared to a mixture of the two drugs. Body weight and food intake data for the co-therapy group nearly overlapped with those for the dual agonist (FIG. 9E, FIG. 9F, and FIG 8C).

Therefore, the dual agonist’s more robust glucose response could not be attributed to the indirect effect of decreased body weight but might be a result of elevated fasting insulin levels (FIG. 9G, right panel).

[00126] Although differences between the dual agonist and the single agonist mixture were not statistically significant, GLP1 -ELP-FG2F21 showed consistently superior pharmacological trends, effects that we hypothesize may prove to be significant in a larger cohort of animals, or when challenging with larger doses of glucose. These trends may be attributable to the dual agonist’s more consistent pharmacokinetic profile compared to the mixed depot, resulting in fewer peaks and valleys and thus maintaining therapeutic levels of both GLP-1 and FGF21 for a larger portion of each dosing cycle.

[00127] A GLP1-ELP-FGF21 dual agonist does not increase risk of hypoglycemia:

Administering GLP-1 and FGF21 in combination was not expected to pose a risk of

hypoglycemia, as each respective agonist has been shown have protective effects: FGF21 lowers glucose levels primarily by increasing insulin sensitivity, while GLP-1 stimulates insulin secretion only in the presence of elevated glucose. However, as a safety check, the dual agonist was tested for increased risk of hypoglycemia during prolonged fasting (nocturnal

hypoglycemia) and following recovery from a prandial glucose spike (reactive hypoglycemia). Db/db mice were treated with 1000 nmol/kg GLP1-ELP-FGF21 or vehicle, and ad hbitum-fed blood glucose levels measured 48 h post-treatment showed significant reductions in the dual agonist cohort (FIG. 10A). Mice were then subjected to an overnight 16 h fast, after which both cohorts maintained blood glucose levels above the traditionally defined threshold of hypoglycemia, 50 - 55 mg/dL, indicating that dual agonist- treated mice tolerated the extended fast. Returning to the GTT, an additional plasma insulin measurement was incorporated at t= : ! Q min to assess the glucose-stimulated insulin secretion capacity of dual agonist-treated mice. Plasma insulin levels during a glucose bolus were not significantly elevated in comparison to vehicle treatment (FIG. 10B), and during recovery from t=60 to t=T20 min post-challenge, blood glucose levels in the GLP1-ELP-FGF21 cohort returned to the pre-injection baselines, -100 mg/dL, without evidence of hypoglycemia. Thus, dual agonist treatment does not appear to increase susceptibility to reactive hypoglycemia due to excessive insulin secretion.

[00128] For reasons of completeness, various aspects of the invention are set out in the following numbered clauses:

[00129] Clause 1. A fusion protein comprising an elastin-like polypeptide (ELP) domain; a GLP-1 receptor agonist domain attached to a N-terminal end of the ELP domain; and a FGF21 receptor agonist domain attached to a C-termina! end of the ELP domain.

[00130] Clause 2. The fusion protein of clause 1, wherein the ELP domain comprises an amino acid sequence of (VPGXG) n (SEQ ID NO: l), wherein X is any amino acid except proline and n is 2 to 200.

[00131] Clause 3. The fusion protein of clause 2, wherein n is 80 to 160.

[00132] Clause 4. The fusion protein of clause 2 or clause 3, wherein X is valine, alanine, leucine, or a combination thereof.

[00133] Clause 5. The fusion protein of any of clauses 2-4, wherein X is a ratio of

valine: alanine of 1 :0 to 10: 1.

[00134] Clause 6. The fusion protein of any of clauses 2-5, wherein n is 120 and X is a ratio of valine: alanine of 4: 1.

[00135] Clause 7. The fusion protein of any of clauses 1 -6, wherein the GLP-1 receptor agonist domain comprises an amino acid sequence selected from the group consisting of (SEQ ID NO:3), (SEQ ID NO:4), (SEQ ID NO: 5), and variants thereof

[00136] Clause 8. The fusion protein of any of clauses 1-7, wherein the FGF21 receptor agonist domain comprises an ammo acid sequence selected from the group consisting of (SEQ ID NO: 6), (SEQ ID NO: 7). (SEQ ID NO:8), (SEQ ID NON) and variants thereof.

[00137] Clause 9. The fusion protein of any of clauses 1-8, wherein the fusion protein has a transition temperature (T t ) of about 25 °C to about 37 °C. [00138] Clause 10. The fusion protein of any of clauses 1-9, wherein the fusion protein has a molecular weight of about 50 kDa to about 100 kDa.

[00139] Clause 1 1. A composition comprising a plurality of fusion proteins according to any one of clauses 1-10, wherein the plurality of fusion proteins assemble into an aggregate above the T t of the fusion protein.

[00140] Clause 12. The composition of clause 11, further comprising a biologically active agent.

[00141] Clause 13. A method of treating a metabolic disease in a subject in need thereof, the method comprising administering to the subject an effective amount of the composition of clause 11 or clause 12.

[00142] Clause 14. The method of clause 13, wherein the metabolic disease is selected from the group consisting of obesity, type 2 diabetes meilitus, pancreatitis, dyslipidemia, nonalcoholic fatty' liver disease (NAFLD), nonalcoholic steatohepatitis (NASH), insulin resistance, hyperinsulinemia, glucose intolerance, hyperglycemia, and a combination thereof.

[00143] Clause 15. The method of clause 13 or clause 14, wherein the composition releases the fusion protein following administration for greater than 3 days.

[00144] Clause 16. The method of any of clauses 13-15, wherein administration of the composition results in the subject having at least one of decreased blood glucose level, decreased body fat, increased insulin production, decreased hemoglobin Ale values, decreased circulating fatty acids, decreased liver fat content, decreased liver inflammation, and decreased liver fibrosis compared to a subject not receiving the administration of the composition

[00145] Clause 17. A method of synthesizing a dual agonist fusion protein, the method comprising transforming a bacteria with a recombinant expression vector comprising a first polynucleotide encoding the fusion protein of any of clauses 1 -10; and culturing the transformed bacteria to express the fusion protein.

[00146] Clause 18. The method of clause 17, wherein the expression vector further comprises a second polynucleotide encoding a translation initiation domain attached to the N-terminal end of the fusion protein.

[00147] Clause 19. The method of clause 18, wherein the translation initiation domain comprises a leader sequence and a protease cleavage site, the protease cleavage site located between the leader sequence and the fusion protein. 148] Clause 20. The method of any of clauses 17-19, wherein culturing is done at less than °C.

i lienees

VPG VG VP (GVG VPG VG VP GVG VPG VG V P GVG V

P G VG VP G V G VP GVG VP GVG VP G V G VP G V G VP GVGVPGVGVPGVGVPGVGVPG VGVPGVGVPG V G VP GVG VPG V G VP GVG VPG V G VP G V G VPG V GVPGVGVPGVGVPGVGVPGVGVPGVGVPGVG VPGVGVPGVG VPG VGVPGVGVPG VGVPGVGV P GVG VP GVG VP GVG VP GVG VP GVG VP GVG VP GVGVPGVGVPGVGVPGVGVPG VGVPGVGVPG VGVPGVGVPGVGVPGVGVPGVGVPGVGVPGV GVPGVGVPGVGVPGVGVPGVGVPGVGVPGVG VPGVGVPGVG VPG VGVPGVGVPG VGVPGVGV PGVGVPGVGVPGVGVPGVGVPGVGVPGVGVP G V G VP G V G VP G V G VP G V GW G VG VP GVG VP G VG VPG VGVPGVGVPG VGVPGVGVPGVG VPGV GVPGVGVPGVGVPGVGVPGVGVPGVGVPGVG VPGVGVPGVGVPGVGVPGVGVPGVGVPGVGV PGVGVPGVGVPGAYPIPDSSPLLOFGGOVROR YLYTDDDQDfEAHLElREDGTWGAAHRSPE

SLLELKALKPGVIQILGVKASRFLCQQPDGAL YGSPHFDPEACSFRERLLEDGYNVYQSEAHG LPLRLPQKDSPNQDATSWGPVRFLPMPGLLH EPQDQAGFLPPEPPDVGSSDPLSMVEGSQGRS PSYASG

ELP 2 o %Ai3, s 2 o-FGF21 GVG VPG VGVPGAG VPG VGVPGVGVPG V G VPG 68.5

VG VP G AGVPGVG VP GVG VPGVG VP G V G VPG A (SEQ ID NO: 14) GVPGVGVPGVGVPGVGVPGVGVPGAGVPGVG VPGV G VPGVGVPGVG VPGAGVPGVGVPG VGV P G VG VP GVG VPG A G VP G VG VPG V G VP G V GYP GVGVPGAGVPGVGVPGVGVPGVGVPGVGVPG AGVPGVGVPG VGVPGVGVPG VGVPGAGVPGV GVPGVGVPGVGVPGVGVPGAGVPGVGVPGVG VPGV GVPGVGVPG AGVPG VGVPGV GVPGVGV P G VG VP GAG VPG VG VP GVG VPG V G VP GVG VP GAGVPGVGVPGVGVPGVGVPGVGVPGAGVPG VO VP G V G VPG VG VP GVG VPG AG VP G V G VPG V G VPGVG VP G V G VP G AGVPG V G VP G VG VPG VG VPGVGVPGAGVPGVGVPGVGVPGVGVPGVGV PGAGVPGVGVPGVGVPGVGVPGVGVPGAGVP GVGVPGVGVPGVGVPGVGVPG AGVPGVGVPG VO VP G V G VPG VG VP G A G VPG VG VP G V G VPG V GVPGVGVPGAGVPGVG VPGVGVPGVG VPGVG VPGAGVPGVGVPGVGVPGVGVPGVGVPGAGV

PGVGVPGVGVPGAYPIPDSSPLLOrGGOVROR

YLYTDDDQDTEAHLEIREDGTWGAAHRSPE

SLLELKALKPGVIQILGVKASRFLCQQPDGAL

YGSPHFDPEACSFRERLLEDGYNVYQSEAHG

LPLRLPQKDSPNQDATSWGPVRFLPMPGLLH

EPQDQAGFLPPEPPDVGSSDPLSMVEGSQGRS

VPGVGVPGVG VPG AGVPGVG VPG VGVPGVGV

PG VG VPG AG VPGVGVPGVG VPG V(3VPGVG VP

GAG GVGVPGVGVPGVGVPGVGVPGAGVPG

VGVPGVGVPGVGVPGVGVPGAGVPGVGVPGV

GVPGVGVPGVGVPGAGVPGVGVPGVGVPGVG

VPG V G VP GAG VPG V G VP G V G VPG VG VP G VG V

PGAGVPGVGVPGVGVPGVGVPGVGVPGAGVP

GVGVPGVGVPGVGVPGVGVPGAGVPGVGVPG

VGVPGVGVPGVGVPGAGVPGVGVPGVGVPGV

GVPGVGVPGAGVPGVGVPGVGVPGVGVPGVG

VPGAGVPGVGVPGVGVPGVGVPGVGVPGAGV

PGVGVPGVGVPGVGVPGVGVPGAGVPGVGVP

G VG VP G V G VP G VG VPG AG VP G VGVPGVG VP G

VGVPGVGVPGAGVPGVGVPGVGVPGVGVPGV

GVPGAGVPGVGVPGVGVPGVGVPGVGVPGAG

VPGVGVPGVGVPGAWgDSSPLLOTGGOVBO

RYLYTDDDQDTEAHLEIREDGTVVGAAHRSP

ESLLELKALKPGVIQILGVKASRFLCQQPDGA ,

AGVGVPGVGVPGAGVPGVGVPGVGVPGVGVP (SEQ ID NO: 16) GVGVPGAGVPGVGVPGVGVPGVGVPGVGVPG AGVPGVGVPGVGVPGVGVPGVGVPGAGVPGV GVPGVGVPGVGVPGVGVPGAGVPGVGVPGVG VPGVGVPGVGVPGAGVPGVGVPGVGVPGVGV PGVGVPGAGVPGVGVPGVGVPGVGVPGVGVP GAGVPGVGVPGVGVPGVGVPGVGVPGAGVPG VGVPGVGVPGVGVPGVGVPGAGVPGVGVPGV GVPGVGVPGVGVPGAGVPGVGVPGVGVPGVG VPGVGVPGAGVPGVGVPGVGVPGVGVPGVGV P G AG VP G VG VPGVG VP G VG VPG V G VP GAG VP GVGVPGVGVPGVGVPGVGVPGAGVPGVGVPG VG VP G V G VPG VG VP G A G VPG VG VP G V G VPG V GVPGVGVPGAGVPGVGVPGVGVPGVGVPGVG VPGAGVPGVGVPGVGVPGVGVPGVGVPGAGV PGVGVPGVGVPGVGVPGVGVPGAGVPGVGVP GVGVPGVGVPGVGVPGAGVPGVGVPGVGVPG VGVPGVGVPGAGVPGVGVPGVGVPGVGVPGV GVPGAGVPGVGVPGVGVPGVGVPGVGVPGAG VPG V G YP G VG VPG

Leader- MSKGPGKWLYFQGA&GEGTFTSBVSSYLEEQA 73.2

AKEFIAWLVKGAGVGVPGVGVPGAGWGVGV

GLP 1 -ELP2o%A!a,i2o- (SEQ ID NO: 17)

P G VG VP G V G VP G VG VP G A G VP G V G VP G V GYP FGF21

G VG VP G VG VP GAG VPG VG VP G VG VPG VG VP G VGVPGAGVPGVGVPGVGVPGVGVPGVGVPGA GVPGVGVPGVGVPGVGVPGVGVPGAGVPGVG VPG V G VP G VG VPG V G VP G AGVPGVG VP G VG V PGVGVPGVGVPGAGVPGVGVPGVGVPGVGVP GVGVPGAGVPGVGVPGVGVPGVGVPGVGVPG AGVPGVGVPGVGVPGVGVPGVGVPGAGVPGV GVPGVGVPGVGVPGVGVPGAGVPGVGVPGVG VPGVGVPGVG VPG AGVPGVG VPG VGVPGVGV

Underlined text designates residues associated with ELPs; bold text designates functional proteins/peptides (FGF21, TEV protease, GLP1); italicized text designates residues associated with linkers or leaders.