Prodrugs comprising an GLP-1/glucagon dual agonist linker hyaluronic acid conjugate

Information

  • Patent Grant
  • 10806797
  • Patent Number
    10,806,797
  • Date Filed
    Thursday, June 2, 2016
    8 years ago
  • Date Issued
    Tuesday, October 20, 2020
    3 years ago
Abstract
The present invention relates to a prodrug or a pharmaceutically acceptable salt thereof comprising an GLP-1/Glucagon agonist linker conjugate Z-L1-L2-L-Y—R20, wherein Y represents an GLP-1/Glucagon agonist moiety; and -L is a linker moiety—by formula (Ia), wherein the dashed line indicates the attachment to one of the amino groups of the GLP-1/Glucagon agonist moiety by forming an amide bond. The invention further relates to pharmaceutical compositions comprising said prodrugs as well as their use as a medicament for treating or preventing diseases or disorders which can be treated by GLP-1/Glucagon agonist.
Description
RELATED APPLICATIONS

This application is a 35 U.S.C. § 371 filing of International Patent Application No. PCT/EP2016/062496, filed Jun. 2, 2016, which claims priority to European Patent Application No. 15305858.1, filed Jun. 5, 2015, the entire disclosures of which are hereby incorporated herein by reference in their entirety.


The present invention relates to GLP-1/Glucagon dual agonist prodrugs, pharmaceutical compositions comprising said prodrugs as well as their use as a medicament for treating or preventing diseases or disorders which can be treated by a GLP-1/Glucagon dual agonist, for example in the treatment of disorders of the metabolic syndrome, including diabetes and obesity, as well as for reduction of excess food intake.


BACKGROUND OF THE INVENTION

GLP-1 Agonists


Exendin-4 is a 39-amino acid peptide, isolated from the salivary secretions of the venomous Gila monster (Heloderma suspectum). It has some sequence similarity to several members of the glucagon-like peptide family, with the highest homology of 53% being to glucagon-like peptide-1 [7-36]-amide (GLP-1). Exendin-4 acts as a agonist on the GLP-1 receptor and bears GLP-1-like insulin sectretagogue action in isolated rat islets. Exendin-4 is a high potency agonist and truncated GLP-1 agonist-(9-39)-amide is an antagonist at the glucagon-like peptide 1-(7-36)-amide receptor of insulin-secreting beta-cells. (see e.g. J. Biol. Chem. 268(26):19650-19655). Exendin-4 (“exenatide”) was approved recently in the US and EU for improving glycemic control in patients with type 2 diabetes taking metformin and/or a sulfonylurea but have not achieved adequate glycemic control.


The amino acid sequence of exendin-4 is shown as SEQ ID NO: 1











HGEGTFTSDLSKQMEEEAVRLFIEWLKNGGPSSGAPPPS-NH2






The amino acid sequence of GLP-1(7-36)-amide is shown as SEQ ID NO 2











HAEGTFTSDVSSYLEGQAAKEFIAWLVKGR-NH2






Glucagon is a 29-amino acid peptide which is released into the bloodstream when circulating glucose is low. Glucagon's amino acid sequence is shown in SEQ ID NO 3.











HSQGTFTSDYSKYLDSRRAQDFVQWLMNT-OH






During hypoglycemia, when blood glucose levels drop below normal, glucagon signals the liver to break down glycogen and release glucose, causing an increase of blood glucose levels to reach a normal level. Hypoglycemia is a common side effect of insulin treated patients with hyperglycemia (elevated blood glucose levels) due to diabetes. Thus, glucagon's most predominant role in glucose regulation is to counteract insulin action and maintain blood glucose levels.


Hoist (Hoist, J. J. Physiol. Rev. 2007, 87, 1409) and Meier (Meier, J. J. Nat. Rev. Endocrinol. 2012, 8, 728) describe that GLP-1 receptor agonists, such as GLP-1, liraglutide and exendin-4, have 3 major pharmacological activities to improve glycemic control in patients with T2DM by reducing fasting and postprandial glucose (FPG and PPG): (i) increased glucose-dependent insulin secretion (improved first- and second-phase), (ii) glucagon suppressing activity under hyperglycemic conditions, (iii) delay of gastric emptying rate resulting in retarded absorption of meal-derived glucose.


GLP-1/Glucagon (Glc) Agonists


Pocai et al (Obesity. 2012; 20:1566-1571; Diabetes 2009, 58, 2258) and Day et al. (Nat Chem Biol 2009; 5:749) describe that dual activation of the GLP-1 and glucagon receptors, e.g., by combining the actions of GLP-1 and glucagon in one molecule leads to a therapeutic principle with anti-diabetic action and a pronounced weight lowering effect.


Peptides which bind and activate both the glucagon and the GLP-1 receptor (Hjort et al. Journal of Biological Chemistry, 269, 30121-30124, 1994; Day J W et al, Nature Chem Biol, 5: 749-757, 2009) and suppress body weight gain and reduce food intake are described in patent applications WO 2008/071972, WO 2008/101017, WO 2009/155258, WO 2010/096052, WO 2010/096142, WO 2011/075393, WO 2008/152403, WO 2010/070251, WO 2010/070252, WO 2010/070253, WO 2010/070255, WO 2011/160630, WO 2011/006497, WO 2011/152181, WO 2011/152182, WO2011/117415, WO2011/117416, and WO 2006/134340, the contents of which are herein incorporated by reference.


In addition, triple co-agonist peptides which not only activate the GLP-1 and the glucagon receptor, but also the GIP receptor (Gastric inhibitory polypeptide) are described in WO 2012/088116 and by VA Gault et al (Biochem Pharmacol, 85, 16655-16662, 2013; Diabetologia, 56, 1417-1424, 2013).


Bloom et al. (WO 2006/134340) disclose that peptides which bind and activate both the glucagon and the GLP-1 receptor can be constructed as hybrid molecules from glucagon and exendin-4, where the N-terminal part (e.g. residues 1-14 or 1-24) originate from glucagon and the C-terminal part (e.g. residues 15-39 or 25-39) originate from exendin-4.


Otzen et al (Biochemistry, 45, 14503-14512, 2006) disclose that N- and C-terminal hydrophobic patches are involved in fibrillation of glucagon, due to the hydrophobicity and/or high β-sheet propensity of the underlying residues.


WO2014/056872 discloses peptides which bind and activate both the glucagon and the GLP-1 receptor that are derived from exendin-4 wherein at least the aminoacid at position 14 bear a side chain for a prolonged halflife.


Peptides used in this invention are exendin-4 peptide analogues comprising leucine in position 10 and glutamine in position 13 and are described in WO2015/086731, WO2015/086732, WO2015/086733.


Peptides also used in this invention are exendin-4 peptide analogues comprising beta-alanine in position 28 and are described in EP15305899.5.


Further a peptide is used in this invention which is a exendin-4 peptide analogue comprising glutamine at position 3, beta-alanine in position 28 and D-alanines in position 29 and 34.


Peptides used in this invention preferably are soluble not only at neutral pH, but also at pH 4.5. Also the chemical stability at pH values of 4.5 to 5 is an important criterion for the long acting prodrug product. The prodrug is preferably formulated in this pH range in order to obtain a shelflife from at least 6 month at 4° C.


Longacting GLP-1/Glucagon Agonists


Ideally, the peptide is formulated in a fashion that provides for a sustained plasma level in human for at least one week after application to a human body resulting in a once-weekly or longer injection frequency.


Current therapy with a long acting GLP-1 agonists is Bydureon® which is exendin-4 in a depot suspension for a once weekly injection based on poly(glycol-co lactic acid) using a 23 gauge needle.


WO2012/173422 describes a GLP-1/Glucagon agonist conjugated to the Fc region of an immunoglobulin for weekly administration wherein the peptide is derived from oxyntomodulin.


Carrier Linked Prodrugs


To enhance physicochemical or pharmacokinetic properties of a drug in vivo, such as its half-live, such drug can be conjugated with a carrier. If the drug is transiently bound to a carrier and/or a linker, such systems are commonly assigned as carrier-linked prodrugs. According to the definitions provided by IUPAC (as given under http://www.chem.qmul.ac.uk/iupac.medchem, accessed on Jul. 22, 2009), a carrier-linked prodrug is a prodrug that contains a temporary linkage of a given active substance with a transient carrier group that produces improved physicochemical or pharmacokinetic properties and that can be easily removed in vivo, usually by a hydrolytic cleavage.


The linkers employed in such carrier-linked prodrugs may be transient, meaning that they are non-enzymatically hydrolytically degradable (cleavable) under physiological conditions (aqueous buffer at pH 7.4, 37° C.) with half-lives ranging from, for example, one hour to three months. Suitable carriers are polymers and can either be directly conjugated to the linker or via a non-cleavable spacer.


Transient polymer conjugation through traceless prodrug linkers combines the advantages of prolonged residence time due to polymer attachment and the recovery of the original pharmacology of the native peptide after release from the polymer conjugate.


Using polymer-linker peptide conjugates, native unchanged peptide is slowly released after application to a patient, governed only by release kinetics of the linker and pharmacokinetics of the polymer carrier. Ideally, release kinetics would be independent from the presence of enzymes like proteases or esterases in body fluids to guarantee a consistent and homogenous release pattern.


WO2008/148839, WO2009/095479 and WO2012/035139 refer to prodrugs comprising drug linker conjugates, where the linker is covalently attached via a cleavable bond to a biologically active moiety, such as the GLP 1-agonist exendin-4. The biologically active moiety is released from the prodrug upon cyclization-activation by cyclic imide formation. The release kinetic is dependent on the pH value and is minimum for storage of the prodrug at pH values from 4.5 to 5 and reach its intended release rate at physiological pH of around 7.4 to 7.5. An GLP-1 agonist-prodrug is described in which the linker is based on L-alanine and the polymeric carrier is a PEG-lysine based hydrogel. Not described are dual GLP-1/Glucagon agonist-prodrugs.


Hyaluronic Acid (HA)


Dhal et al (Journal of Biomedical Nanotechnology, vol 9, 2013, 1-13) report hyaluronic acid as a suitable carrier for drug conjugates. Kong et al. (Biomaterials 31 (2010), 4121-4128) report an exendin-4-hyaluronic acid conjugate which showed an glucose lowering effect over 3 days in mice. The used HA was a linear polymer with a drug load ranging from about 2.4 to 12.%.


In the present invention hydrogels of crosslinked hyaluronic acid are chosen due to their longer residence time as a local depot at the application site than soluble HA. Important criteria for the use of hyaluronic acid (HA) as a carrier polymer is the achievable drug load in the final drug product which is determined by the drug load on the polymer itself and the concentration of the final solution/suspension. Giving the fact that the injection volume for subcutaneous drug depots is practically limited to equal/less than 1 mL, preferably equal/less than 0.6 mL.


The more concentrated the polymer solutions/suspensions of HA is, the more viscous is the formulation which has a negative impact on the syringability of the prodrug formulation. Viscous solutions need injection needles of a larger diameter to limit the force on the plunger of which the syringe is pressed. Also the time for injection is longer.


It is an object of the present invention to provide a GLP-1/Glucagon agonist prodrug for administering as a subcutaneous depot which releases a GLP-1/Glucagon agonist in an active form over the time period of at least 6 days between administrations and which can be injected through 26 gauge needles or even needles of smaller inner diameter for good patient compliance.


An object of the invention is a prodrug or a pharmaceutically acceptable salt thereof comprising a drug linker conjugate of formula (I)

Z-L1-L2-L-Y—R20  (I)

wherein Y is a peptide moiety having the formula (II)









(II)


His-X2-X3-Gly-Thr-Phe-Thr-Ser-Asp-Leu-Ser-Lys-





Gln-Leu-Asp-Glu-Gln-X18-Ala-X20-X21-Phe-Ile-Glu-





Trp-Leu-Ile-X28-Gly-Gly-Pro-X32-Ser-Gly-Ala-Pro-





Pro-Pro-Ser








    • X2 represents an amino acid residue selected from Ser, D-Ser and Aib,

    • X3 represents an amino acid residue selected from Gln and His,

    • X18 represents an amino acid residue selected from Arg and Lys

    • X20 represents an amino acid residue selected from Lys, Gln and His,

    • X21 represents an amino acid residue selected from Asp and Glu,

    • X28 represents an amino acid residue selected from Ser and Ala,

    • X32 represents an amino acid residue selected from Ser and Val,


      or wherein Y is a peptide moiety having the formula (III)












(III)


His-X2-X3-Gly-Thr-Phe-Thr-Ser-Asp-Leu-Ser-Lys-





Gln-Leu-Asp-Glu-Gln-X18-Ala-X20-X21-Phe-Ile-Glu-





Trp-Leu-Ile-X28-Gly-Gly-Pro-X32-Ser-Gly-Ala-Pro-





Pro-Pro-Ser








    • X2 represents an amino acid residue selected from Ser, D-Ser and Aib,

    • X3 represents an amino acid residue selected from Gln and His,

    • X18 represents an amino acid residue selected from Leu and His

    • X20 represents an amino acid residue selected from His, Arg, Lys, and Gln,

    • X21 represents an amino acid residue selected from Asp and Glu,

    • X28 represents an amino acid residue selected from Lys, Ser and Ala,

    • X32 represents an amino acid residue selected from Ser and Val,


      or


      wherein Y is a peptide moiety having the formula (IV)












(IV)


His-X2-X3-Gly-Thr-Phe-Thr-Ser-Asp-Leu-Ser-Lys-





Leu-Leu-Asp-Glu-Gln-X18-Ala-Lys-Asp-Phe-Ile-Glu-





Trp-Leu-Ile-Ala-Gly-Gly-Pro-X32-Ser-Gly-Ala-Pro-





Pro-Pro-Ser








    • X2 represents an amino acid residue selected from Ser, D-Ser and Aib,

    • X3 represents an amino acid residue selected from Gln and His,

    • X18 represents an amino acid residue selected from Arg and Leu,

    • X32 represents an amino acid residue selected from Ser and Val,


      or


      wherein Y is a peptide moiety having the formula (IVa)












(IVa)


H2N-His-Aib-His-Gly-Thr-Phe-Thr-Ser-Asp-Leu-Ser-





Lys-Gln-Leu-X15-Glu-Gln-Leu-Ala-Arg-Asp-Phe-Ile-





Glu-Trp-Leu-Ile-Bal-X29-Gly-X31-X32-Ser-X34-X35-





Pro-Pro-Pro-X39-R20








    • X15 represents an amino acid residue selected from Asp and Glu, (pref. Asp)

    • X29 represents an amino acid residue selected from Gly, D-Ala and Pro, (pref) Gly, D-Ala

    • X31 represents an amino acid residue selected from Pro, His and Trp, (pref. Pro)

    • X32 represents an amino acid residue selected from Ser, His, Pro and Arg, (pref. Ser, His, Pro),

    • X34 represents an amino acid residue selected from Gly and D-Ala,

    • X35 represents an amino acid residue selected from Ala, Pro and Lys, (pref. Ala, Pro)

    • X39 represents Ser or Pro-Pro-Pro,


      or


      wherein Y is a peptide moiety having the formula (IVb)












H2N-His-Aib-Gln-Gly-Thr-Phe-Thr-Ser-Asp-Leu-Ser-





Lys-Leu-Leu-Glu-Glu-Gln-Arg-Ala-Arg-Glu-Phe-Ile-





Glu-Trp-Leu-Ile-Bal-D-Ala-Gly-Pro-Pro-Ser-D-Ala-





Ala-Pro-Pro-Pro-Ser-R20;







or a salt or solvate thereof;


R20 is OH or NH2;


L is a linker of formula (Ia),




embedded image




    • wherein the dashed line indicates the attachment to the N-Terminus of Y by forming an amide bond;

    • X is C(R4R4a); N(R4);

    • R1, R1a, are independently selected from the group consisting of H; and C1-4 alkyl;

    • R2, R2a, are independently selected from the group consisting of H; and C1-4 alkyl;

    • R4, R4a, are independently selected from the group consisting of H; and C1-4 alkyl;

    • wherein R2, R2a, R4 or R4a is substituted with one group L2-L1-Z; wherein R2a,


      L2 is a single chemical bond or is a C1-20 alkyl chain, which is optionally interrupted by one or more groups independently selected from —O— and C(O)N(R3aa) and is optionally substituted with one or more groups independently selected from OH and C(O)N(R3aaR3aaa), wherein R3aa and R3aaa are independently selected from the group consisting of H and C1-4 alkyl; and

    • L2 is attached to L1 via a terminal group selected from the group consisting of







embedded image






      • wherein L2 is attached to the one position indicated with the dashed line and L1 is attached to the position indicated with the other dashed line; and


        L1 is a C1-20 alkyl chain, which is optionally interrupted by one or more groups independently selected from —O— and C(O)N(R5aa) and is optionally substituted with one or more groups independently selected from OH and C(O)N(R5aaR5aaa), wherein R5aa and R5aaa are independently selected from the group consisting of H and C1-4 alkyl; and


        L1 is attached to Z via a terminal amino group forming an amide bond with the carboxy group of the beta-1,3-D-glucuronic acid of the hyaluronic acid of Z;


        Z is a crosslinked hyaluronic acid hydrogel, in which


        0.05 to 20% of the monomeric disaccharide units are crosslinked by a crosslinker; and


        0.2 to 8.5% of the monomeric disaccharide units bear L′-L2-L-Y—R20.groups.







The present invention relates to a prodrug which provides a GLP-1/Glucagon agonist release from a subcutaneous depot in an active form over the time period of at least 6 days between administrations


This helps patients to reduce the frequency of injections, while being able to maintain optimal control the plasma levels of GLP-1/Glucagon agonist and consequently blood glucose.


Further advantages of the crosslinked hyaluronic acid carrier of the invention are the good injectability through a 26 gauge needle or even a needle of a smaller inner diameter.





LEGENDS TO THE FIGURES


FIG. 1a, FIG. 1b, FIG. 1c show various crosslinking chemistries to synthesize hyaluronic acid hydrogels.



FIG. 2a. Magnetic Resonance (MR) images of the HA hydrogel at the injection site, taken at different time points.



FIG. 2b. Magnetic Resonance (MR) images of the polymer suspension containing 1:1 (w/w) HA hydrogel-800 kDa soluble HA at the injection site, taken at different time points.



FIG. 3. In vitro release kinetics of Exendin-4 Seq ID No. 26 dual agonist with weekly linker from the HA hydrogel (example 18). The half-life is 5 days.



FIG. 4 In vivo Effect of HA Hydrogel-Dual Agonist (Seq ID No. 26) Conjugate on Non-fasting Blood Glucose in db/db Mice: shown is the difference level of blood glucose in mmol/L versus the treatment time of the animals for different doses.



FIG. 4b In vivo Effect of HA Hydrogel-Dual Agonist (Seq ID No. 26) Conjugate on Non-fasting Blood Glucose in db/db Mice: shown is the difference level of blood glucose in mmol/L versus the treatment time of the animals for different hydrogel constructs: soluble and crosslinked hyaluronic acid.



FIG. 4c In vivo Effect of HA Hydrogel-Dual agonists (Seq ID No. 45, 46 and 48) Conjugates on Non-fasting Blood Glucose in db/db Mice: shown is the level of blood glucose in mmol/L versus the treatment time of the animals.



FIG. 4d In vivo Effect of HA Hydrogel-Dual Agonist (Seq ID No. 49) Conjugate on Non-fasting Blood Glucose in db/db Mice: shown is the level of blood glucose in mmol/L versus the treatment time of the animals.



FIG. 5 Injectability study: Extrusion Force on syringe plunger for HA hydrogels of different peptide loading.





DETAILED DESCRIPTION

The amino acid sequences of the present invention contain the conventional one letter and three letter codes for naturally occurring amino acids, as well as generally accepted three letter codes for other amino acids, such as Aib (α-aminoisobutyric acid). Furthermore, the following code was used for the amino acid shown in the table below.

















Name
Structure
code









beta alanine


embedded image


Bal







D-alanine

D-Ala










GLP-1/Glucagon agonist bound to linker is referred to as “GLP-1/Glucagon agonist moiety”.


“Protective groups” refers to a moiety which temporarily protects a chemical functional group of a molecule during synthesis to obtain chemoselectivity in subsequent chemical reactions. Protective groups for alcohols are, for example, benzyl and trityl, protective groups for amines are, for example, tert-butyloxycarbonyl, 9-fluorenylmethyloxycarbonyl and benzyl and for thiols examples of protective groups are 2,4,6-trimethoxybenzyl, phenylthiomethyl, acetamidomethyl, p-methoxybenzyloxycarbonyl, tert-butylthio, triphenylmethyl, 3-nitro-2-pyridylthio, 4-methyltrityl.


“Protected functional groups” means a chemical functional group protected by a protective group.


“Acylating agent” means a moiety of the structure R—(C═O)—, providing the acyl group in an acylation reaction, optionally connected to a leaving group, such as acid chloride, N-hydroxy succinimide, pentafluorphenol and para-nitrophenol.


“Alkyl” means a straight-chain or branched carbon chain. Each hydrogen of an alkyl carbon may be replaced by a substituent.


“Aryl” refers to any substituent derived from a monocyclic or polycyclic or fused aromatic ring, including heterocyclic rings, e.g. phenyl, thiophene, indolyl, napthyl, pyridyl, which may optionally be further substituted.


“Acyl” means a chemical functional group of the structure R—(C═O)—, wherein R is an alkyl or aryl.


“C1-4 alkyl” means an alkyl chain having 1-4 carbon atoms, e.g. if present at the end of a molecule: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl tert-butyl, or e.g. —CH2—, —CH2—CH2—, —CH(CH3)—, —CH2—CH2—CH2—, —CH(C2H5)—, —C(CH3)2—, when two moieties of a molecule are linked by the alkyl group. Each hydrogen of a C1-4 alkyl carbon may be replaced by a substituent.


“C1-6 alkyl” means an alkyl chain having 1-6 carbon atoms, e.g. if present at the end of a molecule: C1-4 alkyl, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl; tert-butyl, n-pentyl, n-hexyl, or e.g. —CH2—, —CH2—CH2—, —CH(CH3)—, —CH2—CH2—CH2—, —CH(C2H5)—, —C(CH3)2—, when two moieties of a molecule are linked by the alkyl group. Each hydrogen of a C1-6 alkyl carbon may be replaced by a substituent.


Accordingly, “C1-18 alkyl” means an alkyl chain having 1 to 18 carbon atoms and “C8-18 alkyl” means an alkyl chain having 8 to 18 carbon atoms. Accordingly, “C1-50 alkyl” means an alkyl chain having 1 to 50 carbon atoms.


“Halogen” means fluoro, chloro, bromo or iodo. It is generally preferred that halogen is fluoro or chloro.


“Hyaluronic acid” means a polymer of a disaccharide composed of beta-1,3-D-glucuronic acid and beta-1,4-N-acetyl-D-glucosamine and their respective sodium salts. These polymers are linear.




embedded image


“Disaccharide unit” means the disaccharide composed of beta-1,3-D-glucuronic acid and beta-1,4-N-acetyl-D-glucosamine and their respective sodium salts and is the monomeric building block for HA.


“Crosslinked hyaluronic acid” means a polymer of hyaluronic acid” wherein different chains of HA are covalently connected by a crosslinker, forming a 3-dimensional polymer network. The degree of crosslinking refers the molar ratio of disaccharide units to crosslinker units in the polymer network.


Crosslinked hyaluronic acid” may be derived by different methods. Reaction of HA with the crosslinker, reaction of modified (activated) HA with the crosslinker, reaction of two different modified HA with the crosslinker. Examples may be described in Oh et al, Journal of Controlled Release 141 (2010), 2-12. Example 11 describes the crosslinking of unmodified HA with divinylsulfone. Further methods for preparing crosslinked HA are also depicted in FIG. 1a, FIG. 1b and FIG. 1c: aldehyde (diol oxidation) and subsequent amine reductive amination, hydroxyl mediated alkylation, amide formation reaction, Michael Addition Crosslinking (Thiol-maleimide), diol-epoxide chemistry and others.


“Crosslinker” may be a linear or branched molecule or chemical group, preferably is a linear molecule with at least chemical functional groups on each distal ends


“functionalized hyaluronic acid” means a polymer of hyaluronic acid” wherein HA is chemically modified with a group L1 which bears a chemical functional chemical group at its distal end. The degree of functionalization refers the molar ratio of disaccharide units to L1 units in the polymer.


The term “chemical functional group” refers to but not limited to carboxylic acid and activated derivatives, amino, maleimide, thiol and derivatives, sulfonic acid and derivatives, carbonate and derivatives, carbamate and derivatives, hydroxyl, aldehyde, ketone, hydrazine, isocyanate, isothiocyanate, phosphoric acid and derivatives, phosphonic acid and derivatives, haloacetyl, alkyl halides, acryloyl and other alpha-beta unsaturated michael acceptors, arylating agents like aryl fluorides, hydroxylamine, disulfides like pyridyl disulfide, vinyl sulfone, vinyl ketone, diazoalkanes, diazoacetyl compounds, oxirane, and aziridine.


If a chemical functional group is coupled to another chemical functional group, the resulting chemical structure is referred to as “linkage”. For example, the reaction of an amine group with a carboxyl group results in an amide linkage.


“Reactive functional groups” are chemical functional groups of the backbone moiety, which are connected to the hyperbranched moiety.


“Functional group” is the collective term used for “reactive functional group”, “degradable interconnected functional group”, or “conjugate functional group”.


The terms “blocking group” or “capping group” are used synonymously and refer to moieties which are irreversibly connected to reactive functional groups to render them incapable of reacting with for example chemical functional groups.


The terms “protecting group” or “protective group” refers to a moiety which is reversibly connected to reactive functional groups to render them incapable of reacting with for example other chemical functional groups under specific conditions.


The term “derivatives” refers to chemical functional groups suitably substituted with protecting and/or activation groups or to activated forms of a corresponding chemical functional group which are known to the person skilled in the art. For example, activated forms of carboxyl groups include but are not limited to active esters, such as succinimidyl ester, benzotriazyl ester, nitrophenyl ester, pentafluorophenyl ester, azabenzotriazyl ester, acyl halogenides, mixed or symmetrical anhydrides, acyl imidazole.


The term “non-enzymatically cleavable linker” refers to linkers that are hydrolytically degradable under physiological conditions without enzymatic activity.


The terms “spacer”, “spacer group”, “spacer molecule”, and “spacer moiety” are used interchangeably and if used to describe a moiety present in the hydrogel carrier of the invention, refer to any moiety suitable for connecting two moieties, such as C1-50 alkyl, which fragment is optionally interrupted by one or more groups selected from —NH—, —N(C1-4 alkyl)-, —O—, —S—, —C(O)—, —C(O)NH—, —C(O)N(C1-4 alkyl)-, —O—C(O)—, —S(O)—, —S(O)2—.


The terms “terminal”, “terminus” or “distal end” refer to the position of a functional group or linkage within a molecule or moiety, whereby such functional group may be a chemical functional group and the linkage may be a degradable or permanent linkage, characterized by being located adjacent to or within a linkage between two moieties or at the end of an oligomeric or polymeric chain.


The phrases “in bound form” or “moiety” refer to sub-structures which are part of a larger molecule. The phrase “in bound form” is used to simplify reference to moieties by naming or listing reagents, starting materials or hypothetical starting materials well known in the art, and whereby “in bound form” means that for example one or more hydrogen radicals (—H), or one or more activating or protecting groups present in the reagents or starting materials are not present in the moiety.


It is understood that all reagents and moieties comprising polymeric moieties refer to macromolecular entities known to exhibit variabilities with respect to molecular weight, chain lengths or degree of polymerization, or the number of functional groups. Structures shown for crosslinking reagents, and crosslinked moieties are thus only representative examples.


A reagent or moiety may be linear or branched. If the reagent or moiety has two terminal groups, it is referred to as a linear reagent or moiety. If the reagent or moiety has more than two terminal groups, it is considered to be a branched or multi-functional reagent or moiety.


The linkers employed in such carrier-linked prodrugs are transient, meaning that they are non-enzymatically hydrolytically degradable (cleavable) under physiological conditions (aqueous buffer at pH 7.4, 37° C.) with half-lives ranging from, for example, one hour to three months.


The term “GLP-1/Glucagon agonist hydrogel prodrug” refers to carrier-linked prodrugs of GLP-1/Glucagon agonist, wherein the carrier is a hydrogel. The terms “hydrogel prodrug” and “hydrogel-linked prodrug” refer to prodrugs of biologically active agents transiently linked to a hydrogel and are used synonymously.


A “hydrogel” may be defined as a three-dimensional, hydrophilic or amphiphilic polymeric network capable of taking up large quantities of water. The networks are composed of homopolymers or copolymers, are insoluble due to the presence of covalent chemical or physical (ionic, hydrophobic interactions, entanglements) crosslinks. The crosslinks provide the network structure and physical integrity. Hydrogels exhibit a thermodynamic compatibility with water which allows them to swell in aqueous media. The chains of the network are connected in such a fashion that pores exist and that a substantial fraction of these pores are of dimensions between 1 nm and 1000 nm.


“Free form” of a drug refers to a drug, specifically to GLP-1/Glucagon agonist, in its unmodified, pharmacologically active form, such as after being released from a polymer conjugate.


The terms “drug”, “biologically active molecule”, “biologically active moiety”, “biologically active agent”, “active agent”, are used synonymously and refer to GLP-1/Glucagon agonist, either in its bound or free form.


A “therapeutically effective amount” of GLP-1/Glucagon agonist as used herein means an amount sufficient to cure, alleviate or partially arrest the clinical manifestations of a given disease and its complications. An amount adequate to accomplish this is defined as “therapeutically effective amount”. Effective amounts for each purpose will depend on the severity of the disease or injury as well as the weight and general state of the subject. It will be understood that determining an appropriate dosage may be achieved using routine experimentation, by constructing a matrix of values and testing different points in the matrix, which are all within the ordinary skills of a trained physician.


“Stable” and “stability” means that within the indicated storage time the hydrogel conjugates remain conjugated and do not hydrolyze to a substantial extent and exhibit an acceptable impurity profile relating to GLP-1/Glucagon agonist. To be considered stable, the composition contains less than 5% of the drug in its free form.


The term “pharmaceutically acceptable” means approved by a regulatory agency such as the EMEA (Europe) and/or the FDA (US) and/or any other national regulatory agencies for use in animals, preferably in humans.


“Pharmaceutical composition” or “composition” means one or more active ingredients, and one or more inert ingredients, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients. Accordingly, the pharmaceutical compositions of the present invention encompass any composition made by admixing a compound of the present invention and a pharmaceutically acceptable excipient (pharmaceutically acceptable carrier).


“Dry composition” means that the GLP-1/Glucagon agonist hydrogel prodrug composition is provided in a dry form in a container. Suitable methods for drying are for example spray-drying and lyophilization (freeze-drying). Such dry composition of GLP-1/Glucagon agonist hydrogel prodrug has a residual water content of a maximum of 10%, preferably less than 5% and more preferably less than 2% (determined according to Karl Fischer method). The preferred method of drying is lyophilization. “Lyophilized composition” means that the GLP-1/Glucagon agonist hydrogel polymer prodrug composition was first frozen and subsequently subjected to water reduction by means of reduced pressure. This terminology does not exclude additional drying steps which occur in the manufacturing process prior to filling the composition into the final container.


“Lyophilization” (freeze-drying) is a dehydration process, characterized by freezing a composition and then reducing the surrounding pressure and, optionally, adding heat to allow the frozen water in the composition to sublime directly from the solid phase to gas. Typically, the sublimed water is collected by desublimation.


“Reconstitution” means the addition of a liquid to a dry composition to bring it into the form of a liquid or suspension composition. It is understood that the term “reconstitution” is not limited to the addition of water, but refers to the addition of any liquid, including for example buffers or other aqueous solutions.


“Reconstitution solution” refers to the liquid used to reconstitute the dry composition of an GLP-1/Glucagon agonist hydrogel prodrug prior to administration to a patient in need thereof.


“Container” means any container in which the GLP-1/Glucagon agonist hydrogel prodrug composition is comprised and can be stored until reconstitution.


“Buffer” or “buffering agent” refers to chemical compounds that maintain the pH in a desired range. Physiologically tolerated buffers are, for example, sodium phosphate, succinate, histidine, bicarbonate, citrate and acetate, pyruvate. Antacids such as Mg(OH)2 or ZnCO3 may be also used. Buffering capacity may be adjusted to match the conditions most sensitive to pH stability.


“Excipients” refers to compounds administered together with the therapeutic agent, for example, buffering agents, isotonicity modifiers, preservatives, stabilizers, anti-adsorption agents, oxidation protection agents, or other auxiliary agents. However, in some cases, one excipient may have dual or triple functions.


A “lyoprotectant” is a molecule which, when combined with a protein of interest, significantly prevents or reduces chemical and/or physical instability of the protein upon drying in general and especially during lyophilization and subsequent storage. Exemplary lyoprotectants include sugars, such as sucrose or trehalose; amino acids such as arginine, glycine, glutamate or histidine; methylamines such as betaine; lyotropic salts such as magnesium sulfate; polyols such as trihydric or higher sugar alcohols, e.g. glycerin, erythritol, glycerol, arabitol, xylitol, sorbitol, and mannitol; ethylene glycol; propylene glycol; polyethylene glycol; pluronics; hydroxyalkyl starches, e.g. hydroxyethyl starch (HES), and combinations thereof.


“Surfactant” refers to wetting agents that lower the surface tension of a liquid.


“Isotonicity modifiers” refer to compounds which minimize pain that can result from cell damage due to osmotic pressure differences at the injection depot.


The term “stabilizers” refers to compounds used to stabilize the polymer prodrug. Stabilisation is achieved by strengthening of the protein-stabilising forces, by destabilisation of the denatured state, or by direct binding of excipients to the protein.


“Anti-adsorption agents” refers to mainly ionic or non-ionic surfactants or other proteins or soluble polymers used to coat or adsorb competitively to the inner surface of the composition's container. Chosen concentration and type of excipient depends on the effect to be avoided but typically a monolayer of surfactant is formed at the interface just above the CMC value.


“Oxidation protection agents” refers to antioxidants such as ascorbic acid, ectoine, glutathione, methionine, monothioglycerol, morin, polyethylenimine (PEI), propyl gallate, vitamin E, chelating agents such aus citric acid, EDTA, hexaphosphate, thioglycolic acid.


“Antimicrobial” refers to a chemical substance that kills or inhibits the growth of microorganisms, such as bacteria, fungi, yeasts, protozoans and/or destroys viruses.


“Sealing a container” means that the container is closed in such way that it is airtight, allowing no gas exchange between the outside and the inside and keeping the content sterile.


The term “reagent” or “precursor” refers to an intermediate or starting material used in the assembly process leading to a prodrug of the present invention.


An object of the invention is a prodrug or a pharmaceutically acceptable salt thereof comprising a drug linker conjugate of formula (I)

Z-L1-L2-L-Y—R20  (I)

wherein Y is a peptide moiety having the formula (II)









(II)


His-X2-X3-Gly-Thr-Phe-Thr-Ser-Asp-Leu-Ser-Lys-





Gln-Leu-Asp-Glu-Gln-X18-Ala-X20-X21-Phe-Ile-Glu-





Trp-Leu-Ile-X28-Gly-Gly-Pro-X32-Ser-Gly-Ala-Pro-





Pro-Pro-Ser








    • X2 represents an amino acid residue selected from Ser, D-Ser and Aib,

    • X3 represents an amino acid residue selected from Gln and His,

    • X18 represents an amino acid residue selected from Arg and Lys

    • X20 represents an amino acid residue selected from Lys, Gln and His,

    • X21 represents an amino acid residue selected from Asp and Glu,

    • X28 represents an amino acid residue selected from Ser and Ala,

    • X32 represents an amino acid residue selected from Ser and Val,


      or wherein Y is a peptide moiety having the formula (III)












(III)


His-X2-X3-Gly-Thr-Phe-Thr-Ser-Asp-Leu-Ser-Lys-





Gln-Leu-Asp-Glu-Gln-X18-Ala-X20-X21-Phe-Ile-Glu-





Trp-Leu-Ile-X28-Gly-Gly-Pro-X32-Ser-Gly-Ala-Pro-





Pro-Pro-Ser








    • X2 represents an amino acid residue selected from Ser, D-Ser and Aib,

    • X3 represents an amino acid residue selected from Gln and His,

    • X18 represents an amino acid residue selected from Leu and His

    • X20 represents an amino acid residue selected from His, Arg, Lys, and Gln,

    • X21 represents an amino acid residue selected from Asp and Glu,

    • X28 represents an amino acid residue selected from Lys, Ser and Ala,

    • X32 represents an amino acid residue selected from Ser and Val,


      or


      wherein Y is a peptide moiety having the formula (IV)












(IV)


His-X2-X3-Gly-Thr-Phe-Thr-Ser-Asp-Leu-Ser-Lys-Leu-





Leu-Asp-Glu-Gln-X18-Ala-Lys-Asp-Phe-IIe-Glu-Trp-





Leu-IIe-Ala-Gly-Gly-Pro-X32-Ser-Gly-Ala-Pro-Pro-





Pro-Ser 








    • X2 represents an amino acid residue selected from Ser, D-Ser and Aib,

    • X3 represents an amino acid residue selected from Gln and His,

    • X18 represents an amino acid residue selected from Arg and Leu,

    • X32 represents an amino acid residue selected from Ser and Val,

    • or a salt or solvate thereof;

    • or a salt or solvate thereof;


      R20 is OH or NH2;


      L is a linker of formula (Ia),







embedded image




    • wherein the dashed line indicates the attachment to the N-Terminus of Y by forming an amide bond;

    • X is C(R4R4a); N(R4);

    • R1, R1a, are independently selected from the group consisting of H; and C1-4 alkyl;

    • R2, R2a, are independently selected from the group consisting of H; and C1-4 alkyl;

    • R4, R4a, are independently selected from the group consisting of H; and C1-4 alkyl;

    • wherein R2, R2a, R4 or R4a is substituted with one group L2-L1-Z; wherein


      L2 is a single chemical bond or is a C1-20 alkyl chain, which is optionally interrupted by one or more groups independently selected from —O— and C(O)N(R3aa) and is optionally substituted with one or more groups independently selected from OH and C(O)N(R3aaR3aaa), wherein R3aa and R3aaa are independently selected from the group consisting of H and C1-4 alkyl; and

    • L2 is attached to L1 via a terminal group selected from the group consisting of







embedded image






      • wherein L2 is attached to the one position indicated with the dashed line and L1 is attached to the position indicated with the other dashed line; and


        L1 is a C1-20 alkyl chain, which is optionally interrupted by one or more groups independently selected from —O— and C(O)N(R5aa) and is optionally substituted with one or more groups independently selected from OH and C(O)N(R5aaR5aaa), wherein R5aa and R5aaa are independently selected from the group consisting of H and C1-4 alkyl; and


        L1 is attached to Z via a terminal amino group forming an amide bond with the carboxy group of the beta-1,3-D-glucuronic acid of the hyaluronic acid of Z;


        Z is a crosslinked hyaluronic acid hydrogel, in which


        0.05 to 20% of the monomeric disaccharide units are crosslinked by a crosslinker; and


        0.2 to 8.5% of the monomeric disaccharide units bear L1-L2-L-Y—R20.groups.







Further embodiments of L, L1, L2, Z and Y.


In another embodiment


L is a linker moiety of formula (Ib),




embedded image



wherein the dashed line indicates attachment to Y by forming an amide bond;


R1, R1a, R2a are selected independently from the group consisting of H and C1-4 alkyl;


L2-L1-Z is defined as described above.


In another embodiment


L is a linker moiety of formula (Ib), wherein


R1 is CH3;


R1a is H;


R2a is H; and


L2-L1-Z is defined as described above.


In another embodiment


L is a linker moiety of formula (Ib), wherein


R1 is H;


R1a is CH3;


R2a is H; and


L2-L1-Z is defined as described above.


In another embodiment


L is a linker moiety of formula (Ib), wherein


R1 is CH3;


R1a is CH3;


R2a is H; and


L2-L1-Z is defined as described above.


In another embodiment


L is a linker moiety -L of formula (Ic),




embedded image



wherein the dashed line indicates attachment to Y by forming an amide bond;


R1 is selected from H or C1-4 alkyl, preferably H;


R1a is selected from H or C1-4 alkyl, preferably H;


R2, R2a are independently selected from the group consisting of H and C1-4 alkyl;


wherein L2-L1-Z is defined as described above.


In another embodiment


L is a linker moiety -L of formula (Ic),




embedded image



wherein the dashed line indicates attachment to Y by forming an amide bond;


R1 and R1a are H;


R2, R2a are independently selected from the group consisting of H and CH3;


wherein L2-L1-Z is defined as described above.


In another embodiment


L is a linker moiety -L of formula (Ic), wherein


R1 and R1a are H;


R2 is H and R2a is CH3;


wherein L2-L1-Z is defined as described above.


In another embodiment


L2 is a C1-10 alkyl chain, which is optionally interrupted by one or two groups independently selected from —O— and C(O)N(R3aa) and, wherein R3aa is independently selected from the group consisting of H and C1-4 alkyl; and


L2 is attached to L1 via a terminal group selected from the group consisting of




embedded image




    • wherein L2 is attached to the one position indicated with the dashed line and

    • L1 is attached to the position indicated with the other dashed line; and





In another embodiment


L2 is a C1-6 alkyl chain, which is optionally interrupted by one group selected from —O— and C(O)N(R3aa) and, wherein R3aa is independently selected from the group consisting of H and C1-4 alkyl; and


L2 is attached to L1 via a terminal group selected from the group consisting of




embedded image




    • wherein L2 is attached to the one position indicated with the dashed line and L1 is attached to the position indicated with the other dashed line.





In another embodiment


L2 is —CH2—CH2—CH2—CH2—CH2—C(O)NH— or —CH2—CH2—CH2—CH2—CH2—CH2— and is attached to L1 via the terminal group




embedded image



wherein L2 is attached to the Sulfur atom indicated with the dashed line and L1 is attached to nitrogen atom indicated with the dashed line.


In another embodiment


L1 is a C1-10 alkyl chain, with an amino group on one distal end, which is optionally interrupted by one or two groups independently selected from —O— and C(O)N(R5aa) and, wherein R5aa is independently selected from the group consisting of H and C1-4 alkyl.


A further embodiment relates to prodrugs, wherein


Z is a crosslinked hyaluronic acid hydrogel, in which


0.05 to 15% of the monomeric disaccharide units are crosslinked by a crosslinker.


A further embodiment relates to prodrugs, wherein


Z is a crosslinked hyaluronic acid hydrogel, in which


1 to 10% of the monomeric disaccharide units are crosslinked by a crosslinker.


A further embodiment relates to prodrugs, wherein


Z is a crosslinked hyaluronic acid hydrogel, in which


0.2 to 8.5% of the monomeric disaccharide units bear L1-L2-L-Y—R20.groups.


A further embodiment relates to prodrugs, wherein


Z is a crosslinked hyaluronic acid hydrogel, in which


0.2 to 6% of the monomeric disaccharide units bear L1-L2-L-Y—R20.groups.


A further embodiment relates to prodrugs, wherein


Z is a crosslinked hyaluronic acid hydrogel, in which


0.2 to 5% of the monomeric disaccharide units bear L1-L2-L-Y—R20.groups.


A further embodiment relates to prodrugs, wherein


Z is a crosslinked hyaluronic acid hydrogel, in which


0.4 to 4% of the monomeric disaccharide units bear L1-L2-L-Y—R20.groups.


In another embodiment the GLP-1/Glucagon agonist prodrug has a structure as represented by formula (V)




embedded image


In another embodiment the GLP-1/Glucagon agonist prodrug has a structure as represented by formula (VI) (Aib-linker)




embedded image


In another embodiment the GLP-1/Glucagon agonist prodrug has a structure as represented by formula (VII)




embedded image


A further embodiment relates to a group of prodrugs having a peptide moiety Y of formula (II), wherein

    • X2 represents an amino acid residue selected from Ser, D-Ser and Aib,
    • X3 represents an amino acid residue selected from Gln and His,
    • X18 represents Arg
    • X20 represents an amino acid residue selected from Lys, Gln and His,
    • X21 represents an amino acid residue selected from Asp and Glu,
    • X28 represents an amino acid residue selected from Ser and Ala,
    • X32 represents an amino acid residue selected from Ser and Val.


A further embodiment relates to a group of prodrugs having a peptide moiety Y of formula (II), wherein

    • X2 represents an D-Ser
    • X3 represents His,
    • X18 represents Arg
    • X20 represents Lys,
    • X21 represents an amino acid residue selected from Asp and Glu,
    • X28 represents an amino acid residue selected from Ser and Ala,
    • X32 represents an amino acid residue selected from Ser and Val.


A further embodiment relates to a group of prodrugs having a peptide moiety Y of formula (II), wherein

    • X2 represents D-Ser,
    • X3 represents an amino acid residue selected from Gln and His,
    • X18 represents Arg,
    • X20 represents Lys,
    • X21 represents an amino acid residue selected from Asp and Glu,
    • X28 represents Ala,
    • X32 represents an amino acid residue selected from Ser and Val.


A further embodiment relates to a group of prodrugs having a peptide moiety Y of formula (II), wherein

    • X2 represents an amino acid residue selected from Ser, D-Ser and Aib,
    • X3 represents His,
    • X18 represents Arg,
    • X20 represents Lys,
    • X21 represents Asp,
    • X28 represents Ala,
    • X32 represents an amino acid residue selected from Ser and Val.


A further embodiment relates to a group of prodrugs having a peptide moiety Y of formula (II), wherein

    • X2 represents an amino acid residue selected from Ser, D-Ser and Aib,
    • X3 represents an amino acid residue selected from Gln and His,
    • X18 represents an amino acid residue selected from Arg and Lys,
    • X20 represents Lys,
    • X21 represents an amino acid residue selected from Asp and Glu,
    • X28 represents an amino acid residue selected from Ser and Ala,
    • X32 represents an amino acid residue selected from Ser and Val.


A further embodiment relates to a group of prodrugs having a peptide moiety Y of formula (II), wherein

    • X2 represents an amino acid residue selected from Ser, D-Ser and Aib,
    • X3 represents an amino acid residue selected from Gln and His,
    • X18 represents an amino acid selected from Arg and Lys,
    • X20 represents an amino acid residue selected from Lys, Gln and His,
    • X21 represents Asp,
    • X28 represents an amino acid residue selected from Ser and Ala,
    • X32 represents an amino acid residue selected from Ser and Val.


A further embodiment relates to a group of prodrugs having a peptide moiety Y of formula (II), wherein

    • X2 represents an amino acid residue selected from Ser, D-Ser and Aib,
    • X3 represents an amino acid residue selected from Gln and His,
    • X18 represents an amino acid selected from Arg and Lys,
    • X20 represents an amino acid residue selected from Lys, Gln and His,
    • X21 represents an amino acid residue selected from Asp and Glu,
    • X28 represents an amino acid residue selected from Ser and Ala,
    • X32 represents Ser.


A further embodiment relates to a group of prodrugs having a peptide moiety Y of formula (II), wherein

    • X2 represents an amino acid residue selected from Ser, D-Ser and Aib,
    • X3 represents an amino acid residue selected from Gln and His,
    • X18 represents Arg
    • X20 represents Lys,
    • X21 represents an amino acid residue selected from Asp and Glu,
    • X28 represents Ala,
    • X32 represents Val.


A further embodiment relates to a group of prodrugs having a peptide moiety Y of formula (III), wherein

    • X2 represents an amino acid residue selected from Ser, D-Ser and Aib,
    • X3 represents an amino acid residue selected from Gln and His,
    • X18 represents Leu
    • X20 represents an amino acid residue selected from His, Arg, Lys and Gln,
    • X21 represents an amino acid residue selected from Asp and Glu,
    • X28 represents an amino acid residue selected from Lys, Ser and Ala,
    • X32 represents an amino acid residue selected from Ser and Val.


A further embodiment relates to a group of prodrugs having a peptide moiety Y of formula (III), wherein

    • X2 represents Aib,
    • X3 represents an amino acid residue selected from Gln and His,
    • X18 represents an amino acid residue selected from His and Leu;
    • X20 represents an amino acid residue selected from His, Arg, Lys and Gln,
    • X21 represents an amino acid residue selected from Asp and Glu,
    • X28 represents an amino acid residue selected from Lys, Ser and Ala,
    • X32 represents an amino acid residue selected from Ser and Val.


A further embodiment relates to a group of prodrugs having a peptide moiety Y of formula (III), wherein

    • X2 represents Aib,
    • X3 represents His,
    • X18 represents Leu,
    • X20 represents Lys,
    • X21 represents an amino acid residue selected from Asp and Glu,
    • X28 represents an amino acid residue selected from Ser and Ala,
    • X32 represents an amino acid residue selected from Ser and Val.


A further embodiment relates to a group of prodrugs having a peptide moiety Y of formula (III), wherein

    • X2 represents an amino acid residue selected from Ser, D-Ser and Aib,
    • X3 represents His,
    • X18 represents an amino acid residue selected from His and Leu,
    • X20 represents an amino acid residue selected from His, Arg, Lys and Gln,
    • X21 represents an amino acid residue selected from Asp and Glu,
    • X28 represents an amino acid residue selected from Lys, Ser and Ala,
    • X32 represents an amino acid residue selected from Ser and Val.


A further embodiment relates to a group of prodrugs having a peptide moiety Y of formula (III), wherein

    • X2 represents an amino acid residue selected from Ser, D-Ser and Aib,
    • X3 represents Gln,
    • X18 represents Leu,
    • X20 represents Lys,
    • X21 represents an amino acid residue selected from Asp and Glu,
    • X28 represents Ala,
    • X32 represents Ser.


A further embodiment relates to a group of prodrugs having a peptide moiety Y of formula (III), wherein

    • X2 represents an amino acid residue selected from Ser, D-Ser and Aib,
    • X3 represents an amino acid residue selected from Gln and His,
    • X18 represents an amino acid residue selected from His and Leu,
    • X20 represents Lys,
    • X21 represents an amino acid residue selected from Asp and Glu,
    • X28 represents an amino acid residue selected from Lys, Ser and Ala,
    • X32 represents an amino acid residue selected from Ser and Val.


A further embodiment relates to a group of prodrugs having a peptide moiety Y of formula (III), wherein

    • X2 represents an amino acid residue selected from Ser, D-Ser and Aib,
    • X3 represents an amino acid residue selected from Gln and His,
    • X18 represents an amino acid residue selected from His and Leu,
    • X20 represents an amino acid residue selected from His, Arg, Lys and Gln,
    • X21 represents Asp,
    • X28 represents an amino acid residue selected from Lys, Ser and Ala,
    • X32 represents an amino acid residue selected from Ser and Val.


A further embodiment relates to a group of prodrugs having a peptide moiety Y of formula (III), wherein

    • X2 represents Aib,
    • X3 represents an amino acid residue selected from Gln and His,
    • X18 represents Leu,
    • X20 represents an amino acid residue selected from Lys and Gln,
    • X21 represents Glu,
    • X28 represents Ala,
    • X32 represents Ser.


A further embodiment relates to a group of prodrugs having a peptide moiety Y of formula (III), wherein

    • X2 represents an amino acid residue selected from Ser, D-Ser and Aib,
    • X3 represents an amino acid residue selected from Gln and His,
    • X18 represents an amino acid residue selected from His and Leu,
    • X20 represents an amino acid residue selected from His, Arg, Lys and Gln,
    • X21 represents an amino acid residue selected from Asp and Glu,
    • X28 represents Ala,
    • X32 represents an amino acid residue selected from Ser and Val.


A further embodiment relates to a group of prodrugs having a peptide moiety Y of formula (III), wherein

    • X2 represents an amino acid residue selected from Ser, D-Ser and Aib,
    • X3 represents an amino acid residue selected from Gln and His,
    • X18 represents an amino acid residue selected from His and Leu,
    • X20 represents an amino acid residue selected from His, Arg, Lys and Gln,
    • X21 represents an amino acid residue selected from Asp and Glu,
    • X28 represents an amino acid residue selected from Lys, Ser and Ala,
    • X32 represents an amino acid residue selected from Ser and Val.


A further embodiment relates to a group of prodrugs having a peptide moiety Y of formula (II) or (III), wherein

    • X2 represents an amino acid residue selected from Aib and D-Ser,
    • X3 represents His,
    • X18 represents an amino acid residue selected from Asp and Glu,
    • X28 represents an amino acid residue selected from Leu and Arg,
    • X20 represents Lys,
    • X21 represents an amino acid residue selected from Asp and Glu,
    • X28 represents an amino acid residue selected from Ser and Ala,
    • X32 represents an amino acid residue selected from Ser and Val.


A further embodiment relates to a group of prodrugs having a peptide moiety of formula (IV), wherein

    • X2 represents D-Ser,
    • X3 represents an amino acid residue selected from Gln and His,
    • X18 represents an amino acid residue selected from Arg and Leu,
    • X32 represents an amino acid residue selected from Ser and Val.


A further embodiment relates to a group of prodrugs having a peptide moiety of formula (IV), wherein

    • X2 represents an amino acid residue selected from Ser, D-Ser and Aib,
    • X3 represents His,
    • X18 represents an amino acid residue selected from Arg and Leu, particularly Leu,
    • X32 represents an amino acid residue selected from Ser and Val.


A further embodiment relates to a group of prodrugs having a peptide moiety of formula (IV), wherein

    • X2 represents D-Ser,
    • X3 represents Gln,
    • X18 represents Arg,
    • X32 represents an amino acid residue selected from Ser and Val.


A further embodiment relates to a group of prodrugs having a peptide moiety of formula (IV), wherein

    • X2 represents an amino acid residue selected from Ser, D-Ser and Aib,
    • X3 represents an amino acid residue selected from Gln and His,
    • X18 represents an amino acid residue selected from Arg and Leu,
    • X32 represents Ser.


A further embodiment relates to a group of prodrugs having a peptide moiety of formula (IVa)









(IVa)


H2N-His-Aib-His-Gly-Thr-Phe-Thr-Ser-Asp-Leu-Ser-





Lys-Gln-Leu-X15-Glu-Gln-Leu-Ala-Arg-Asp-Phe-Ile-





Glu-Trp-Leu-Ile-Bal-X29-Gly-X31-X32-Ser-X34-X35-





Pro-Pro-Pro-X39-R20







wherein
    • X15 represents an amino acid residue selected from Asp and Glu, (pref. Asp)
    • X29 represents an amino acid residue selected from Gly, D-Ala and Pro, (pref) Gly, D-Ala
    • X31 represents an amino acid residue selected from Pro, His and Trp, (pref. Pro)
    • X32 represents an amino acid residue selected from Ser, His, Pro and Arg, (pref. Ser, His, Pro),
    • X34 represents an amino acid residue selected from Gly and D-Ala,
    • X35 represents an amino acid residue selected from Ala, Pro and Lys, (pref. Ala, Pro)
    • X39 represents Ser or Pro-Pro-Pro,
    • or a salt or solvate thereof.


A further embodiment relates to a group of prodrugs having a peptide moiety of formula (IVa), wherein

    • X15 represents Asp,
    • X29 represents an amino acid residue selected from Gly, D-Ala and Pro,
    • X31 represents an amino acid residue selected from Pro, His and Trp,
    • X32 represents an amino acid residue selected from Ser, His, Pro and Arg,
    • X34 represents an amino acid residue selected from Gly and D-Ala,
    • X35 represents an amino acid residue selected from Ala, Pro and Lys,
    • X39 represents Ser.


A further embodiment relates to a group of prodrugs having a peptide moiety of formula (IVa), wherein

    • X15 represents an amino acid residue selected from Asp and Glu,
    • X29 represents Gly,
    • X31 represents an amino acid residue selected from Pro, His and Trp,
    • X32 represents an amino acid residue selected from Ser, His, Pro and Arg,
    • X34 represents an amino acid residue selected from Gly and D-Ala,
    • X35 represents an amino acid residue selected from Ala, Pro and Lys,
    • X39 represents Ser or Pro-Pro-Pro.


A further embodiment relates to a group of prodrugs having a peptide moiety of formula (IVa), wherein

    • X15 represents an amino acid residue selected from Asp and Glu,
    • X29 represents Gly,
    • X31 represents Pro,
    • X32 represents an amino acid residue selected from Ser, His and Pro,
    • X34 represents Gly,
    • X35 represents Ala,
    • X39 represents Ser.


A further embodiment relates to a group of prodrugs having a peptide moiety of formula (IVa), wherein

    • X15 represents Asp,
    • X29 represents D-Ala,
    • X31 represents Pro,
    • X32 represents Pro,
    • X34 represents D-Ala,
    • X35 represents an amino acid residue selected from Ala and Pro,
    • X39 represents Ser or Pro-Pro-Pro.


A further embodiment relates to a group of prodrugs having a peptide moiety of formula (IVa), wherein

    • X15 represents an amino acid residue selected from Asp and Glu,
    • X29 represents an amino acid residue selected from Gly, D-Ala and Pro,
    • X31 represents Pro,
    • X32 represents an amino acid residue selected from Ser, His, Pro and Arg,
    • X34 represents an amino acid residue selected from Gly and D-Ala,
    • X35 represents an amino acid residue selected from Ala, Pro and Lys,
    • X39 represents Ser or Pro-Pro-Pro.


A further embodiment relates to a group of prodrugs having a peptide moiety of formula (IVa), wherein

    • X15 represents Asp,
    • X29 represents Gly,
    • X31 represents His,
    • X32 represents Pro,
    • X34 represents Gly,
    • X35 represents an amino acid residue selected from Ala and Lys,
    • X39 represents Ser.


A further embodiment relates to a group of prodrugs having a peptide moiety of formula (IVa), wherein

    • X15 represents Asp,
    • X29 represents an amino acid residue selected from Gly and Pro,
    • X31 represents Pro,
    • X32 represents Ser,
    • X34 represents Gly,
    • X35 represents Ala,
    • X39 represents Ser.


A further embodiment relates to a group of prodrugs having a peptide moiety of formula (IVa), wherein

    • X15 represents an amino acid residue selected from Asp and Glu,
    • X29 represents an amino acid residue selected from Gly, D-Ala and Pro,
    • X31 represents an amino acid residue selected from Pro, His and Trp,
    • X32 represents Pro,
    • X34 represents an amino acid residue selected from Gly and D-Ala,
    • X35 represents an amino acid residue selected from Ala, Pro and Lys,
    • X39 represents Ser or Pro-Pro-Pro.


A further embodiment relates to a group of prodrugs having a peptide moiety of formula (IVa), wherein

    • X15 represents Asp,
    • X29 represents an amino acid residue selected from Gly and Pro,
    • X31 represents Pro,
    • X32 represents His,
    • X34 represents Gly,
    • X35 represents Ala,
    • X39 represents Ser.


A further embodiment relates to a group of prodrugs having a peptide moiety of formula (IVa), wherein

    • X15 represents an amino acid residue selected from Asp and Glu,
    • X29 represents an amino acid residue selected from Gly, D-Ala and Pro,
    • X31 represents an amino acid residue selected from Pro, His and Trp,
    • X32 represents an amino acid residue selected from Ser, His, Pro and Arg,
    • X34 represents Gly,
    • X35 represents an amino acid residue selected from Ala, Pro and Lys,
    • X39 represents Ser or Pro-Pro-Pro.


A further embodiment relates to a group of prodrugs having a peptide moiety of formula (IVa), wherein

    • X15 represents Asp,
    • X29 represents D-Ala,
    • X31 represents Pro,
    • X32 represents an amino acid residue selected from Ser and Pro,
    • X34 represents D-Ala,
    • X35 represents an amino acid residue selected from Ala and Pro,
    • X39 represents Ser or Pro-Pro-Pro.


A further embodiment relates to a group of prodrugs having a peptide moiety of formula (IVa), wherein

    • X15 represents an amino acid residue selected from Asp and Glu,
    • X29 represents an amino acid residue selected from Gly, D-Ala and Pro,
    • X31 represents an amino acid residue selected from Pro, His and Trp,
    • X32 represents an amino acid residue selected from Ser, His, Pro and Arg,
    • X34 represents an amino acid residue selected from Gly and D-Ala,
    • X35 represents Ala,
    • X39 represents Ser.


A further embodiment relates to a group of prodrugs having a peptide moiety of formula (IVa), wherein

    • X15 represents Asp,
    • X29 represents Gly,
    • X31 represents an amino acid residue selected from Pro and His,
    • X32 represents Pro,
    • X34 represents Gly,
    • X35 represents Lys,
    • X39 represents Ser.


A further embodiment relates to a group of prodrugs having a peptide moiety of formula (IVa), wherein

    • X15 represents Asp,
    • X29 represents an amino acid residue selected from Gly and D-Ala,
    • X31 represents Pro,
    • X32 represents Pro,
    • X34 represents an amino acid residue selected from Gly and D-Ala,
    • X35 represents Pro,
    • X39 represents Pro-Pro-Pro.


A further embodiment relates to a group of prodrugs having a peptide moiety of formula (IVa), wherein

    • X15 represents an amino acid residue selected from Asp and Glu,
    • X29 represents an amino acid residue selected from Gly, D-Ala and Pro,
    • X31 represents an amino acid residue selected from Pro, His and Trp,
    • X32 represents an amino acid residue selected from Ser, His, Pro and Arg,
    • X34 represents an amino acid residue selected from Gly and D-Ala,
    • X35 represents an amino acid residue selected from Ala, Pro and Lys,
    • X39 represents Ser.


In one embodiment Y refers to an GLP-1/Glucagon agonist of Seq. ID No 60.


In one embodiment Y refers to an GLP-1/Glucagon agonist selected from sequences Seq. ID No 4 to 60.


In one embodiment Y refers to an GLP-1/Glucagon agonist selected from sequences Seq. ID No 4 to 44.


In one embodiment Y refers to an GLP-1/Glucagon agonist selected from sequences Seq. ID No 4 to 22.


In one embodiment Y refers to an GLP-1/Glucagon agonist selected from sequences Seq. ID No 23 to 39.


In one embodiment Y refers to an GLP-1/Glucagon agonist selected from sequences Seq. ID No 40 to 44.


In one embodiment Y refers to an GLP-1/Glucagon agonist selected from sequences Seq. ID No 45 to 59.


In one embodiment Y refers to an GLP-1/Glucagon agonist selected from sequences Seq. ID No 18, 21 and 26.


In one embodiment Y refers to an GLP-1/Glucagon agonist selected from sequences Seq. ID No 18, 21, 26, 45, 48, 49 and 60.










TABLE 1 





SEQ



ID
sequence
















1
H-G-E-G-T-F-T-S-D-L-S-K-Q-M-E-E-E-A-V-R-L-F-I-E-W-L-K-N-G-G-P-S-



S-G-A-P-P-P-S-NH2





2
H-A-E-G-T-F-T-S-D-V-S-S-Y-L-E-G-Q-A-A-K-E-F-I-A-W-L-V-K-G-R-NH2





3
H-S-Q-G-T-F-T-S-D-Y-S-K-Y-L-D-S-R-R-A-Q-D-F-V-Q-W-L-M-N-T





4
H-S-Q-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-R-A-K-E-F-I-E-W-L-I-A-G-G-P-S-S-



G-A-P-P-P-S-NH2





5
H-S-Q-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-R-A-K-E-F-I-E-W-L-I-A-G-G-P-V-S-



G-A-P-P-P-S-NH2





6
H-dSer-Q-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-R-A-K-D-F-I-E-W-L-I-A-G-G-P-



S-S-G-A-P-P-P-S-NH2





7
H-dSer-Q-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-R-A-K-D-F-I-E-W-L-I-A-G-G-P-



V-S-G-A-P-P-P-S-NH2





8
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-R-A-K-D-F-I-E-W-L-I-A-G-G-P-S-



S-G-A-P-P-P-S-NH2





9
H-Aib-Q-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-R-A-Q-D-F-I-E-W-L-I-A-G-G-P-S-



S-G-A-P-P-P-S-NH2





10
H-Aib-Q-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-R-A-K-D-F-I-E-W-L-I-S-G-G-P-S-



S-G-A-P-P-P-S-NH2





11
H-Aib-Q-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-R-A-K-D-F-I-E-W-L-I-A-G-G-P-S-



S-G-A-P-P-P-S-NH2





12
H-Aib-Q-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-R-A-H-D-F-I-E-W-L-I-A-G-G-P-S-



S-G-A-P-P-P-S-NH2





13
H-Aib-Q-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-R-A-K-E-F-I-E-W-L-I-A-G-G-P-S-



S-G-A-P-P-P-S-NH2





14
H-Aib-Q-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-R-A-K-D-F-I-E-W-L-I-A-G-G-P-V-



S-G-A-P-P-P-S-NH2





15
H-Aib-Q-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-R-A-Q-D-F-I-E-W-L-I-S-G-G-P-S-



S-G-A-P-P-P-S-NH2





16
H-S-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-R-A-K-D-F-I-E-W-L-I-A-G-G-P-V-S-



G-A-P-P-P-S-NH2





17
H-dSer-Q-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-R-A-K-E-F-I-E-W-L-I-A-G-G-P-



V-S-G-A-P-P-P-S-NH2





18
H-dSer-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-R-A-K-D-F-I-E-W-L-I-A-G-G-P-



V-S-G-A-P-P-P-S-NH2





19
H-S-Q-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-R-A-K-D-F-I-E-W-L-I-A-G-G-P-V-



S-G-A-P-P-P-S-NH2





20
H-Aib-Q-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-R-A-K-E-F-I-E-W-L-I-S-G-G-P-S-



S-G-A-P-P-P-S-NH2





21
H-dSer-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-R-A-K-D-F-I-E-W-L-I-A-G-G-P-



S-S-G-A-P-P-P-S-NH2





22
H-dSer-Q-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-K-A-K-D-F-I-E-W-L-I-A-G-G-P-



S-S-G-A-P-P-P-S-





23
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-R-D-F-I-E-W-L-I-A-G-G-P-S-



S-G-A-P-P-P-S-NH2





24
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-H-D-F-I-E-W-L-I-A-G-G-P-S-



S-G-A-P-P-P-S-NH2





25
H-dSer-Q-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-K-D-F-I-E-W-L-I-A-G-G-P-



S-S-G-A-P-P-P-S-NH2





26
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-K-D-F-I-E-W-L-I-A-G-G-P-S-



S-G-A-P-P-P-S-NH2





27
H-S-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-K-D-F-I-E-W-L-I-A-G-G-P-S-S-



G-A-P-P-P-S-NH2





28
H-S-Q-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-K-D-F-I-E-W-L-I-A-G-G-P-S-S-



G-A-P-P-P-S-NH2





29
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-K-E-F-I-E-W-L-I-A-G-G-P-S-



S-G-A-P-P-P-S-NH2





30
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-K-D-F-I-E-W-L-I-S-G-G-P-S-



S-G-A-P-P-P-S-NH2





31
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-K-D-F-I-E-W-L-I-K-G-G-P-S-



S-G-A-P-P-P-S-NH2





32
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-K-D-F-I-E-W-L-I-A-G-G-P-V-



S-G-A-P-P-P-S-NH2





33
H-dSer-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-K-D-F-I-E-W-L-I-A-G-G-P-



S-S-G-A-P-P-P-S-NH2





34
H-S-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-K-D-F-I-E-W-L-I-A-G-G-P-S-S-



G-A-P-P-P-S-NH2





35
H-Aib-Q-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-K-E-F-I-E-W-L-I-A-G-G-P-S-



S-G-A-P-P-P-S-NH2





36
H-Aib-Q-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-K-D-F-I-E-W-L-I-A-G-G-P-S-



S-G-A-P-P-P-S-NH2





37
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-H-A-K-D-F-I-E-W-L-I-A-G-G-P-S-



S-G-A-P-P-P-S-NH2





38
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-Q-D-F-I-E-W-L-I-A-G-G-P-S-



S-G-A-P-P-P-S-NH2





39
H-Aib-Q-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-Q-E-F-I-E-W-L-I-A-G-G-P-S-



S-G-A-P-P-P-S-NH2





40
H-dSer-Q-G-T-F-T-S-D-L-S-K-L-L-D-E-Q-R-A-K-D-F-I-E-W-L-I-A-G-G-P-



S-S-G-A-P-P-P-S-NH2





41
H-Aib-H-G-T-F-T-S-D-L-S-K-L-L-D-E-Q-L-A-K-D-F-I-E-W-L-I-A-G-G-P-S-



S-G-A-P-P-P-S-NH2





42
H-dSer-Q-G-T-F-T-S-D-L-S-K-L-L-D-E-Q-R-A-K-D-F-I-E-W-L-I-A-G-G-P-



V-S-G-A-P-P-P-S-NH2





43
H-dSer-H-G-T-F-T-S-D-L-S-K-L-L-D-E-Q-L-A-K-D-F-I-E-W-L-I-A-G-G-P-



S-S-G-A-P-P-P-S-NH2





44
H-S-H-G-T-F-T-S-D-L-S-K-L-L-D-E-Q-L-A-K-D-F-I-E-W-L-I-A-G-G-P-S-S-



G-A-P-P-P-S-NH2





45
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-R-D-F-I-E-W-L-I-Bal-G-G-P-



S-S-G-A-P-P-P-S-NH2





46
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-R-D-F-I-E-W-L-I-Bal-G-G-P-



P-S-G-A-P-P-P-S-NH2





47
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-R-D-F-I-E-W-L-I-Bal-G-G-P-



P-S-G-P-P-P-P-P-P-P-NH2





48
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-R-D-F-I-E-W-L-I-Bal-dAla-



G-P-P-S-dAla-P-P-P-P-P-P-P-NH2





49
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-R-D-F-I-E-W-L-I-Bal-G-G-P-



H-S-G-A-P-P-P-S-NH2





50
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-R-D-F-I-E-W-L-I-Bal-P-G-P-



S-S-G-A-P-P-P-S-NH2





51
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-R-D-F-I-E-W-L-I-Bal-P-G-P-



P-S-G-A-P-P-P-S-NH2





52
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-R-D-F-I-E-W-L-I-Bal-P-G-P-



H-S-G-A-P-P-P-S-NH2





53
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-R-D-F-I-E-W-L-I-Bal-G-G-P-



P-S-G-K-P-P-P-S-NH2





54
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-R-D-F-I-E-W-L-I-Bal-G-G-H-



P-S-G-A-P-P-P-S-NH2





55
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-R-D-F-I-E-W-L-I-Bal-G-G-W-



P-S-G-A-P-P-P-S-NH2





56
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-R-D-F-I-E-W-L-I-Bal-G-G-H-



P-S-G-K-P-P-P-S-NH2





57
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-E-E-Q-L-A-R-D-F-I-E-W-L-I-Bal-G-G-P-



P-S-G-A-P-P-P-S-NH2





58
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-R-D-F-I-E-W-L-I-Bal-G-G-P-



R-S-G-A-P-P-P-S-NH2





59
H-Aib-H-G-T-F-T-S-D-L-S-K-Q-L-D-E-Q-L-A-R-D-F-I-E-W-L-I-Bal-dAla-



G-P-P-S-dAla-A-P-P-P-S-NH2





60
H-Aib-Q-G-T-F-T-S-D-L-S-K-L-L-E-E-Q-R-A-R-E-F-I-E-W-L-I-Bal-dAla-



G-P-P-S-dAla-A-P-P-P-S-NH2









Another embodiment is the peptide of Seq. ID No. 60 and its use as pharmaceutical.


In case the GLP-1/Glucagon agonist prodrugs comprising the compounds according to formula (I) contain one or more acidic or basic groups, the invention also comprises their corresponding pharmaceutically or toxicologically acceptable salts, in particular their pharmaceutically utilizable salts. Thus, the GLP-1/Glucagon agonist prodrugs comprising the compounds of the formula (I) which contain acidic groups can be used according to the invention, for example, as alkali metal salts, alkaline earth metal salts or as ammonium salts. More precise examples of such salts include sodium salts, potassium salts, calcium salts, magnesium salts or salts with ammonia or organic amines such as, for example, ethylamine, ethanolamine, triethanolamine or amino acids. GLP-1/Glucagon agonist prodrugs comprising the compounds of the formula (I) which contain one or more basic groups, i.e. groups which can be protonated, can be present and can be used according to the invention in the form of their addition salts with inorganic or organic acids. Examples for suitable acids include hydrogen chloride, hydrogen bromide, phosphoric acid, sulfuric acid, nitric acid, methanesulfonic acid, p-toluenesulfonic acid, naphthalenedisulfonic acids, oxalic acid, acetic acid, tartaric acid, lactic acid, salicylic acid, benzoic acid, formic acid, propionic acid, pivalic acid, diethylacetic acid, malonic acid, succinic acid, pimelic acid, fumaric acid, maleic acid, malic acid, sulfaminic acid, phenylpropionic acid, gluconic acid, ascorbic acid, isonicotinic acid, citric acid, adipic acid, and other acids known to the person skilled in the art. If the GLP-1/Glucagon agonist prodrugs comprising the compounds of the formula (I) simultaneously contain acidic and basic groups in the molecule, the invention also includes, in addition to the salt forms mentioned, inner salts or betaines (zwitterions). The respective salts according to GLP-1/Glucagon agonist prodrugs comprising the formula (I) can be obtained by customary methods which are known to the person skilled in the art like, for example by contacting these with an organic or inorganic acid or base in a solvent or dispersant, or by anion exchange or cation exchange with other salts. The present invention also includes all salts of the GLP-1/Glucagon agonist prodrugs comprising the compounds of the formula (I) which, owing to low physiological compatibility, are not directly suitable for use in pharmaceuticals but which can be used, for example, as intermediates for chemical reactions or for the preparation of pharmaceutically acceptable salts.


Process of Making


The peptides Y may be prepared by convenient methods known in the art.


The linkers L are prepared by methods as described in the examples and as disclosed in WO2009/095479, WO2011/012718 and WO2012/035139.


The hydrogel-linked GLP-1/Glucagon agonist prodrug of the present invention can be prepared by synthesizing the building blocks activated hyaluronic acid hydrogel Z-L1* and activated peptide linker conjugate L2*-L-Y.


Activated groups L1* and L2* are used to conjugate peptide to the polymers.


Scheme 1 shows different types of linking chemistries which can be used to conjugate the peptide with self-immolative linkers to the polymer. Thus, besides thiol-maleimide chemistry, other biorthogonal chemistries can be used. In scheme 1 the dashed lines indicates the positions where L1 and L2 are attached.


After loading the GLP-1/Glucagon agonist-linker conjugate to the functionalized hyaluronic acid hydrogel, all remaining functional groups are optionally capped with a suitable blocking reagent to prevent undesired side-reactions.


In the case of a functionalized maleimido group-containing HA-hydrogel, a thiol containing compound such as mercaptoethanol is a suitable blocking agent.




embedded image


embedded image


Another aspect of the present invention are functionalized intermediates comprising a GLP-1/Glucagon agonist-linker conjugate L2*-L-Y.


One embodiment of a GLP-1/Glucagon agonist-linker conjugate L2*-L-Y comprises a thiol functionalization, resulting in the formula

HS-L2-L-Y

    • wherein L2, L and Y have the meanings as described above.


One embodiment of thiol functionalized a GLP-1/Glucagon agonist-linker conjugate L2*-L-Y is a GLP-1/Glucagon agonist-linker conjugate of formula (VIII)




embedded image


One embodiment of thiol functionalized GLP-1/Glucagon agonist-linker conjugate L2*-L-Y is a GLP-1/Glucagon agonist-linker conjugate of formula (IX)




embedded image


One embodiment of thiol functionalized a GLP-1/Glucagon agonist-linker conjugate L2*-L-Y is a GLP-1/Glucagon agonist-linker conjugate of formula (X)




embedded image


Pharmaceutical Composition


Another aspect of the present invention is a pharmaceutical composition comprising a prodrug of the present invention or a pharmaceutically acceptable salt thereof together with a pharmaceutically acceptable excipient. The pharmaceutical composition is further described in the following paragraphs.


The composition of GLP-1/Glucagon agonist-hydrogel prodrug may be provided as a suspension composition or as a dry composition. In one embodiment the pharmaceutical composition of GLP-1/Glucagon agonist-hydrogel prodrug is a dry composition. Suitable methods of drying are, for example, spray-drying and lyophilization (freeze-drying). Preferably, the pharmaceutical composition of GLP-1/Glucagon agonist-hydrogel prodrug is dried by lyophilization.


In another embodiment the pharmaceutical composition of GLP-1/Glucagon agonist-hydrogel prodrug is a ready to use suspension.


In another embodiment the pharmaceutical composition of GLP-1/Glucagon agonist-hydrogel prodrug is a ready to use suspension wherein the prodrug is swollen in water/buffer to a concentration of 0.5 to 8% (w/v).


In another embodiment the pharmaceutical composition of GLP-1/Glucagon agonist-hydrogel prodrug is a ready to use suspension wherein the prodrug is swollen in water/buffer to a concentration of 1 to 4% (w/v).


In another embodiment the pharmaceutical composition of GLP-1/Glucagon agonist-hydrogel prodrug is a ready to use suspension wherein the prodrug is swollen in water/buffer to a concentration of 1.5 to 3% (w/v).


Preferably, the GLP-1/Glucagon agonist hydrogel prodrug is sufficiently dosed in the composition to provide therapeutically effective amount of GLP-1/Glucagon agonist for at least three days in one application. More preferably, one application of the GLP-1/Glucagon agonist hydrogel prodrug is sufficient for one week.


The pharmaceutical composition of GLP-1/Glucagon agonist-hydrogel prodrug according to the present invention contains one or more excipients.


Excipients used in parenteral compositions may be categorized as buffering agents, isotonicity modifiers, preservatives, stabilizers, anti-adsorption agents, oxidation protection agents, viscosifiers/viscosity enhancing agents, or other auxiliary agents. In some cases, these ingredients may have dual or triple functions. The compositions of GLP-1/Glucagon agonist-hydrogel prodrugs according to the present invention contain one or more than one excipient, selected from the groups consisting of:

  • (i) Buffering agents: physiologically tolerated buffers to maintain pH in a desired range, such as sodium phosphate, bicarbonate, succinate, histidine, citrate and acetate, sulphate, nitrate, chloride, pyruvate. Antacids such as Mg(OH)2 or ZnCO3 may be also used. Buffering capacity may be adjusted to match the conditions most sensitive to pH stability
  • (ii) Isotonicity modifiers: to minimize pain that can result from cell damage due to osmotic pressure differences at the injection depot. Glycerin and sodium chloride are examples. Effective concentrations can be determined by osmometry using an assumed osmolality of 285-315 mOsmol/kg for serum
  • (iii) Preservatives and/or antimicrobials: multidose parenteral preparations require the addition of preservatives at a sufficient concentration to minimize risk of patients becoming infected upon injection and corresponding regulatory requirements have been established. Typical preservatives include m-cresol, phenol, methylparaben, ethylparaben, propylparaben, butylparaben, chlorobutanol, benzyl alcohol, phenylmercuric nitrate, thimerosol, sorbic acid, potassium sorbate, benzoic acid, chlorocresol, and benzalkonium chloride
  • (iv) Stabilizers: Stabilisation is achieved by strengthening of the protein-stabilising forces, by destabilisation of the denatured stater, or by direct binding of excipients to the protein. Stabilizers may be amino acids such as alanine, arginine, aspartic acid, glycine, histidine, lysine, proline, sugars such as glucose, sucrose, trehalose, polyols such as glycerol, mannitol, sorbitol, salts such as potassium phosphate, sodium sulphate, chelating agents such as EDTA, hexaphosphate, ligands such as divalent metal ions (zinc, calcium, etc.), other salts or organic molecules such as phenolic derivatives. In addition, oligomers or polymers such as cyclodextrins, dextran, dendrimers, PEG or PVP or protamine or HSA may be used
  • (v) Anti-adsorption agents: Mainly ionic or inon-ionic surfactants or other proteins or soluble polymers are used to coat or adsorb competitively to the inner surface of the composition's or composition's container. E.g., poloxamer (Pluronic F-68), PEG dodecyl ether (Brij 35), polysorbate 20 and 80, dextran, polyethylene glycol, PEG-polyhistidine, BSA and HSA and gelatines. Chosen concentration and type of excipient depends on the effect to be avoided but typically a monolayer of surfactant is formed at the interface just above the CMC value
  • (vi) Lyo- and/or cryoprotectants: During freeze- or spray drying, excipients may counteract the destabilising effects caused by hydrogen bond breaking and water removal. For this purpose sugars and polyols may be used but corresponding positive effects have also been observed for surfactants, amino acids, non-aqueous solvents, and other peptides. Trehalose is particularly efficient at reducing moisture-induced aggregation and also improves thermal stability potentially caused by exposure of protein hydrophobic groups to water. Mannitol and sucrose may also be used, either as sole lyo/cryoprotectant or in combination with each other where higher ratios of mannitol:sucrose are known to enhance physical stability of a lyophilized cake. Mannitol may also be combined with trehalose. Trehalose may also be combined with sorbitol or sorbitol used as the sole protectant. Starch or starch derivatives may also be used
  • (vii) Oxidation protection agents: antioxidants such as ascorbic acid, ectoine, methionine, glutathione, monothioglycerol, morin, polyethylenimine (PEI), propyl gallate, vitamin E, chelating agents such as citric acid, EDTA, hexaphosphate, thioglycolic acid
  • (viii) Viscosifiers or viscosity enhancers: retard settling of the particles in the vial and syringe and are used in order to facilitate mixing and resuspension of the particles and to make the suspension easier to inject (i.e., low force on the syringe plunger). Suitable viscosifiers or viscosity enhancers are, for example, carbomer viscosifiers like Carbopol 940, Carbopol Ultrez 10, cellulose derivatives like hydroxypropylmethylcellulose (hypromellose, HPMC) or diethylaminoethyl cellulose (DEAE or DEAE-C), colloidal magnesium silicate (Veegum) or sodium silicate, hydroxyapatite gel, tricalcium phosphate gel, xanthans, carrageenans like Satia gum UTC 30, aliphatic poly(hydroxy acids), such as poly(D,L- or L-lactic acid) (PLA) and poly(glycolic acid) (PGA) and their copolymers (PLGA), terpolymers of D,L-lactide, glycolide and caprolactone, poloxamers, hydrophilic poly(oxyethylene) blocks and hydrophobic poly(oxypropylene) blocks to make up a triblock of poly(oxyethylene)-poly(oxypropylene)-poly(oxyethylene) (e.g. Pluronic®), polyetherester copolymer, such as a polyethylene glycol terephthalate/polybutylene terephthalate copolymer, sucrose acetate isobutyrate (SAIB), dextran or derivatives thereof, combinations of dextrans and PEG, polydimethylsiloxane, collagen, chitosan, polyvinyl alcohol (PVA) and derivatives, polyalkylimides, poly (acrylamide-co-diallyldimethyl ammonium (DADMA)), polyvinylpyrrolidone (PVP), glycosaminoglycans (GAGs) such as dermatan sulfate, chondroitin sulfate, keratan sulfate, heparin, heparan sulfate, hyaluronan, ABA triblock or AB block copolymers composed of hydrophobic A-blocks, such as polylactide (PLA) or poly(lactide-co-glycolide) (PLGA), and hydrophilic B-blocks, such as polyethylene glycol (PEG) or polyvinyl pyrrolidone. Such block copolymers as well as the abovementioned poloxamers may exhibit reverse thermal gelation behavior (fluid state at room temperature to facilitate administration and gel state above sol-gel transition temperature at body temperature after injection).
  • (ix) Spreading or diffusing agent: modifies the permeability of connective tissue through the hydrolysis of components of the extracellular matrix in the interstitial space such as but not limited to hyaluronic acid, a polysaccharide found in the intercellular space of connective tissue. A spreading agent such as but not limited to hyaluronidase temporarily decreases the viscosity of the extracellular matrix and promotes diffusion of injected drugs.
  • (x) Other auxiliary agents: such as wetting agents, viscosity modifiers, antibiotics, hyaluronidase. Acids and bases such as hydrochloric acid and sodium hydroxide are auxiliary agents necessary for pH adjustment during manufacture


In one embodiment the composition of GLP-1/Glucagon agonist-hydrogel prodrug contains one or more than one viscosifier and/or viscosity modifying agent.


In another embodiment the composition of GLP-1/Glucagon agonist-hydrogel prodrug contains hyaluronic acid as viscosifier and/or viscosity modifying agent.


In another embodiment the composition of GLP-1/Glucagon agonist-hydrogel prodrug comprises hyaluronic acid as viscosifier and/or viscosity modifying agent in a concentration of 5 to 30 wt %.


In another embodiment the composition of GLP-1/Glucagon agonist-hydrogel prodrug comprises hyaluronic acid as viscosifier and/or viscosity modifying agent of a molecular weight of 200 kDa to 6 million kDa.


In another embodiment the composition of GLP-1/Glucagon agonist-hydrogel prodrug comprises hyaluronic acid as viscosifier and/or viscosity modifying agent of a molecular weight of 500 kDa to 3 million kDa.


The term “excipient” preferably refers to a diluent, adjuvant, or vehicle with which the therapeutic is administered. Such pharmaceutical excipient can be sterile liquids. Water is a preferred excipient when the pharmaceutical composition is administered orally. Saline and aqueous dextrose are preferred excipients when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions are preferably employed as liquid excipients for injectable solutions.


Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. The composition, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsions, tablets, pills, capsules, powders, sustained-release formulations and the like. Such compositions will contain a therapeutically effective amount of the therapeutic, preferably in purified form, together with a suitable amount of excipient so as to provide the form for proper administration to the patient. The formulation should suit the mode of administration.


In a general embodiment a pharmaceutical composition of the present invention whether in dry form or as a suspension or in another form may be provided as single or multiple dose composition.


In one embodiment of the present invention, the dry composition of GLP-1/Glucagon agonist-hydrogel prodrug is provided as a single dose, meaning that the container in which it is supplied contains one pharmaceutical dose.


Thus, in another aspect of the present invention the composition is provided as a single dose composition.


In another aspect of the present invention the composition is comprised in a container. In one embodiment the container is a dual-chamber syringe. Especially the dry composition according to the present invention is provided in a first chamber of the dual-chamber syringe and reconstitution solution is provided in a second chamber of the dual-chamber syringe.


Prior to applying the dry composition of GLP-1/Glucagon agonist-hydrogel prodrug to a patient in need thereof, the dry composition is reconstituted. Reconstitution can take place in the container in which the dry composition of GLP-1/Glucagon agonist-hydrogel prodrug is provided, such as in a vial, syringe, dual-chamber syringe, ampoule, and cartridge. Reconstitution is done by adding a predefined amount of reconstitution solution to the dry composition. Reconstitution solutions are sterile liquids, such as water or buffer, which may contain further additives, such as preservatives and/or antimicrobials. If the GLP-1/Glucagon agonist-hydrogel prodrug composition is provided as single dose, the reconstituion solution may contain one or more preservative and/or antimicrobial. Preferably, the reconstitution solution is sterile water.


An additional aspect of the present invention relates to the method of administration of a reconstituted GLP-1/Glucagon agonist hydrogel prodrug composition. The GLP-1/Glucagon agonist hydrogel prodrug composition can be administered by methods of injection or infusion, including intradermal, subcutaneous, intramuscular, intravenous, intraosseous, and intraperitoneal.


A further aspect is a method of preparing a reconstituted composition comprising a therapeutically effective amount of an GLP-1/Glucagon agonist hydrogel prodrug, and optionally one or more pharmaceutically acceptable excipients, wherein the GLP-1/Glucagon agonist is transiently linked to a hydrogel, the method comprising the step of

    • contacting the composition of the present invention with a reconstitution solution.


Another aspect is a reconstituted composition comprising a therapeutically effective amount of a GLP-1/Glucagon agonist hydrogel prodrug, and optionally one or more pharmaceutically acceptable excipients, wherein the GLP-1/Glucagon agonist is transiently linked to a hydrogel obtainable by the method above.


Another aspect of the present invention is the method of manufacturing a dry composition of GLP-1/Glucagon agonist-hydrogel prodrug. In one embodiment, such suspension composition is made by

    • (i) admixing the GLP-1/Glucagon agonist-hydrogel prodrug with one or more excipients,
    • (ii) transferring amounts equivalent to single or multiple doses into a suitable container,
    • (iii) drying the composition in said container, and
    • (iv) sealing the container.


Suitable containers are vials, syringes, dual-chamber syringes, ampoules, and cartridges.


Another aspect is a kit of parts. When the administration device is simply a hypodermic syringe then the kit may comprise the syringe, a needle and a container comprising the dry GLP-1/Glucagon agonist-hydrogel prodrug composition for use with the syringe and a second container comprising the reconstitution solution. In more preferred embodiments, the injection device is other than a simple hypodermic syringe and so the separate container with reconstituted GLP-1/Glucagon agonist-hydrogel prodrug is adapted to engage with the injection device such that in use the liquid composition in the container is in fluid connection with the outlet of the injection device. Examples of administration devices include but are not limited to hypodermic syringes and pen injector devices. Particularly preferred injection devices are the pen injectors in which case the container is a cartridge, preferably a disposable cartridge.


A preferred kit of parts comprises a needle and a container containing the composition according to the present invention and optionally further containing a reconstitution solution, the container being adapted for use with the needle. Preferably, the container is a dual-chamber syringe.


In another aspect, the invention provides a cartridge containing a composition of GLP-1/Glucagon agonist-hydrogel prodrug as hereinbefore described for use with a pen injector device. The cartridge may contain a single dose or multiplicity of doses of GLP-1/Glucagon agonist.


In one embodiment of the present invention the suspension composition of GLP-1/Glucagon agonist-hydrogel prodrug does not only comprise an GLP-1/Glucagon agonist-hydrogel prodrug and one or more than one excipients, but also other biologically active agents, either in their free form or as prodrugs. Preferably, such additional one or more biologically active agent is a prodrug, more preferably a hydrogel prodrug. Such biologically active agents include, but are not limited to, compounds of the following classes:


Injectability


Preferably, the formulation can be administered by injection through a needle smaller than 0.26 mm inner diameter (26 Gauge), even more preferably through a needle smaller than 0.18 mm inner diameter (28 Gauge), and most preferably through a needle small than 0.16 mm inner diameter (30 Gauge).


It is understood that the terms “can be administered by injection”, “injectable” or “injectability” refer to a combination of factors such as a certain force applied to a plunger of a syringe containing the biodegradable HA hydrogel according to the invention swollen in a liquid at a certain concentration (w/v) and at a certain temperature, a needle of a given inner diameter connected to the outlet of such syringe, and the time required to extrude a certain volume of the biodegradable hydrogel according to the invention from the syringe through the needle.


In order to provide for injectability, a volume of 1 mL of the GLP-1/glucagon agonist prodrugs according to the invention swollen in water and contained in a syringe (holding a plunger of a diameter of 4.7 mm) can be extruded at room temperature within 10 seconds by applying a force of equal/less than 20 Newton through a needle of 26 gauge.


A preferred injectability is a volume of 1 mL of the GLP-1/glucagon agonist prodrugs according to the invention swollen in water and contained in a syringe (holding a plunger of a diameter of 4.7 mm) which can be extruded at room temperature within 10 seconds by applying a force of equal/less than 20 Newton through a needle of 30 gauge.


Suprisingly it was found that the HA carrier of the invention needs lesser force for injection the higher the peptide loading on the polymer is (FIG. 5).


In order to provide for injectability, a volume of 1 mL of the GLP-1/glucagon agonist prodrugs according to the invention swollen in water/buffer to a concentration of at least 1.5% (w/v) and contained in a syringe holding a plunger of a diameter of 4.7 mm can be extruded at room temperature within 10 seconds by applying a force of less than 30 Newton through a needle of 30 gauge.


More preferably injectability is achieved for a GLP-1/glucagon agonist prodrug according to the invention swollen in water/buffer to a concentration of at least 2% (w/v) by applying a force of less than 30 Newton through a needle of 30 gauge.


Most preferably injectability is achieved for a GLP-1/glucagon agonist prodrug according to the invention swollen in water/buffer to a concentration of at least 2% (w/v) by applying a force of less than 20 Newton through a needle of 30 gauge.


An important characteristic of the prodrug is the forming of a stable depot which stays its application site. The degradation of the polymer should start after release of the drug.


Combination Therapy


The prodrugs of the present invention, dual agonists for the GLP-1 and glucagon receptors, can be widely combined with other pharmacologically active compounds, such as all drugs mentioned in the Rote Liste 2015, e.g. with all weight-reducing agents or appetite suppressants mentioned in the Rote Liste 2015, chapter 1, all lipid-lowering agents mentioned in the Rote Liste 2015, chapter 58, all antihypertensives and nephroprotectives, mentioned in the Rote Liste 2015, or all diuretics mentioned in the Rote Liste 2015, chapter 36.


The active ingredient combinations can be used especially for a synergistic improvement in action. They can be applied either by separate administration of the active ingredients to the patient or in the form of combination products in which a plurality of active ingredients are present in one pharmaceutical preparation. When the active ingredients are administered by separate administration of the active ingredients, this can be done simultaneously or successively.


Most of the active ingredients mentioned hereinafter are disclosed in the USP Dictionary of USAN and International Drug Names, US Pharmacopeia, Rockville 2011.


Other active substances which are suitable for such combinations include in particular those which for example potentiate the therapeutic effect of one or more active substances with respect to one of the indications mentioned and/or which allow the dosage of one or more active substances to be reduced.


Therapeutic agents which are suitable for combinations include, for example, antidiabetic agents such as:


Insulin and Insulin derivatives, for example: Glargine/Lantus®, 270-330 U/mL of insulin glargine (EP 2387989 A), 300 U/mL of insulin glargine (EP 2387989 A), Glulisin/Apidra®, Detemir/Levemir®, Lispro/Humalog®/Liprolog®, Degludec/DegludecPlus, Aspart, basal insulin and analogues (e.g. LY-2605541, LY2963016, NN1436), PEGylated insulin Lispro, Humulin®, Linjeta, SuliXen®, NN1045, Insulin plus Symlin, PE0139, fast-acting and short-acting insulins (e.g. Linjeta, PH20, NN1218, HinsBet), (APC-002)hydrogel, oral, inhalable, transdermal and sublingual insulins (e.g. Exubera®, Nasulin®, Afrezza, Tregopil, TPM 02, Capsulin, Oral-Lyn®, Cobalamin® oral insulin, ORMD-0801, NN1953, NN1954, NN1956, VIAtab, Oshadi oral insulin). Additionally included are also those insulin derivatives which are bonded to albumin or another protein by a bifunctional linker.


GLP-1, GLP-1 analogues and GLP-1 receptor agonists, for example: Lixisenatide/AVE0010/ZP10/Lyxumia, Exenatide/Exendin-4/Byetta/Bydureon/ITCA 650/AC-2993, Liraglutide/Victoza, Semaglutide, Taspoglutide, Syncria/Albiglutide, Dulaglutide, rExendin-4, CJC-1134-PC, PB-1023, TTP-054, Langlenatide/HM-11260C, CM-3, GLP-1 Eligen, ORMD-0901, NN-9924, NN-9926, NN-9927, Nodexen, Viador-GLP-1, CVX-096, ZYOG-1, ZYD-1, GSK-2374697, DA-3091, MAR-701, MAR709, ZP-2929, ZP-3022, TT-401, BHM-034. MOD-6030, CAM-2036, DA-15864, ARI-2651, ARI-2255, Exenatide-XTEN and Glucagon-Xten.


DPP-4 inhibitors, for example: Alogliptin/Nesina, Trajenta/Linagliptin/BI-1356/Ondero/Trajenta/Tradjenta/Trayenta/Tradzenta, Saxagliptin/Onglyza, Sitagliptin/Januvia/Xelevia/Tesave/Janumet/Velmetia, Galvus/Vildagliptin, Anagliptin, Gemigliptin, Teneligliptin, Melogliptin, Trelagliptin, DA-1229, Omarigliptin/MK-3102, KM-223, Evogliptin, ARI-2243, PBL-1427, Pinoxacin.


SGLT2 inhibitors, for example: Invokana/Canaglifozin, Forxiga/Dapagliflozin, Remoglifozin, Sergliflozin, Empagliflozin, Ipragliflozin, Tofogliflozin, Luseogliflozin, LX-4211, Ertuglifozin/PF-04971729, RO-4998452, EGT-0001442, KGA-3235/DSP-3235, LIK066, SBM-TFC-039,


Biguanides (e.g. Metformin, Buformin, Phenformin), Thiazolidinediones (e.g. Pioglitazone, Rivoglitazone, Rosiglitazone, Troglitazone), dual PPAR agonists (e.g. Aleglitazar, Muraglitazar, Tesaglitazar), Sulfonylureas (e.g. Tolbutamide, Glibenclamide, Glimepiride/Amaryl, Glipizide), Meglitinides (e.g. Nateglinide, Repaglinide, Mitiglinide), Alpha-glucosidase inhibitors (e.g. Acarbose, Miglitol, Voglibose), Amylin and Amylin analogues (e.g. Pramlintide, Symlin).


GPR119 agonists (e.g. GSK-263A, PSN-821, MBX-2982, APD-597, ZYG-19, DS-8500), GPR40 agonists (e.g. Fasiglifam/TAK-875, TUG-424, P-1736, JTT-851, GW9508).


Other suitable combination partners are: Cycloset, inhibitors of 11-beta-HSD (e.g. LY2523199, BMS770767, RG-4929, BMS816336, AZD-8329, HSD-016, BI-135585), activators of glucokinase (e.g. TTP-399, AMG-151, TAK-329, GKM-001), inhibitors of DGAT (e.g. LCQ-908), inhibitors of protein tyrosinephosphatase 1 (e.g. Trodusquemine), inhibitors of glucose-6-phosphatase, inhibitors of fructose-1,6-bisphosphatase, inhibitors of glycogen phosphorylase, inhibitors of phosphoenol pyruvate carboxykinase, inhibitors of glycogen synthase kinase, inhibitors of pyruvate dehydrokinase, alpha2-antagonists, CCR-2 antagonists, SGLT-1 inhibitors (e.g. LX-2761).


One or more lipid lowering agents are also suitable as combination partners, such as for example: HMG-CoA-reductase inhibitors (e.g. Simvastatin, Atorvastatin), fibrates (e.g. Bezafibrate, Fenofibrate), nicotinic acid and the derivatives thereof (e.g. Niacin),


PPAR-(alpha, gamma or alpha/gamma) agonists or modulators (e.g. Aleglitazar),


PPAR-delta agonists, ACAT inhibitors (e.g. Avasimibe), cholesterol absorption inhibitors (e.g. Ezetimibe), Bile acid-binding substances (e.g. cholestyramine, colesevelam), ileal bile acid transport inhibitors, MTP inhibitors, or modulators of PCSK9.


HDL-raising compounds such as: CETP inhibitors (e.g. Torcetrapib, Anacetrapid, Dalcetrapid, Evacetrapid, JTT-302, DRL-17822, TA-8995) or ABC1 regulators.


Other suitable combination partners are one or more active substances for the treatment of obesity, such as for example: Sibutramine, Tesofensine, Orlistat, antagonists of the cannabinoid-1 receptor, MCH-1 receptor antagonists, MC4 receptor agonists, NPY5 or NPY2 antagonists (e.g. Velneperit), beta-3-agonists, leptin or leptin mimetics, agonists of the 5HT2c receptor (e.g. Lorcaserin), or the combinations of bupropione/naltrexone, bupropione/zonisamide, bupropione/phentermine or pramlintide/metreleptin.


Other Suitable Combination Partners are:


Further gastrointestinal peptides such as Peptide YY 3-36 (PYY3-36) or analogues thereof, pancreatic polypeptide (PP) or analogues thereof.


Glucagon receptor agonists or antagonists, GIP receptor agonists or antagonists, ghrelin antagonists or inverse agonists, Xenin and analogues thereof.


Moreover, combinations with drugs for influencing high blood pressure, chronic heart failure or atherosclerosis, such as e.g.: Angiotensin II receptor antagonists (e.g. telmisartan, candesartan, valsartan, losartan, eprosartan, irbesartan, olmesartan, tasosartan, azilsartan), ACE inhibitors, ECE inhibitors, diuretics, beta-blockers, calcium antagonists, centrally acting hypertensives, antagonists of the alpha-2-adrenergic receptor, inhibitors of neutral endopeptidase, thrombocyte aggregation inhibitors and others or combinations thereof are suitable.


Use


In another aspect, this invention relates to the use of a prodrug according to the invention or a physiologically acceptable salt thereof combined with at least one of the active substances described above as a combination partner, for preparing a medicament which is suitable for the treatment or prevention of diseases or conditions which can be affected by binding to the receptors for GLP-1 and glucagon and by modulating their activity.


Said compositions are for use in a method of treating or preventing diseases or disorders known for GLP-1/Glucagon agonist and GLP-1/Glucagon agonist agonists, for example, for treatment and prevention of hyperglycemia and for treatment and prevention of diabetes mellitus of any type, e.g. insulin-dependent diabetes mellitus, non insulin dependent diabetes mellitus, prediabetes or gestational diabetes mellitus, for prevention and treatment of metabolic syndrome and/or obesity and/or eating disorders, insulin resistance syndrome, lowering plasma lipid level, reducing the cardiac risk, reducing the appetite, reducing the body weight, etc.


The compounds of the invention are useful in the treatment or prevention of hepatosteatosis, preferably non-alcoholic liver-disease (NAFLD) and non-alcoholic steatohepatitis (NASH).


The use of the prodrugs according to the invention, or a physiologically acceptable salt thereof, in combination with one or more active substances may take place simultaneously, separately or sequentially.


The use of the prodrug according to the invention, or a physiologically acceptable salt thereof, in combination with another active substance may take place simultaneously or at staggered times, but particularly within a short space of time. If they are administered simultaneously, the two active substances are given to the patient together; if they are used at staggered times, the two active substances are given to the patient within a period of less than or equal to 12 hours, but particularly less than or equal to 6 hours.


Consequently, in another aspect, this invention relates to a medicament which comprises a prodrug according to the invention or a physiologically acceptable salt of such a compound and at least one of the active substances described above as combination partners, optionally together with one or more inert carriers and/or diluents.


The compound according to the invention, or physiologically acceptable salt or solvate thereof, and the additional active substance to be combined therewith may both be present together in one formulation, for example a suspension, or separately in two identical or different formulations, for example as so-called kit-of-parts.


Yet another aspect of the present invention is a method of treating, controlling, delaying or preventing in a mammalian patient, preferably in a human, in need of the treatment of one or more conditions comprising administering to said patient a therapeutically effective amount of a prodrug of the present invention or a pharmaceutical composition of the present invention or a pharmaceutically acceptable salt thereof.


EXAMPLES

Materials and Methods


Abbreviations employed are as follows:

  • AA amino acid
  • AcOH acetic acid
  • AcOEt ethyl acetate
  • cAMP cyclic adenosine monophosphate
  • Bn benzyl
  • Boc tert-butyloxycarbonyl
  • BOP (benzotriazol-1-yloxy)tris(dimethylamino)phosphonium hexafluorophosphate
  • BSA bovine serum albumin
  • tBu tertiary butyl
  • DBU 1,3-diazabicyclo[5.4.0]undecene
  • DCC N,N-dicyclohexylcarbodiimid
  • DCM dichloromethane
  • Dde 1-(4,4-dimethyl-2,6-dioxocyclohexylidene)-ethyl
  • ivDde 1-(4,4-dimethyl-2,6-dioxocyclohexylidene)3-methyl-butyl
  • DIC N,N′-diisopropylcarbodiimide
  • DIPEA N,N-diisopropylethylamine
  • DMAP dimethylamino-pyridine
  • DMEM Dulbecco's modified Eagle's medium
  • DMF dimethyl formamide
  • DMSO dimethylsulfoxide
  • DTT DL dithiotreitol
  • EDC 1-Ethyl-3-(3-dimethylaminopropyl)carbodiimid
  • EDT ethanedithiol
  • EDTA ethylenediaminetetraacetic acid
  • eq stoichiometric equivalent
  • EtOH ethanol
  • FBS fetal bovine serum
  • Fmoc fluorenylmethyloxycarbonyl
  • HATU O-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium
  • hexafluorophosphate
  • HBSS Hanks' Balanced Salt Solution
  • HBTU 2-(1H-benzotriazol-1-yl)-1,1,3,3-tetramethyl-uronium hexafluorophosphate
  • HEPES 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid
  • HOBt 1-hydroxybenzotriazole
  • HOSu N-hydroxysuccinimide
  • s HPLC High Performance Liquid Chromatography
  • HTRF Homogenous Time Resolved Fluorescence
  • IBMX 3-isobutyl-1-methylxanthine
  • LC/MS Liquid Chromatography/Mass Spectrometry
  • Mal 3-maleimido propyl
  • Mal-PEG6-NHS N-(3-maleimidopropyl)-21-amino-4,7,10,13,16,19-hexaoxa-heneicosanoic acid NHS ester
  • Me methyl
  • MeOH methanol
  • Mmt 4-methoxytrityl
  • MS mass spectrum/mass spectrometry
  • MTBE methyl tert.-butyl ether
  • MW molecular mass
  • NHSN-hydroxy succinimide
  • Palm palmitoyl
  • iPrOH 2-propanol
  • PBS phosphate buffered saline
  • PEG polyethylene glycole
  • PK pharmacokinetic
  • PyBOP benzotriazole-1-yl-oxy-tris-pyrrolidino-phosphonium hexafluorophosphate
  • Phth phthalimido
  • RP-HPLC reversed-phase high performance liquid chromatography
  • rpm rounds per minute
  • RT room temperature
  • SEC size exclusion chromatography
  • TCEP tris(2-carboxyethyl)phosphine hydrochloride
  • TES triethylsilane
  • TFA trifluoroacetic acid
  • s THF tetrahydrofurane
  • TMEDA N,N,N′N′-tetramethylethylene diamine
  • Tris tris(hydroxymethyl)aminomethane
  • Trt trityl
  • UPLC Ultra Performance Liquid Chromatography
  • UV ultraviolet
  • V volume


Example 1

General Synthesis of Peptidic Compounds


Materials:


Different Rink-Amide resins (4-(2′,4′-Dimethoxyphenyl-Fmoc-aminomethyl)-phenoxyacetamido-norleucylaminomethyl resin, Merck Biosciences; 4-[(2,4-Dimethoxyphenyl)(Fmoc-amino)methyl]phenoxy acetamido methyl resin, Agilent Technologies) were used for the synthesis of peptide amides with loadings in the range of 0.3-0.4 mmol/g.


Fmoc protected natural amino acids were purchased from Protein Technologies Inc., Senn Chemicals, Merck Biosciences, Novabiochem, Iris Biotech, Nagase or Bachem. The following standard amino acids were used throughout the syntheses: Fmoc-L-Ala-OH, Fmoc-L-Arg(Pbf)-OH, Fmoc-L-Asn(Trt)-OH, Fmoc-L-Asp(OtBu)-OH, Fmoc-L-Cys(Trt)-OH, Fmoc-L-Gln(Trt)-OH, Fmoc-L-Glu(OtBu)-OH, Fmoc-Gly-OH, Fmoc-L-His(Trt)-OH, Fmoc-L-Ile-OH, Fmoc-L-Leu-OH, Fmoc-L-Lys(Boc)-OH, Fmoc-L-Met-OH, Fmoc-L-Phe-OH, Fmoc-L-Pro-OH, Fmoc-L-Ser(tBu)-OH, Fmoc-L-Thr(tBu)-OH, Fmoc-L-Trp(Boc)-OH, Fmoc-L-Tyr(tBu)-OH, Fmoc-L-Val-OH.


In addition, the following special amino acids were purchased from the same suppliers as above: Fmoc-L-Lys(ivDde)-OH, Fmoc-Aib-OH, Fmoc-D-Ser(tBu)-OH, Fmoc-D-Ala-OH, Boc-L-His(Boc)-OH (available as toluene solvate) and Boc-L-His(Trt)-OH, Fmoc-L-Nle-OH, Fmoc-L-Met(O)—OH, Fmoc-L-Met(O2)-OH, Fmoc-(S)MeLys(Boc)-OH, Fmoc-(R)MeLys(Boc)-OH, Fmoc-(S)MeOrn(Boc)-OH and Boc-L-Tyr(tBu)-OH.


The solid phase peptide syntheses were performed for example on a Prelude Peptide Synthesizer (Protein Technologies Inc) or similar automated synthesizer using standard Fmoc chemistry and HBTU/DIPEA activation. DMF was used as the solvent. Deprotection: 20% piperidine/DMF for 2×2.5 min. Washes: 7×DMF. Coupling 2:5:10 200 mM AA/500 mM HBTU/2M DIPEA in DMF 2× for 20 min. Washes: 5×DMF.


All the peptides that had been synthesized were cleaved from the resin with King's cleavage cocktail consisting of 82.5% TFA, 5% phenol, 5% water, 5% thioanisole, 2.5% EDT. The crude peptides were then precipitated in diethyl or diisopropyl ether, centrifuged, and lyophilized. Peptides were analyzed by analytical HPLC and checked by ESI mass spectrometry. Crude peptides were purified by a conventional preparative HPLC purification procedure.


Analytical HPLC/UPLC


Method A: detection at 215 nm




  • column: Aeris Peptide, 3.6 μm, XB-C18 (250×4.6 mm) at 60° C.

  • solvent: H2O+0.1% TFA: ACN+0.1% TFA (flow 1.5 ml/min)

  • gradient: 90:10 (0 min) to 90:10 (3 min) to 10:90 (43 min) to 10:90 (48 min) to 90:10 (49 min) to 90:10 (50 min)


    Method B: detection at 220 nm

  • column: Zorbax, 5 μm, C18 (250×4.6 mm) at 25° C.

  • solvent: H2O+0.1% TFA: 90% ACN+10% H2O+0.1% TFA (flow 1.0 ml/min)

  • gradient: 100:0 (0 min) to 98:2 (2 min) to 30:70 (15 min) to 5:95 (20 min) to 0:100 (25 min) to 0:100 (30 min) to 98:2 (32 min) to 98:2 (35 min)


    Method C1: detection at 210-225 nm, optionally coupled to a mass analyser Waters LCT Premier, electrospray positive ion mode

  • column: Waters ACQUITY UPLC® BEH™ C18 1.7 μm (150×2.1 mm) at 50° C.

  • solvent: H2O+1% FA: ACN+1% FA (flow 0.5 ml/min)

  • gradient: 95:5 (0 min) to 95:5 (1.80 min) to 80:20 (1.85 min) to 80:20 (3 min) to 60:40 (23 min) to 25:75 (23.1 min) to 25:75 (25 min) to 95:5 (25.1 min) to 95:5 (30 min)


    Method C2: detection at 210-225 nm, optionally coupled to a mass analyser Waters LCT Premier, electrospray positive ion mode

  • column: Waters ACQUITY UPLC® BEH™ C18 1.7 μm (150×2.1 mm) at 50° C.

  • solvent: H2O+1% FA: ACN+1% FA (flow 0.6 ml/min)

  • gradient: 95:5 (0 min) to 95:5 (1 min) to 65:35 (2 min) to 65:35 (3 min) to 45:55 (23 min) to 25:75 (23.1 min) to 25:75 (25 min) to 95:5 (25.1 min) to 95:5 (30 min)


    Method C3: detection at 210-225 nm, optionally coupled to a mass analyser Waters LCT Premier, electrospray positive ion mode

  • column: Waters ACQUITY UPLC® BEH™ C18 1.7 μm (150×2.1 mm) at 50° C.

  • solvent: H2O+1% FA: ACN+1% FA (flow 1 ml/min)

  • gradient: 95:5 (0 min) to 95:5 (1 min) to 65:35 (2 min) to 65:35 (3 min) to 45:55 (20 min) to 2:98 (20.1 min) to 2:98 (25 min) to 95:5 (25.1 min) to 95:5 (30 min)


    Method C4:


    detection at 210-225 nm, optionally coupled to a mass analyser Waters LCT Premier, electrospray positive ion mode

  • column: Waters ACQUITY UPLC® BEH™ C18 1.7 μm (150×2.1 mm) at 50° C.

  • solvent: H2O+1% FA: ACN+1% FA (flow 1 ml/min)

  • gradient: 95:5 (0 min) to 95:5 (1.80 min) to 80:20 (1.85 min) to 80:20 (3 min) to 60:40 (23 min) to 2:98 (23.1 min) to 2:98 (25 min) to 95:5 (25.1 min) to 95:5 (30 min)


    Method D: detection at 214 nm

  • column: Waters X-Bridge C18 3.5 μm 2.1×150 mm

  • solvent: H2O+0.5% TFA: ACN (flow 0.55 ml/min)

  • gradient: 90:10 (0 min) to 40:60 (5 min) to 1:99 (15 min)


    Method E: detection at 210-225 nm, optionally coupled to a mass analyser Waters LCT Premier, electrospray positive ion mode

  • column: Waters ACQUITY UPLC® BEH™ C18 1.7 μm (150×2.1 mm) at 50° C.

  • solvent: H2O+1% FA: ACN+1% FA (flow 0.9 ml/min)

  • gradient: 95:5 (0 min) to 95:5 (2 min) to 35:65 (3 min) to 65:35 (23.5 min) to 5:95 (24 min) to 95:5 (26 min) to 95:5 (30 min)



General Preparative HPLC Purification Procedure:


The crude peptides were purified either on an Äkta Purifier System or on a Jasco semiprep HPLC System. Preparative RP-C18-HPLC columns of different sizes and with different flow rates were used depending on the amount of crude peptide to be purified. Acetonitrile+0.05 to 0.1% TFA (B) and water+0.05 to 0.1% TFA (A) were employed as eluents. Alternatively, a buffer system consisting of acetonitrile and water with minor amounts of acetic acid was used. Product-containing fractions were collected and lyophilized to obtain the purified product, typically as TFA or acetate salt.


Solubility and Stability-Testing of Exendin-4 Derivatives


Prior to the testing of solubility and stability of a peptide batch, its content was determined. Therefore, two parameters were investigated, its purity (HPLC-UV) and the amount of salt load of the batch (ion chromatography).


Example 2

For solubility testing, the target concentration was 1.0 mg/mL pure compound. Therefore, solutions from solid samples were prepared in different buffer systems with a concentration of 1.0 mg/mL compound based on the previously determined content. HPLC-UV was performed after 2 h of gentle agitation from the supernatant, which was obtained by 20 min of centrifugation at 4000 rpm.


The solubility was then determined by comparison with the UV peak areas obtained with a stock solution of the peptide at a concentration of 2 mg/mL in pure water or a variable amount of acetonitrile (optical control that all of the compound was dissolved). This analysis also served as starting point (t0) for the stability testing.


Example 3

For stability testing, an aliquot of the supernatant obtained for solubility was stored for 7 days at 25° C. or 40° C. After that time course, the sample was centrifuged for 20 min at 4000 rpm and the supernatant was analysed with HPLC-UV.


For determination of the amount of the remaining peptide, the peak areas of the target compound at t0 and t7 were compared, resulting in “% remaining peptide”, following the equation

% remaining peptide=[(peak area peptide t7)×100]/peak area peptide t0.


The amount of soluble degradation products was calculated from the comparison of the sum of the peak areas from all observed impurities reduced by the sum of peak areas observed at t0 (i.e. to determine the amount of newly formed peptide-related species). This value was given in percentual relation to the initial amount of peptide at t0, following the equation:

% soluble degradation products={[(peak area sum of impurities t7)−(peak area sum of impurities t0)]×100}/peak area peptide t0


The potential difference from the sum of “% remaining peptide” and “% soluble degradation products” to 100% reflects the amount of peptide which did not remain soluble upon stress conditions following the equation

% precipitate=100−([% remaining peptide]+[% soluble degradation products])


This precipitate includes non-soluble degradation products, polymers and/or fibrils, which have been removed from analysis by centrifugation.


The chemical stability is expressed as “% remaining peptide”.


Anion Chromatography


Instrument: Dionex ICS-2000, pre/column: Ion Pac AG-18 2×50 mm (Dionex)/AS18 2×250 mm (Dionex), eluent: aqueous sodium hydroxide, flow: 0.38 mL/min, gradient: 0-6 min: 22 mM KOH, 6-12 min: 22-28 mM KOH, 12-15 min: 28-50 mM KOH, 15-20 min: 22 mM KOH, suppressor: ASRS 300 2 mm, detection: conductivity.


As HPLC/UPLC method, method D or E has been used.


Example 4: In Vitro Data on GLP-1 and Glucagon Receptor

Potencies of peptidic compounds at the GLP-1 and glucagon receptors were determined by exposing cells expressing human glucagon receptor (hGlucagon R) or human GLP-1 receptor (hGLP-1 R) to the listed compounds at increasing concentrations and measuring the formed cAMP as described in example 27.


The results are shown in Table 2:









TABLE 2







EC50 values of exendin-4 derivatives at GLP-1


and Glucagon receptors (indicated in pM)












SEQ ID
EC50
EC50
SEQ ID
EC50
EC50


NO
hGLP-1R
hGlucagon-R
NO
hGLP-1R
hGlucagon-R















4
1.7
6.2
24
1.2
11.1


5
2.7
5.0
25
4.6
259.0


6
4.9
24.3
26
0.9
33.8


7
8.5
20.6
27
1.0
44.8


8
1.2
5.3
28
1.1
57.7


9
1.5
35.6
29
0.4
21.3


10
1.6
47.5
30
0.5
10.9


11
0.9
68.0
31
0.6
24.6


12
1.0
35.6
32
1.2
29.8


13
1.4
44.9
33
2.5
94.1


14
1.6
77.5
34
1.1
41.5


15
1.0
18.3
35
1.0
555.0


16
3.7
8.2
36
1.0
581.0


17
6.0
10.1
37
1.4
58.1


18
2.6
7.9
38
0.8
11.5


19
1.7
4.7
39
1.0
401.0


20
0.8
26.7
40
3.5
12.1


21
1.9
8.9
41
2.2
25.7


22
4.4
26.1
43
2.0
53.9


23
1.2
3.9
44
1.8
20.0


45
2.1
7.9
46
2.4
8.0


47
5.3
13.4
48
2.1
10.0


49
1.8
6.5
50
1.8
4.2


51
2.3
3.7
52
2.1
4.2


53
2.4
10.5
54
1.4
7.7


55
2.4
10.5
56
1.9
9.4


57
1.4
4.7
58
1.4
11.5


59
n.a.
n.a.
60
1.8
6.5









Linker Synthesis
Example 5
Synthesis of Linker Reagent 5c

Linker reagent 5c was synthesized according to the following scheme:




embedded image


Synthesis of Linker Reagent Intermediate 5a

m-Methoxytrityl chloride (3 g, 9.71 mmol) was dissolved in DCM (20 mL) and added dropwise to a solution of ethylenediamine (6.5 mL, 97.1 mmol) in DCM (20 mL). After two hours the solution was poured into diethyl ether (300 mL) and washed three times with 30/1 (v/v) brine/0.1 M NaOH solution (50 ml each) and once with brine (50 mL). The organic phase was dried over Na2SO4 and volatiles were removed under reduced pressure. Mmt-protected intermediate (3.18 g, 9.56 mmol) was used in the next step without further purification.


The Mmt-protected intermediate (3.18 g, 9.56 mmol) was dissolved in anhydrous DCM (30 mL). 6-(S-Tritylmercapto)hexanoic acid (4.48 g, 11.47 mmol), PyBOP (5.67 g, 11.47 mmol) and DIPEA (5.0 mL, 28.68 mmol) were added and the mixture was agitated for 30 min at RT. The solution was diluted with diethyl ether (250 mL) and washed three times with 30/1 (v/v) brine/0.1 M NaOH solution (50 mL each) and once with brine (50 mL). The organic phase was dried over Na2SO4 and volatiles were removed under reduced pressure. 5a was purified by flash chromatography.


Yield: 5.69 g (8.09 mmol).


MS: m/z 705.4=[M+H]+(MW calculated=705.0).


Synthesis of Linker Reagent Intermediate 5b

To a solution of 5a (3.19 g, 4.53 mmol) in anhydrous THF (50 mL) was added BH3.THF (1 M solution, 8.5 mL, 8.5 mmol) and the solution was stirred for 16 h at RT. Further BH3.THF (1 M solution, 14 mL, 14 mmol) was added and stirred for 16 h at RT. The reaction was quenched by addition of methanol (8.5 mL). N,N-dimethyl-ethylenediamine (3 mL, 27.2 mmol) was added and the solution was heated to reflux and stirred for three h. Reaction mixture was allowed to cool down to RT and was then diluted with ethyl acetate (300 mL), washed with saturated, aqueous Na2CO3 solution (2×100 mL) and saturated, aqueous NaHCO3 solution (2×100 mL). The organic phase was dried over Na2SO4 and volatiles were removed under reduced pressure to obtain crude amine intermediate (3.22 g).


The amine intermediate (3.22 g) was dissolved in DCM (5 mL). Boc2O (2.97 g, 13.69 mmol) dissolved in DCM (5 mL) and DIPEA (3.95 mL, 22.65 mmol) were added and the mixture was agitated at RT for 30 min. The mixture was purified by flash chromatography to obtain the crude Boc- and Mmt-protected intermediate (3.00 g).


MS: m/z 791.4=[M+H]+, 519.3=[M-Mmt+H]+ (MW calculated=791.1).


0.4 M aqueous HCl (48 mL) was added to a solution of the Boc- and Mmt-protected intermediate in acetonitrile (45 mL). The mixture was diluted with acetonitrile (10 mL) and stirred for 1 h at RT. Subsequently, the pH value of the reaction mixture was adjusted to 5.5 by addition of 5 M NaOH solution. Acetonitrile was removed under reduced pressure and the aqueous solution was extracted with DCM (4×100 mL). The combined organic phases were dried over Na2SO4 and volatiles were removed under reduced pressure. Crude 5b was used in the next step without further purification.


Yield: 2.52 g (3.19 mmol).


MS: m/z 519.3=[M+H]+ (MW calculated=519.8 g/mol).


Synthesis of Linker Reagent 5c

Intermediate 5b (985 mg, 1.9 mmol) and p-nitrophenyl chloroformate (330 mg, 2.5 mmol) were dissolved in anhydrous THF (10 mL). DIPEA (0.653 mL, 3.7 mmol) was added and the mixture was stirred for 2 h at RT. The solution was acidified by addition of acetic acid (1 mL). 5c was purified by RP-HPLC.


Yield: 776 mg, (1.13 mmol).


MS m/z 706.3=[M+Na]+ (MW calculated=706.3).


Synthesis of Peptide Linker Reagent

a) Peptide Synthesis


The solid phase peptide syntheses were performed for example on a Prelude Peptide Synthesizer (Protein Technologies Inc) or similar automated synthesizer using standard Fmoc chemistry and HBTU/DIPEA activation. DMF was used as the solvent. Deprotection: 20% piperidine/DMF for 2×2.5 min. Washes: 7×DMF. Coupling 2:5:10 200 mM AA/500 mM HBTU/2M DIPEA in DMF 2× for 20 min. Washes: 5×DMF.


b) N-Terminal Elongation with D-Ala


0.9 mmol resin bound peptide (equivalent to 4 g resin) synthesized as described in step a with a free amino group at the N-terminus was split into five equal portions. Each portion was suspended in 15 ml DMF and subsequently 2.5 eq. Fmoc-D-Ala-OH, 2.5 eq. HATU, 2.5 eq. HOAt and 2.5 eq. DIPEA were added. The mixture was agitated for 16 h at ambient temperature. Then the reaction mixture was removed by filtration and the resin was washed 3 times with 18 ml DMF, 18 ml DCM, 18 ml iso-propanol, 18 ml diethyl ether. The remaining solvents were removed in vacuo.


c) Fmoc Deprotection, Linker Attachment and Cleavage


0.37 mmol resin bound peptide (equivalent to 1.6 g resin) synthesized as described in step b with a free amino group at the N-terminus was split into two equal portions. Each portion was suspended in 12 ml of a 20% solution of piperidine in DMF and agitated for 5 min. The solvent was removed and the procedure was repeated twice.


The resin bound peptide was washed 5 times with 12 ml DMF.


Then the resin was suspended in 10 ml DMF and 2.5 eq. of tert-butyl (2-(((4-nitrophenoxy)carbonyl)amino)ethyl)(6-(tritylthio)hexyl)carbamate 5c and 2.5 eq. DIPEA were added. The mixture was agitated for 16 h at ambient temperature. Then the reaction mixture was removed by filtration and the resin was washed 3 times with 15 ml DMF, 15 ml DCM, 15 ml iso-propanol, 15 ml diethyl ether. The remaining solvents were removed in vacuo. The reaction was tested for completion by a Kaisertest


Then a mixture of TFA/DTT/TIS/H2O/thioanisole/Bu4NBr (100/3/2/3/1/0.05) was added and the mixture was agitated for 3.5 h. The mixture was filtrated and the resin was washed with 1 ml TFA. The combined filtrates were added to 100 ml cooled diethyl ether. The precipitate was isolated by centrifugation and it was washed 2 times with 100 ml diethyl ether.


The crude product was purified via preparative HPLC on a Waters column (XBridge, BEH130, Prep C18 5 μM) using an acetonitrile/water gradient (both buffers with 0.1% TFA). The purified intermediate was immediately lyophilized and stored in an Ar atmosphere or used directly for the next step.


Example 6
Synthesis of GLP-1/Glucagon Agonist Linker Reagent 6d (Ala Linker)

GLP-1/Glucagon agonist linker reagent 6d was synthesized according to the following scheme:




embedded image


Synthesis of GLP-1/Glucagon agonist linker reagent intermediate 6a: Fully side chain protected GLP-1/Glucagon agonist with free N-terminus on resin (2.00 g, 0.2 mmol, loading approximately 0.1 mmol/g) was transferred into a 20 mL syringe equipped with a filter frit. 8 mL of anhydrous DMF was drawn into the syringe and the syringe was shaken (600 rpm) for 15 min in order to pre-swell the resin. The solvent was discarded, and a solution of Fmoc-D-alanine-OH (187 mg, 0.6 mol), PyBOP (312 mg, 0.6 mmol), and DIPEA (174 μL, 1.0 mmol) in anhydrous DMF (4 mL) was drawn into the syringe. The syringe was shaken at RT and 600 rpm for 60 min. The solution was discharged, and the resin was washed ten times with DMF.


Fmoc-deprotection was performed as described above.


Synthesis of GLP-1/Glucagon Agonist Linker Reagent Intermediate 6b

A solution of 5c (137 mg, 0.4 mmol) in anhydrous DMF (3 mL) was added to the resin 6a (0.2 mmol), followed by a solution of DIPEA (80 μL, 0.46 mmol) in anhydrous DMF (4.5 mL), and the reaction mixture was shaken (600 rpm) at 22° C. for 15 hours.


The resin was washed ten times with DMF and ten times with DCM and dried in vacuo.


Synthesis of GLP-1/Glucagon Agonist Linker Reagent Intermediate 6c

3-Nitro-2-pyridine-sulfenyl chloride (48 mg, 0.25 mmol) was given into a syringe containing 6b (0.05 mmol, 0.5 g). Anhydrous DCM (4 mL) was drawn into the syringe and the mixture was shaken (600 rpm) at RT. After 2 h the solution was discarded and the resin was washed 14 times with DCM and dried in vacuo.


Synthesis of GLP-1/Glucagon Agonist Linker Reagent Intermediate 6d

In a round bottom flask o-cresol (1.5 mL), thioanisole (1.5 mL), DTT (1.125 g), TES (1.125 mL), and water (1.5 mL) were dissolved in TFA (37.5 mL). 6c (0.15 mmol, 1.5 g) was added to the stirred (250-350 rpm) solution at RT in order to obtain a homogeneous suspension. Stirring was continued for 45 min. The solution was separated from the resin beads by filtration, the beads were washed with TFA twice (2 mL each) and the washing solutions were combined with the filtrate. TFA was removed from the combined solutions in a stream of nitrogen.


Crude 6d was precipitated from the concentrated solution (approx. 10 mL) by addition of diethyl ether (30 mL) and vigorous shaking. After centrifugation (2 min, 5000 rpm) the supernatant was discarded and the precipitate was washed with diethyl ether twice (20 mL each).


Dried precipitate was dissolved in a solution of TCEP (114 mg, 0.39 mmol) in 30 ml 1/19 (v/v) acetonitrile/water containing 0.01% TFA (v/v). Mixture was incubated for 15 hours at RT. 6d was purified by RP-HPLC as described in Materials and Methods using a 150×30 mm Waters XBridge™ BEH300 C18 10 μm column and a flow of 40 ml/min.


Up to 12 mL of the mixture were loaded on the column. The elution was performed using a linear gradient from 5% to 30% solvent B (5 min) followed by a linear gradient from 30% to 35% solvent B (40 min). Fractions containing product 6d were pooled and lyophilized. Purity: 86% (215 nm)


Yield: 85.2 mg (19.2 μmol, starting from 2.00 g resin).


MS m/z 1486.7=[M+3H]3+, (MW calculated=4460.0 g/mol).


Example 7 (Asn Linker)
Synthesis of Linker Reagent 7f

Linker reagent 7f was synthesized according to the following scheme:




embedded image


To a cooled (0° C.) solution of N-Methyl-N-boc-ethylendiamine (0.5 mL, 2.79 mmol) and NaCNBH3 (140 mg, 2.23 mmol) in MeOH (10 mL) and acetic acid (0.5 mL) was added a solution of 2,4,6-trimethoxybenzaldehyde (0.547 mg, 2.79 mmol) in EtOH (10 mL). The mixture was stirred at RT for 2 h, acidified with 2 M HCl (1 mL) and neutralized with saturated aqueous Na2CO3 (50 mL). Evaporation of all volatiles, DCM extraction of the resulting aqueous slurry and concentration of the organic fractions yielded N-Methyl-N-boc-N′-tmob-ethylendiamine (7a) as a crude oil which was purified by RP-HPLC.


Yield: 593 mg (1.52 mmol)


MS: m/z 377.35=[M+Na]+, (calculated=377.14).


N-Fmoc-N-Me-Asp(OtBu)-OH (225 mg, 0.529 mmol) was dissolved in DMF (3 mL) and 7a (300 mg, 0.847 mmol), HATU (201 mg, 0.529 mmol), and collidine (0.48 mL, 3.70 mmol) were added. The mixture was stirred at RT for 2 h to yield 7b. For fmoc deprotection, piperidine (0.22 mL, 2.16 mmol) was added and stirring was continued for 1 h. Acetic acid (1 mL) was added, and 7c was purified by RP-HLPC.


Yield: 285 mg (0.436 mmol as TFA salt)


MS: m/z 562.54=[M+Na]+, (calculated=562.67).


6-Tritylmercaptohexanoic acid (0.847 g, 2.17 mmol) was dissolved in anhydrous DMF (7 mL). HATU (0.825 g, 2.17 mmol), and collidine (0.8 mL, 6.1 mmol) and 7c (0.78 g, 1.44 mmol) were added. The reaction mixture was stirred for 60 min at RT, acidified with AcOH (1 mL) and purified by RP-HPLC. Product fractions were neutralized with saturated aqueous NaHCO3 and concentrated. The remaining aqueous phase was extracted with DCM and 7d was isolated upon evaporation of the solvent.


Yield: 1.4 g (94%)


MS: m/z 934.7=[M+Na]+, (calculated=934.5).


To a solution of 7d (1.40 mg, 1.53 mmol) in MeOH (12 mL) and H2O (2 mL) was added LiOH (250 mg, 10.4 mmol) and the reaction mixture was stirred for 14 h at 70° C. The mixture was acidified with AcOH (0.8 mL) and 7e was purified by RP-HPLC. Product fractions were neutralized with saturated aqueous NaHCO3 and concentrated. The aqueous phase was extracted with DCM and 7e was isolated upon evaporation of the solvent.


Yield: 780 mg (60%)


MS: m/z 878.8=[M+Na]+, (calculated=878.40).


To a solution of 7e (170 mg, 0.198 mmol) in anhydrous DCM (4 mL) were added DCC (123 mg, 0.59 mmol) and N-hydroxy-succinimide (114 mg, 0.99 mmol), and the reaction mixture was stirred at RT for 1 h. The mixture was filtered, and the filtrate was acidified with 0.5 mL AcOH and 7f purified by RP-HPLC. Product fractions were neutralized with saturated aqueous NaHCO3 and concentrated. The remaining aqueous phase was extracted with DCM and 7f was isolated upon evaporation of the solvent.


Yield: 154 mg (0.161 mmol)


MS: m/z 953.4=[M+H]+, (calculated=953.43).


Alternatively, linker reagent 7f was synthesized according to the following procedure: Alternative reaction scheme:




embedded image


To a solution of N-Methyl-N-boc-ethylenediamine (2 g, 11.48 mmol) and NaCNBH3 (819 mg, 12.63 mmol) in MeOH (20 mL) was added 2,4,6-trimethoxybenzaldehyde (2.08 mg, 10.61 mmol) portion wise. The mixture was stirred at RT for 90 min, acidified with 3 M HCl (4 mL) and stirred further 15 min. The reaction mixture was added to saturated NaHCO3 solution (200 mL) and extracted 5× with CH2Cl2. The combined organic phases were dried over Na2SO4 and the solvents were evaporated in vacuo. The resulting N-Methyl-N-boc-N′-tmob-ethylenediamine (7a) was completely dried in high vacuum and used in the next reaction step without further purification.


Yield: 3.76 g (11.48 mmol, 89% purity, 7a: double Tmob protected product=8:1)


MS: m/z 355.22=[M+H]+, (calculated=354.21).


To a solution of 7a (2 g, 5.65 mmol) in CH2Cl2 (24 ml) COMU (4.84 g, 11.3 mmol), N-Fmoc-N-Me-Asp(OBn)-OH (2.08 g, 4.52 mmol) and collidine (2.65 mL, 20.34 mmol) were added. The reaction mixture was stirred for 3 h at RT, diluted with CH2Cl2 (250 mL) and washed 3× with 0.1 M H2SO4 (100 ml) and 3× with brine (100 ml). The aqueous phases were re extracted with CH2Cl2 (100 ml). The combined organic phases were dried over Na2SO4, filtrated and the residue concentrated to a volume of 24 mL. 7 g was purified using flash chromatography.


Yield: 5.31 g (148%, 6.66 mmol)


MS: m/z 796.38=[M+H]+, (calculated=795.37).


To a solution of 7 g [5.31 g, max. 4.51 mmol ref. to N-Fmoc-N-Me-Asp(OBn)-OH] in THF (60 mL) DBU (1.8 mL, 3% v/v) was added. The solution was stirred for 12 min at RT, diluted with CH2Cl2 (400 ml) and washed 3× with 0.1 M H2SO4 (150 ml) and 3× with brine (150 ml). The aqueous phases were re extracted with CH2Cl2 (100 ml). The combined organic phases were dried over Na2SO4 and filtrated. 7h was isolated upon evaporation of the solvent and used in the next reaction without further purification.


MS: m/z 574.31=[M+H]+, (calculated=573.30).


7h (5.31 g, 4.51 mmol, crude) was dissolved in acetonitrile (26 mL) and COMU (3.87 g, 9.04 mmol), 6-Tritylmercaptohexanoic acid (2.12 g, 5.42 mmol) and collidine (2.35 mL, 18.08 mmol) were added. The reaction mixture was stirred for 4 h at RT, diluted with CH2Cl2 (400 ml) and washed 3× with 0.1 M H2SO4 (100 ml) and 3× with brine (100 ml). The aqueous phases were re extracted with CH2Cl2 (100 ml). The combined organic phases were dried over Na2SO4, filtrated and 7i was isolated upon evaporation of the solvent. Product 7i was purified using flash chromatography.


Yield: 2.63 g (62%, 94% purity)


MS: m/z 856.41=[M+H]+, (calculated=855.41).


To a solution of 7i (2.63 g, 2.78 mmol) in i-PrOH (33 mL) and H2O (11 mL) was added LiOH (267 mg, 11.12 mmol) and the reaction mixture was stirred for 70 min at RT. The mixture was diluted with CH2Cl2 (200 ml) and washed 3× with 0.1 M H2SO4 (50 ml) and 3× with brine (50 ml). The aqueous phases were re-extracted with CH2Cl2 (100 ml). The combined organic phases were dried over Na2SO4, filtrated and 7e was isolated upon evaporation of the solvent. 7j was purified using flash chromatography.


Yield: 2.1 g (88%)


MS: m/z 878.4=[M+Na]+, (calculated=878.40).


To a solution of 7e (170 mg, 0.198 mmol) in anhydrous DCM (4 mL) were added DCC (123 mg, 0.59 mmol), and a catalytic amount of DMAP. After 5 min N-hydroxy-succinimide (114 mg, 0.99 mmol) was added and the reaction mixture was stirred at RT for 1 h. The reaction mixture was filtered, the solvent was removed in vacuo and the residue was taken up in 90% acetonitrile plus 0.1% TFA (3.4 ml). The crude mixture was purified by RP-HPLC. Product fractions were neutralized with 0.5 M pH 7.4 phosphate buffer and concentrated. The remaining aqueous phase was extracted with DCM and 7f was isolated upon evaporation of the solvent.


Yield: 154 mg (81%)


MS: m/z 953.4=[M+H]+, (calculated=953.43).


Example 8
Synthesis of Linker Reagent 8e

Linker reagent 8e was synthesized according to the following scheme:




embedded image


Synthesis of linker reagent intermediate 8b was performed under nitrogen atmosphere. A solution of amine 8a (1.69 g, 4.5 mmol, for preparation see WO-A 2009/133137) in 30 mL THF (dry, mol. sieve) was cooled to 0° C. Butyl chloroformate (630 μl, 4.95 mmol) in 3 mL THF (dry, mol. sieve) and DIPEA (980 μl, 5.63 mmol) were added. Mixture was stirred for 10 min at 0° C., cooling was removed and mixture stirred for further 20 min at RT. 1 M LiAlH4 in THF (9 mL, 9 mmol) was added and mixture was refluxed for 1.5 h. Reaction was quenched by slowly adding methanol (11 mL) and 100 mL sat. Na/K tartrate solution. Mixture was extracted with ethyl acetate, organic layer was dried over Na2SO4 and solvent was evaporated under reduced pressure. Crude product 8b (1.97 g) was used in the next step without further purification.


MS: m/z 390.2=[M+H]+ (MW calculated=389.6).


A solution of crude product 8b (1.97 g), N-(bromoethyl)-phthalimide (1.43 g, 5.63 mmol) and K2CO3 (1.24 g, 9.0 mmol) in 120 mL acetonitrile was refluxed for 6 h. 60 mL of a sat. NaHCO3 solution was added and mixture was extracted 3× with ethyl acetate. Combined organics were dried (Na2SO4) and solvent was removed under reduced pressure. Phthalimide 8c was purified on silica by using heptane (containing 0.02 NEt3) and an ascending amount of ethyl acetate (containing 0.02% NEt3) as eluents.


Yield: 0.82 g (1.46 mmol)


MS: m/z 563.3=[M+H]+ (MW calculated=562.8).


Phthalimide 8c (819 mg 1.46 mmol) was dissolved in 35 mL ethanol and hydrazine hydrate (176 μl, 3.64 mmol) was added. Mixture was refluxed for 3 h. Precipitate was filtered off. Solvent was removed under reduced pressure and residue was treated with 15 mL dichloromethane. Precipitate was filtered off and dichloromethane was removed under reduced pressure. Residue was purified by RP HPLC. Pooled HPLC fractions were adjusted to pH 7 by adding NaHCO3 and extracted several times with dichloromethane. Combined organics were dried (Na2SO4) and solvent was removed under reduced pressure to yield amine 8d.


Yield: 579 mg (1.34 mmol)


MS: m/z 433.3=[M+H]+(MW calculated=432.7).


Para-nitrophenyl chloroformate (483 mg, 2.40 mmol) was dissolved in 10 mL dichloromethane (dry, mol. sieve). A solution of amine 8d (1.00 g, 2.31 mmol) in 5 mL dichloromethane (dry, mol. sieve) and 1.8 mL of sym-collidine were added and mixture was stirred at room temperature for 40 min. Dichloromethane was removed under reduced pressure, residue was acidified with acetic acid and purified by RP-HPLC to yield para-nitrophenyl carbamate 8e.


Yield: 339 mg (0.57 mmol)


MS: m/z 598.3=[M+H]+ (MW calculated=597.8).


Synthesis of Peptide-Linker—Polymer Conjugates
Example 9

Synthesis of GLP/Glucagon agonist thiol linker 1 was synthesized according to the following scheme:




embedded image


To 400 mg of the resin bound Fmoc-protected Exendin-4 Seq. ID. 26 on rink resin in a peptide synthesis vessel was added 4 ml of 20% piperidine in DMF and the reaction mixture was stirred for 5 minutes. The process was repeated three times. The resin bound peptide was subsequently washed with 4 ml of DMF three times.


To 80 mg (2.5 eqv.) of the linker 1a and 60 mg (3.0 eqv.) of PyBOP was added 5 mL of anhydrous DMF. To this solution was added 16 μl (3.0 eqv.) of diisoproyldiethyamine (DIPEA). The resulting solution was added to above processed resin bound Exendin-4 Seq ID. 3126-resin. The mixture was allowed to proceed for 18 hr. at 25° C. At the end of this time, the resin filtered and washed with anhydrous DMF (5 ml×5), dichloromethane (5 ml×5), isopropanol (5 ml×5) and diethyl ether (5 ml×5).


The above resin bound peptide-linker conjugate was treated with 10 ml of a solution containing trifluoroacetic acid (TFA)/triethylsilane(TES)/dithiothreitol (DTT)/Thioanisole/water (100/2/3/1/2). The reaction mixture was shaken at 25° C. for 3 hr. At the end of reaction, the solvent was filtered off and the resin was washed with dichloromethane (5×10 ml). The filtrate was concentrated under reduced pressure. The residue was poured into 50 ml ice-cold ether. White solid precipitated out. The mixture was centrifuged and decanted. The solid was washed with diethyl ether (2×20 ml). The peptide-linker was purified by reverse phase HPLC (30×100 mm C18 column, 25-40% AcCN/H2O w/0.1% TFA in 20 min.). The appropriate fractions were collected and evaporated to dryness yielding 16 mg of the desired product as a white solid. The product was identified my mass spectrometry (molecular ion peak, m/z 1469, z=3).


Example 10

GLP/Glucagon thiol linker 2 was synthesized according to the following scheme:




embedded image


To 1.8 g of the resin bound Fmoc-protected Exendin-4 Seq ID. 26 on rink resin in a peptide synthesis vessel was added 20 ml of 20% piperidine in DMF and the reaction mixture was stirred for 5 minutes. The process was repeated three times. The resin bound peptide was subsequently washed with 20 ml of DMF three times.


To 163 mg (2.5 eqv.) Fmoc-D-Ala-OH, 199 mg (2.5 eqv.) of HATU and 71 mg (2.5 eqv.) of HOAt was added 15 ml of anhydrous DMF. To this solution was added 92 μl (2.5 eqv.) of diisoproyldiethyamine (DIPEA). The resulting solution was added to above processed resin bound Exendin-4 Seq ID. 26-resin. The mixture was allowed to proceed for 16 hr. at 25° C. At the end of this time, the resin filtered and washed with anhydrous DMF (20 ml×5), dichloromethane (20 ml×5), isopropanol (20 ml×5) and diethyl ether (20 ml×5).


To the above resin bound Fmoc-protected D-Ala-Exendin-4-Seq ID. 26 on rink resin in a peptide synthesis vessel was added 15 ml of 20% piperidine in DMF and the reaction mixture was stirred for 5 minutes. The process was repeated three times. The resin bound peptide was subsequently washed with 15 ml of DMF three times.


To 352 mg (2.5 eqv.) of the linker 2a was added 15 mL of anhydrous DMF. To this solution was added 92 μl (2.5 eqv.) of diisoproyldiethyamine (DIPEA). The resulting solution was added to above processed resin bound D-Ala-Exendin-4 Seq ID. 26-resin. The mixture was allowed to proceed for 16 hr. at 25° C. At the end of this time, the resin filtered and washed with anhydrous DMF (15 ml×5), dichloromethane (15 ml×5), isopropanol (15 ml×5) and diethyl ether (15 ml×5).


The above resin bound peptide-linker conjugate was treated with 40 ml of a solution containing trifluoroacetic acid (TFA)/triethylsilane(TES)/dithiothreitol (DTT)/Thioanisole/water (100/2/3/1/2). The reaction mixture was shaken at 25° C. for 1 hr. At the end of reaction, the solvent was filtered. The filtrate was poured into 400 ml ice-cold ether. White solid precipitated out. The mixture was centrifuged and decanted. The solid was washed with diethyl ether (2×200 ml). The peptide-linker was purified by reverse phase HPLC (30×100 mm C18 column, 25-40% AcCN/H2O w/0.1% TFA in 20 min.). The appropriate fractions were collected and evaporated to dryness yielding 104 mg of the desired product as a white solid. The product was identified my mass spectrometry (molecular ion peak, m/z 1465.3, z=3 and m/z 1099.2, z=4).


Synthesis of Hyaloronic Acid Hydrogels
Example 11

Divinyl sulfone crosslinked hyaluronic acid was synthesized according to the following scheme:




embedded image


Example 11a

To 0.2M sodium hydroxide (168.9 g) was added sodium chloride (23.4 g) with stirring until dissolved. To the solution under rapid mechanical stirring was added sodium hyaluronate (25.4 g, 400-500 KDa) which continued for 2h. The resulting polymer solution has a concentration of ˜12% w/w. A solution of divinylsulfone (0.41 mL, 0.48 g) in isopropanol (1.6 mL) was prepared and added (5×0.4 mL) over ˜30 sec. The mixture was stirred for an additional 2 min and poured into a 23×28×6.5 cm glass tray and sealed with a plastic cover. After standing at RT for 4h the gel was transferred as a single piece to a solution of 1M hydrochloric acid (100.1 g) in 0.9% saline (3 kg). It was agitated gently at RT. After 24h the pH of the solution was 2.28. The solution discarded leaving a gel (416.2 g). To the gel was then added 0.9% saline (3 kg) and it was agitated gently at RT for 18h. To the mixture was added 1M sodium hydroxide (9.7 mL) at 0, 2, 4, 6 and 8h. The gel was gently agitated for a further 24h at RT at which time the pH of the gel was 6.65. The gel was stored at 2-8° C. for 120h and then 10 mM sodium phosphate solution pH 7.4 (2 L) was added. The gel was agitated for an additional 21 h and the wash discarded leaving a gel (1036.2 g) with a final polymer concentration of 2.4%.


Example 11b. Alternative Synthesis of Divinylsulphone Crosslinked Hyaluronic Acid

To 35 g of sodium hyaluronate was added 946 mL of sterile water. The reaction mixture was kept at 2-8° C. for 7 days, during which time a clear solution has formed. To this solution was added 1M 111 mL of 1.0 M sodium hydroxide solution and the resulting reaction mixture was stirred vigorously for 5 min. The reaction mixture was kept at 2-8° C. for 90 min. Subsequently a suspension of 6.7 mL of divinylsulphone in 10 mL of sterile water was to the polymer solution and the resulting reaction mixture was stirred vigorously for 5 minutes. Subsequently, reaction mixture was stored at 2-8° C. for 150 minutes followed by for 90 minutes at 25° C. The polymer gel thus formed was washed with 0.9% sterile saline for four days. The pH of the suspension was adjusted to 7.0 with either 1.) M NaOH or 1.0 M HCl. The final concentration of the gel suspension was 0.58%.


Example 12
Synthesis of 1-(tert-butoxycarbonyl) amino 3-(3-maleimidopropyl) aminopropane



embedded image


In 250 ml round bottomed flask were taken 3.0 g of 1-(tert-Butoxycarbonyl) amino 3-aminopropane) and 100 mL of anhydrous chloroform. The reaction mixture was stirred at 25° C. until a clear solution was formed. To this solution was added N-succinimidyl 3-maleimidopropionate (5.05 g) with stirring until dissolved followed 3.42 mL of diisoproylethylamine. The resulting reaction mixture was stirred at 25° C. for 18h. The solution was washed with 1M hydrochloric acid (50 mL), 10% brine (50 mL), saturated sodium bicarbonate (50 mL), semi-saturated brine (50 mL). The organic phase was isolated and was dried over anhydrous sodium sulfate. After removing the sodium sulfate by filtration, the solution was concentrated under reduced pressure and the residue was purified by silica gel column chromatography using ethyl acetate: hexanes gradient as the mobile phase. Removal of the solvent under reduced pressure followed by vacuum drying offered the desired product as an off white solid (4.03 g).


Example 13
Synthesis of 1-(tert-Butyloxycarbonyl)amino 8-(3-maleimidopropyl) amino-3,6-dioxaoctane

In 100 ml round bottomed flask were taken 1-(tert-butyloxycarbonyl) amino 8-amino-3,6-dioxaoctane (1.007 g) and 25 mL of anhydrous acetonitrile (25 mL). The reaction mixture was stirred under nitrogen until dissolved. To this solution was added N-succinimidyl 3-maleimidopropionate (1.302 g) and the reaction mixture was stirred under nitrogen at 25° C. for 6h. At the end of this time, the solvent was removed under reduced pressure. The residue was purified silica gel column chromatography using dichloromethane containing 2-6% of methanol. After removing the solvent under reduced pressure and drying the residue under vacuum yielded 1.08 g of the desired product as an off white solid.


Example 14
Synthesis of 3-(3-maleimido-propyl) aminopropane Functionalized Hyaluronic Acid



embedded image


To 15 g of divinyl sulfone crosslinked suspension (Example 1) was added 45 ml of deionized (DI) water and the resulting suspension was stirred at 25° C. for 10 minutes. After adding 45 ml of ethanol to the suspension, it was stirred for additional 60 min. It was followed by addition of 60 mL of ethanol and 10 minute of stirring. To this suspension was added 0.24 g of 4-(4,6-dimethoxy-1,3,5-triazin-2-yl)-4-methyl-morpholinium chloride (DMT-MM) dissolved in 5 mL of ethanol. The resulting reaction mixture was stirred at 25° for 60 mins. To 70 mg of 1-(tert-Butoxycarbonyl) amino 3-(3-maleimidopropyl) aminopropane (example 2) dissolved in 2 mL of anhydrous dichloromethane was added 2 mL of trifluoroacetic acid. The resulting reaction was stirred at room temperature for 2h. The solution was concentrated under reduced pressure. The resulting residue was treated with 3 mL of methanol and evaporated to dryness. After repeating this process one more time the residue was dissolved in ethanol (5 mL) and the pH adjusted to 6-6.5 with 10% N-methylmorpholine in ethanol. The resulting solution was added to the above hyaluronic acid suspension. The vial was rinsed with ethanol (5 mL) and added to the slurry. After stirring for 18h at 25° C., saturated brine (2 mL) was added to the reaction mixture. The suspension was treated with ethanol (3×30 mL, 2×15 mL) to precipitate the polymer gel. The reaction mixture was centrifuged at 4000 rpm and the supernatant decanted. The residue was hydrated in DI water (20 mL) for ˜20 min and was precipitated from ethanol (4×10 mL). Glucosamine assay method suggests the degree of substitution to be 19 mole %.


Example 15
Synthesis of 3-(3-maleimido-propyl) aminopropane Functionalized Hyaluronic Acid

To 20 g of divinyl sulfone crosslinked suspension (Example 1) was added 60 ml of deionized (DI) water and the resulting suspension was stirred at 25° C. for 10 minutes. After adding 60 ml of ethanol to the suspension, it was stirred for additional 60 min. It was followed by addition of 60 mL of ethanol and 10 minute of stirring. To this suspension was added 0.325 g of DMT-MM dissolved in 10 mL of ethanol. The resulting reaction mixture was stirred at 25° for 60 mins. To 0.38 g of 1-(tert-Butoxycarbonyl) amino 3-(3-maleimidopropyl) aminopropane (example 2) dissolved in 2.5 mL of anhydrous dichloromethane was added 2.5 mL of trifluoroacetic acid. The resulting reaction was stirred at room temperature for 2h. The solution was concentrated under reduced pressure. The resulting residue was treated with 3 mL of methanol and evaporated to dryness. After repeating this process one more time, the residue was dissolved in ethanol (5 mL) and the pH adjusted to 6-6.5 with 10% N-methylmorpholine in ethanol. The resulting solution was added to the above hyaluronic acid suspension. The vial was rinsed with ethanol (5 mL) and added to the slurry. After stirring for 18h at 25° C., saturated brine (2 mL) was added to the reaction mixture. The suspension was treated with ethanol (3×30 mL, 2×15 mL) to precipitate the polymer gel. The reaction mixture was centrifuged at 4000 rpm and the supernatant decanted. The residue was hydrated in DI water (20 mL) for ˜20 min and was precipitated from ethanol (4×10 mL). Glucosamine assay method suggests the degree of substitution to be 24 mole %.


Example 16a

General Method for the Synthesis of 3-(3-Maleimido-Propyl) Aminopropane Functionalized HA Hydrogel


To appropriate amount divinylsulphone crosslinked HA suspension (Example 11b) was added sterile saline to obtain a gel concentration of ˜1% w/v. The resulting suspension was stirred at 25° C. for 15-30 minutes. A water miscible organic solvent (preferably ethanol) was added to the suspension and the resulting suspension was stirred for additional 30-60 min. To this suspension was added appropriate amount of an ethanolic solution of 4-(4,6-dimethoxy-1,3,5-triazin-2-yl)-4-methyl-morpholinium chloride (DMT-MM). The container containing DMT-MM solution was rinsed twice with ethanol and the washings were added to the above suspension. The resulting reaction mixture was stirred at 25° C. for 90 minutes. Appropriate amount of 1-(tert-butoxycarbonyl) amino 3-(3-maleimidopropyl) aminopropane was dissolved in dichloromethane. To this solution was added trifluoroacetic acid to give a 1:1 (v/v) solution. After stirring at room temperature for 60-90 minutes, the reaction mixture was evaporated to dryness under reduced pressure. The residue was dissolved in ethanol and was added to the above suspension. The container containing maleimide derivative was rinsed twice with ethanol and the washings were added to the suspension. The pH of the suspension was adjusted to pH 6.4-6.6 using an organic or inorganic base (for example 10% N-methylmorpholine in ethanol). After stirring for 16-20h at 25° C., the suspension was treated with ethanol to a volume of 60-65% v/v. The solvent was removed from the reaction mixture either by centrifugation at 120G followed by decanting the supernatant or by applying a slight overpressure of N2 gas to the system and filtering through a glass frit or filter membrane. The residue was subsequently treated with sterile 20 mM succinate saline (0.9%) at pH 3.8 for ˜15-20 min and was precipitated by adding ethanol to a volume of 60-65% v/v. The solvent is removed from the reaction mixture by following the above procedure. This procedure was repeated one more time.


Example 16b
Synthesis of 3-(3-maleimido-propyl) aminopropane Functionalized HA Hydrogels at a Degree of Substitution of 20 Mole %

To 5.65 g of the HA hydrogel suspension (Example 11b) was added 7.5 mL of sterile saline and the resulting suspension was gently stirred for 15 minutes. To this suspension was added 3 mL of ethanol and the resulting reaction mixture was gently stirred for 60 min. To this suspension was added 94 mg of DMT-MM dissolved in 3 mL of ethanol. The vial containing the DMT-MM was washed with ethanol (2×1 mL) and the washings were added to the suspension. The reaction mixture was allowed to stir gently at ambient temperature for at 90 minutes. To 21.7 mg of 1-(tert-Butoxycarbonyl) amino 3-(3-maleimidopropyl)aminopropane was added 0.25 mL of dichloromethane and the reaction was gently mixed until a clear solution was formed. To this solution was added 0.25 mL of trifluoroacetic acid and was gently mixed at ambient temperature for 75 min. Subsequently, the solution was evaporated to dryness. The residue was dissolved in 3 mL of ethanol and the resulting solution added to the above hydrogel suspension. The vial containing the maleimide reagent was rinsed with ethanol (2×1 mL) and the washings were added to the reaction mixture. The pH of the reaction mixture was then adjusted to 6.41 using 10% v/v N-methylmorpholine in ethanol and the resulting reaction mixture was shaken gently for 18 hours at ambient temperature. At the end of this time, the pH of the reaction mixture adjusted to pH 3.89 by treating with 1.0 M HCl. The gel was precipitated by adding ethanol (4×3.5 mL). The suspension was centrifuged at 120G for 2 min at 20° C. and the supernatant was carefully removed using a pipette. The residue was rehydrated in 12 mL of 20.0 mM SBS buffer (pH 3.8) by gentle shaking/mixing for 15 minutes. The suspension was subjected to additional ethanol treatment (4×5 ml, 1×4 mL). After each ethanol treatment, the suspension was centrifuged at 120G for 2 min at 20° C. followed by careful removal of the supernatant. Finally, the vial was inverted, allowed to drain for 15 minutes and the supernatant was decanted yielding 0.8876 g of the wet gel.


Example 16c
Synthesis of 3-(3-maleimido-propyl) aminopropane Functionalized Hyaluronic Acid

To 10 g of divinyl sulfone crosslinked suspension (Example 11a) was added 40 ml of deionized (DI) water and the resulting suspension was stirred at 25° C. for 10 minutes. After adding 30 ml of ethanol to the suspension, it was stirred for additional 60 minutes. It was followed by addition of 60 mL of ethanol and 10 minute of stirring. To this suspension was added 0.162 g of DMT-MM dissolved in 7.5 mL of ethanol. The resulting reaction mixture was stirred at 25° C. for 60 mins. To 29 mg of 1-(tert-Butoxycarbonyl) amino 3-(3-maleimidopropyl) aminopropane (example 2) dissolved in 0.5 mL of anhydrous dichloromethane was added 0.5 mL of trifluoroacetic acid. The resulting reaction was stirred at room temperature for 2h. The solution was concentrated under reduced pressure. The resulting residue was treated with 2 mL of methanol and evaporated to dryness. After repeating this process one more time, the residue was dissolved in ethanol (7.5 mL) and 9.5 μL of N-methylmorpholine. The solution was added the above hyaluronic acid suspension and pH adjusted to 7.1 with using N-methylmorpholine. The vial was rinsed with ethanol (5 mL) and added to the slurry. After stirring for 21 h at 25° C., saturated brine (2 mL) was added and the gel was precipitated by adding ethanol (4×15 mL). The slurry was allowed to settle for 15 min and the supernatant was decanted. The hydrogel was hydrated in 20 mM PBS pH5 (35 mL) and was precipitated by adding ethanol (5×15 mL). The process was repeated, the sample centrifuged at 2500 rpm for 1.5 min, and the supernatant decanted. Glucosamine assay method suggests the degree of substitution to be 24 mole %.


Example 17
Synthesis of 8-(3-maleimidopropyl) amino-3,6-dioxaoctane Functionalized Hyaluronic Acid

In a 250 mL round bottomed flask were taken 0.5 g of lyophilized HA (example 11a) and 50 mL of 0.9% saline solution. The reaction mixture was allowed to stir gently at 25° C. for one hr. To this suspension was added 40 mL of ethanol and the suspension was stirred for 5 min. In a 10 mL vial were taken 0.35 g of DMT-MM and 5 mL of ethanol. The solution was added to the HA suspension. The vial was rinsed with 5 mL of ethanol and added to the suspension. The resulting reaction mixture was stirred at 25° C. for 1 hr. Subsequently, 86 mg of 1-(tert-Butyloxycarbonyl) amino 8-(3-maleimidopropyl) amino-3,6-dioxaoctane dissolved in 0.5 mL of anhydrous dichloromethane was treated with 0.5 mL of trifluoroacetic acid was added to the. The solution was stirred at room temperature from 1 h. The solution was concentrated under reduced pressure, dissolved in 2 mL of ethanol and evaporated to dryness under reduced pressure. The ethanol treatment was repeated twice. The residue was dissolved in 5 mL of ethanol/water (1:1) and the pH of the solution was adjusted to 6.5 using N-methylmorpholine (60 μL). The resulting solution was added to the above suspension. After stirring for 4h at 25° C., pH of suspension was adjusted to 3.75 with 0.1 M HCl and the gel was precipitated by adding ethanol (7×25 mL). The slurry was allowed to settle for 30 min and 80% of the supernatant removed decantation. The remaining suspension was centrifuged at 1500 rpm for 5 min. After removing the solvent, the residue was treated with 0.9% saline and precipitated from ethanol (7×15 mL). The residue was dried by lyophilization. The degree substitution, as determined by glucosamine assay was found to be 7.3 mol %.


Example 18
Synthesis of 2-pyridyldithiol Group Containing Hyaluronic Acid

To a rapidly stirring solution of 200 mg sodium hyaluronate (molecular weight=500 kDa) dissolved in 24 mL of deionized water was added 16 mL acetonitrile in dropwise manner. After addition of acetonitrile was complete, 17.6 mg of chlorodimethoxytriazine in 2 mL of water/acetonitrile (1:1) to the reaction mixture followed by 20 μL of N-methylmorpholine. The reaction was stirred at 25° C. for 1 hr. Subsequently, 26.8 mg of 2-((3-nitropyridin-2-yl)thio)ethanamine hydrochloride dissolved in 1 mL of deionized water was added. The reaction was allowed to stir for 18 hr. The pH of the reaction was adjusted to 6.0 by adding 1M HCl. To the resulting reaction mixture was added 15 mL of pre-washed Amberlite® CG-120 (Na+ form) and the reaction mixture was stirred for 20 minutes. The resin was filtered off and was washed with deionized water (2×5 mL). The Amberlite® CG-120 (Na+ form) treatment process was repeated twice. The solution was diluted with water to form a solution containing 20 vol % of acetonitrile. The solution was spin-filtered using Macrosep® centrifugal devices (30K molecular weight cutoff). The retentate was washed with deionized water (5×200 ml). The retentates were combined and lyophilized yielding 120 mg of the product as an off white solid. The degree of modification was 10%. Another 2-pyridyldithiol group containing hyaluronic acid was synthesized using sodium hyaluronate of molecular weight 70 kDa following the similar procedure.


Example 19
Synthesis of Soluble Maleimide Functionalized HA

In a 100 mL flask were taken 200 mg of hyaluronic acid, sodium salt (mol. wt.=500 kDa) and 24 ml of DI water. It was stirred until a clear solution was obtained. To the rapidly stirred HA solution was added 16 mL of ethanol was added in a. 4-(4,6-Dimethoxy-1,3,5-triazin-2-yl)-4-methylmorpholinium chloride (55 mg, 0.2 mmol) in water/ethanol was added along with N-methylmorpholine (20 μl, 0.2 mmol) and the reaction was aged for one hour. An aqueous solution of 1-(2-(2-aminoethoxy)ethyl)-1H-pyrrole-2,5-dione.trifluoroacetate salt (60 mg, 0.2 mmol) was added. The reaction was left over night. The pH is adjusted to be slightly acidic and pre-washed Amberlite® CG-120, Na+ form (≥15 ml) was added to the reaction mixture and stirred for 20 minutes. The resin was filtered off and washed with water. To the filtrate was again added pre-washed Amberlite® CG-120, Na+ form (≥15 ml) and the same procedure as above was followed. The whole cycle was repeated once more. The solution was diluted with water to form an aqueous solution containing <20% ethanol. The solution was spin-filtered using four Macrosep® centrifugal devices from PALL, 30K molecular weight cutoff (5000 rpm, 15 minutes spin time. The membrane is cleared each time by gently stroking a spatula over it and vigorously shaking the sealed devise). The retentate was washed several times with deionized water (>200 ml). The resulting modified HA concentrates were combined and lyophilized. Recovery is between 50-75%. Degree of modification 20% according to NMR.


Example 20
Estimation of Maleimide Content in Hyaluronic Acid Hydrogels

Estimation of maleimide groups incorporated to HA hydrogels was performed by a colorimetric analysis method. 5-Thio 2-nitrobenzoic acid was prepared by the reduction of 5,5′-dithiobis-(2-nitrobenzoic acid) with Tris-(2-carboxyethyl) phosphine hydrochloride (TCEP) in PBS buffer at pH 7.5. A 20 mol % excess of 5,5′-dithiobis-(2-nitrobenzoic acid) was used to prevent side reactions with TCEP. A predetermined amount of maleimide functionalized hydrogel suspended in 20 mM Succinate buffered saline (SBS) at pH 3.5. Above 5-Thio 2-nitrobenzoic acid solution was added to the hydrogel suspension and the reaction mixture was vortex mixed (2×10 seconds) and was subsequently stirred gently at 25° C. for 45 min. The suspension was subsequently centrifuged at 25° C. for 10 min and an aliquot of the supernatant taken. The absorbance of the supernatant was measured at 412 nm. The concentration 5-thio 2-nitrobenzoic acid in the solution was estimated using a calibration curve. Maleimide concentration in the hydrogels is equivalent to the moles of thiol reacted, which is calculated from the difference between the amount 5-thio 2-nitrobenzoic acid added and that present in the supernatant.


Example 21
Synthesis of Near Infrared Dye (IRDye800CW) Conjugated HA Hydrogel



embedded image


Example 21 a. Synthesis 3-nitro-2-(2′-amino-ethyldisulfanyl)pyridine Hydrochloride (NEA) Conjugated Divinyl Sulfone Cross-Linked Hyaluronic Acid

A suspension containing 5.1 g (23 mg/g of water) of divinylsulfone cross-linked hyaluronic acid hydrogel (example 5) and 20 mL of sterile water was gentle stirred at 25° C. for 15 min. To this suspension was added ethanol (20 mL) and the resulting reaction mixture was stirred 25° C. for one hr. To this suspension was added 0.081 g of DMT-MM dissolved in 5 mL of ethanol. The vial containing DMT-MM solution was rinsed with 2.5 mL of ethanol and the washing was added to the suspension. The reaction mixture was stirred at 25° C. for 1 hr. followed by addition of 0.017 g of NEA. After stirring the reaction mixture for 22h at 25° C., 2 mL of brine was added the gel. The solvent was removed by centrifugation was the residue was washed by multiple ethanol treatment (4×10 mL, 1×5 mL). The suspension was centrifuged at 2500 rpm for 5 min followed by at 5000 rpm for 5 min (the temperature of the centrifuge was maintained at 5° C.). The supernatant was decanted and residue was allowed equilibrate for 20 min in sterile water (20 mL). Subsequently, it was subjected to two ethanol treatment (4×10 mL). The slurry was allowed to settle for 30 min and the supernatant decanted. The ethanol treatment process was repeated one more time and the wet gel was stored at 2° C. until next step.


Example 21b. Synthesis of NIR Dye Conjugated HA Hydrogel

Under aseptic condition, 0.224 g NEA modified hyaluronic acid (example 15a) and 13 mL of sterile water were taken in a 50 mL sterile reaction vial and the mixture was allowed to shake gently for 15 min at 25° C. To this suspension was added 20 mL of ethanol and the suspension was stirred for 1 hr. at 25° C. Subsequently, 14 mg of TCEP was added to the suspension and the reaction mixture was stirred for 16 hr. at 25° C. At the end of the reaction, 2 mL of brine was added to the reaction mixture. Subsequently, the gel was subjected to 5 cycles of ethanol treatment (5 mL) and centrifugation (5000 rpm, 5 min., 5° C.). After removal of the supernatant, the residue was treated with 10 mL of in 0.9% sterile saline followed 5 round of ethanol (5 mL) treatment and centrifugation as mentioned above. The residue was suspended in 10 mL of sterile 0.9% saline. To this suspension was added 1.8 mg of maleimide functionalized IRDye800CW dissolved in sterile 0.9% saline (3 mL) using a sterile dehydrogenating filter. The filter was rinsed with 5 mL of ethanol and ethanol/saline 1:1 (2×5 mL) and the washing were added to the reaction mixture. The reaction vessel was agitated gently in dark for 2 hr. at 25° C. and was kept at 2° C. for 66h. Subsequently 11.5 mg of N-methylmaleimide dissolved in 3 mL of ethanol was added to the reaction mixture and the suspension it was shaken gently for additional 3 hr at 25° C. At the end of the reaction, the suspension was centrifuged (5000 rpm, 5° C., 2×15 min) and 20 mL of supernatant was removed. The remaining suspension was precipitated from ethanol (5×5 mL) and further centrifuged (5000 rpm, 5° C., 5 min) and the supernatant was decanted. 10 mL of 0.9% sterile saline was added to the residue, followed by 5 mL of ethanol. The suspension was centrifuged (5000 rpm, 5 min, 5° C.) and supernatant was decanted. The ethanol treatment and centrifugation process was repeated four more time. The residue was treated with 10 mL of sterile 0.9% saline and stored at 2° C. under dark by wrapping the vial with an aluminum foil.


Conjugation of Linker Thio Peptides to Maleimide Functionalized HA Hydrogel
Example 22

General procedure for the thiol terminated trace-linker bearing peptides to maleimide functionalized HA hydrogels.


In a sterile and depyrogenated reactor with medium porosity frit or filter was taken appropriate amount of the maleimide modified HA hydrogel (example 3). Subsequently, appropriate amount of sterile filtered 20 mMol SB buffer (containing 15% v/v propylene glycol and 0.01% w/v Tween 20, pH 3.8) was added to the reaction such that the concentration of the resulting suspension is ˜1% w/w. The suspension was allowed to mix for 30-90 minutes with gentle shaking. At the end of this time, appropriate amount of thiol terminated trace-linker bearing peptide dissolved in sterile filtered 20 mMol SB buffer (containing 15% v/v propylene glycol and 0.01% w/v Tween 20, pH 3.8) was added to the reactor and the resulting reaction mixture was allowed to shake gently at ambient temperature for 1.5-24 hours. At the end of the reaction, the supernatant was removed by filtration using a slight excess pressure of nitrogen or by centrifugation of the suspension. The residue was treated with sterile filtered 20 mM SBS buffer (containing 15% v/v propylene glycol and 0.01% w/v Tween 20, pH 3.0) to prepare a suspension 0.7 w/v %, shaken for 3 minutes, centrifuged and the supernatant was removed by decantation. This process was repeated five times. The residue was treated with 10 mM solution of 1-Hydroxy-2-mercaptoethane dissolved in sterile filtered 20 mM SBS buffer (containing 15% v/v propylene glycol and 0.01% w/v Tween 20, pH 3.0) to prepare a ˜1 wt % suspension and was allowed to stir gently for 30 minutes with gentle shaking/mixing. The solvent was removed by centrifugation followed by decantation as mentioned above. This process of 1-Hydroxy-2-mercaptoethane treatment was repeated four times. The residue was suspended in sterile filtered 20 mM SBS buffer (containing 15% v/v propylene glycol and 0.01% w/v Tween 20, pH 3.0) to prepare a suspension of ˜0.5 wt % concentration and mixed for 3 minutes followed by removal of the by centrifugation and decantation. The resulting residue was suspended in 20 mM SBS buffer (containing 15% v/v propylene glycol and 0.01% w/v Tween 20, pH 6.5) to prepare a 0.7 wt % suspension and stirred for 20 minutes and filtered. After repeating this process one more time, the residue was in 20 mM SBS buffer (containing 15% v/v propylene glycol and 0.01% w/v Tween 20, pH 4.5) to prepare a 0.5 wt. % suspension, stirred for 15 minutes, and filtered. This process was repeated once. The residue was suspended in sterile water (pH 4.5), stirred for 5 minutes, and filtered. The process was repeated five times and. The residue was aseptically filtered using a sterile membrane filter and lyophilized to dryness.


Table 3 summarizes results on the syntheses of various HA hydrogel conjugates of dual agonist peptides obtained by varying reaction conditions and nature of traceless linker.


















Amount of


Amount





maleimide


of
Yield of


modified
Maleimide

Peptide
hydrogel-


HA
content in

linker
peptide
Peptide
Peptide


hydrogel
hydrogel
Linker
used
conjugate
loading
loading


(mg)
(mol %)
type
(mg)
(mg)
(wt. %)
(mol %)





















36
3.4
Asn
2.1
20
4
0.41


35
4.9
Asn
2.4
31
2.4
0.24


297
10.8
Asn
30.6
228
4
0.41


57.5
3.8
Asn
15
66.3
11.1
1.2


54
3.8
Asn
23
77.8
15.9
2.18


48
5.7
Asn
30
60.8
21.0
2.6


73.2
5.2
Asn
15.8
51.6
11
1.2


137
N.D.
Asn
62.5
150
21.7
2.69


90
9.8
Aib
13.3
78
6.5
0.69


50
8.8
D-Ala
3.7
49.3
N.D.
N.D.


41
6.2
D-Ala
9.2
52
N.D.
N.D.
















TABLE 3b







The following conjugates were prepared as described in examples 14 to 22













Amount of

Maleimide


Yield of
Peptide


crosslinked

loading in

Amount of
lyophilized
content


HA
HA
hydrogel after

Peptide
hydrogel-
by


hydrogel
content
activation
Linker
linker used
peptide
NMR


(g)
in gel (%)
(mol %)
type
(mg)
conjugate (g)
(mol %)
















200
2.3
12.1
Aib
508
6.4
N.D.


40
2.3
14.8
Aib
102
1.2
N.D.


40
2.3
9.6
Aib
108
0.84
N.D.


40
2.3
13.6
Aib
203
0.89
6.5


40
2.3
11.0
Aib
102
0.82
3.6


40
2.3
12.5
Aib
104
0.71
5.9


40
2.3
16.8
Aib
108
0.49
9.0


40
2.3
8.0
Aib
102
1.2
N.D.


40
2.3
13.5
Aib
102
1.1
4.4


30
2.6
22
Aib
127
0.67
5.3


30
2.6
18.2
Aib
201
0.45
6.5









Conjugation of linker 7 containing thiol functionalized Exendin-4 Seq ID No. 26 with maleimide functionalized hyaluronic acid.




embedded image


To 53 mg of 3-(3-maleimidopropyl) aminopropane modified DVS-HA hydrogel (example 15) was added 7 mL of the 20 mM SBS containing, 0.01 wt % Tween® 20, propylene glycol (15% v/v). The pH of buffer is 3.8. The suspension was gently agitated at 25° C. for 90 min. To this suspension was added 2.1 mg of thiol functionalized Exendin-4 Seq ID 26 with the linker of example 7 dissolved in 1 ml of the above buffer. The peptide vial was rinsed with buffer (2×0.5 mL) and the washing was added to suspension. The reaction mixture was gently agitated at 25° C. for 18h. The suspension was subsequently centrifuged at 1750 rpm for 2 min and the supernatant was carefully decanted. The hydrogel was suspended in 10 mL of 20 mM SBS buffer containing 0.01 wt % Tween® 20, propylene glycol 15% v/v, pH 3, stirred for 2 min, centrifuged at 1750 rpm for 2 min, and the solvent decanted. This process was repeated four more times. The residue was treated with 2 mL of 10 mM 1-Hydroxy-2-mercaptoethane dissolved in 20 mM SBS pH 3 and the reaction was allowed to proceed for 30 min. The reaction mixture was centrifuged at 2000 rpm for 2 min and the solvent decanted. This process was repeated three more times. The residue was suspended in 10 mL of 20 mM SBS containing 0.01 wt % Tween® 20, pH 3, stirred for 2 min, centrifuged at 2000 rpm for 2 min and the solvent decanted. The residue was subsequently suspended in 10 mL of acidified sterile water (pH 3.5) containing 0.01 wt % Tween® 20, stirred for 10 min, centrifuged at 5000 rpm for 5 min and the solvent was decanted. This process was repeated with 8, 7 and 6 mL of the above acidified sterile water. The sample was lyophilized yielding 78 mg of the HA hydrogel conjugated peptide as an off white solid with 4 wt. % loading of peptide in the hydrogel.


Example 23

Conjugation of linker containing thiol functionalized Exendin-4 Seq ID 26 with maleimide functionalized hyaluronic acid.


Reaction and work up were performed using de-oxygenated buffers and under nitrogen atmosphere. To 36 mg of 3-(3-maleimidopropyl) aminopropane modified DVS-HA hydrogel (example 15) was added 6 mL of 20 mM SBS, (containing 0.01 wt % Tween® 20, 10% v/v propylene glycol). The pH of the medium was adjusted 6. The suspension was gently shaken at 25° C. for 30 min. To this suspension was added 2.4 mg of thiol functionalized Exendin-4 Seq ID 26 with linker (example 7) dissolved in 1 ml of the above buffer (pH 6). The peptide vial was rinsed with buffer (2×0.5 mL) and the washing was added to suspension. The reaction mixture was gently shaken at 25° C. for 90 min. The suspension was subsequently centrifuged at 4000 rpm for 2 min and the supernatant was carefully decanted. The hydrogel was suspended in 10 ml of 20 mM SBS at pH 3 (containing 0.01 wt % Tween® 20, and 10 v/v % propylene glycol), mixed for 2 min, centrifuged at 3005G for 2 min, and the solvent decanted. This process was repeated four more times. The residue was treated with 2 mL of 10 mM 1-hydroxy-2-mercaptoethane in 20 mM SBS pH3 for 30 min, centrifuged at 4000 rpm for 2 min and the solvent decanted. This process was repeated three more times. The residue was suspended in 10 mL of 20 mM SBS containing 0.01 wt % Tween® 20 (pH 3), mixed for 2 min, centrifuged at 4000 rpm for 2 min, and the solvent was decanted. This process was repeated four more times. The residue was suspended in acidified (pH 3.5) sterile water containing 0.01 wt % Tween® 20, mixed for 10 min, centrifuged at 5000 rpm for 5 min, and the solvent decanted. This process was repeated using 8, 7 and 6 mL of the above sterile water. The residue was lyophilized to yield 31 mg HA conjugated peptide as an off white solid with 2.4 wt % peptide loading in the hydrogel.


Example 24
Procedure for Estimation of Peptide Loading in the Hydrogel-Peptide Conjugates

A predetermined amount HA hydrogel-peptide conjugate was suspended in CHES buffer (pH9.5) and the suspension was allowed to gently stir at 70° C. The suspension was centrifuged and the aliquot was analyzed for peptide content by HPLC method. The HPLC method comprises of using q C-18 Kinetics column (inner diameter=4.6 mm and length=100 mm, particle size 2.6 μm, Phenomenex) using Agilent 1100 LC. The composition of mobile phase A is 90% water/10% Acetonitrile/0.1% Trifluoroacetic acid (TFA) and the mobile phase B is 10% Acetonitrile/90% water/0.09% TFA. The gradient is from mobile phase 25% B to 55% B in 8 minutes. The flow rate was kept at 1 mL/min. Pure peptides were used as standards to quantify the released peptide from the hydrogel.


Example 25
Determination of In Vivo Residence Time of the HA Hydrogel

The residence time of hydrogels in vivo in the subcutaneous space were investigated by magnetic resonance (MR) imaging. The intensity of water proton contrast inside the hydrogel was used to assess the hydrogels residence time in vivo. For this purpose, CAnN.Cg-Foxn1 nu/Crl mice were used as the animals. The hydrogel was injected using a 31 G needle by following all approved animal care protocols. The MR image of the injection site was taken regularly over a period of time. In one group, the hydrogel was injected and in the other group, a suspension of containing 1:1 (w/w) mixture hydrogel and 800,000 Da soluble HA was used. In the case of pure hydrogel, the gel was evident for 3 weeks with slow loss in intensity (FIG. 2). On the other hand, for the mixture containing soluble HA, the MR signal, which was intense on day 1, has significantly reduced on day 4 (2). This suggests that to improve the residence time of the polymer carrier, it needs to be crosslinked to achieve very high molecular weight.



FIG. 2a. MR of Image of the HA hydrogel at the injection site as a function of time.



FIG. 2b. MR of Image of the polymer suspension containing 1:1 (w/w) HA hydrogel-800 kDa soluble HA at the injection site as a function of time.


Example 26
Release Kinetics In Vitro

An aliquot of GLP-1/Glucagon agonist linker hydrogel 8 (0.5 mg GLP-1/Glucagon agonist) was transferred into a syringe equipped with a filter frit and washed 5 times with pH 7.4 phosphate buffer (60 mM, 3 mM EDTA, 0.01% Tween-20). The hydrogel was suspended in the same buffer and incubated at 37° C. At defined time points (after 1-7 days incubation time each) the supernatant was exchanged and liberated GLP-1/Glucagon agonist was quantified by RP-HPLC at 215 nm. UV-signals correlating to liberated GLP-1/Glucagon agonist were integrated and plotted against incubation time. Curve-fitting software was applied to estimate the corresponding halftime of release.



FIG. 3. In vitro release kinetics of Exendin-4 Seq ID 26 dual agonist with linker from the HA hydrogel (example 23). The half-life is ˜5 days.


Example 27
In Vitro Cellular Assays for GLP-1 Receptor, Glucagon Receptor and GIP Receptor Efficacy

Agonism of peptides for the receptors was determined by functional assays measuring cAMP response of HEK-293 cell lines stably expressing human GIP, GLP-1 or glucagon receptor.


cAMP content of cells was determined using a kit from Cisbio Corp. (cat. no. 62AM4PEC) based on HTRF (Homogenous Time Resolved Fluorescence). For preparation, cells were split into T175 culture flasks and grown overnight to near confluency in medium (DMEM/10% FBS). Medium was then removed and cells washed with PBS lacking calcium and magnesium, followed by proteinase treatment with accutase (Sigma-Aldrich cat. no. A6964). Detached cells were washed and resuspended in assay buffer (1×HBSS; 20 mM HEPES, 0.1% BSA, 2 mM IBMX) and cellular density determined. They were then diluted to 400000 cells/ml and 25 μl-aliquots dispensed into the wells of 96-well plates. For measurement, 25 μl of test compound in assay buffer was added to the wells, followed by incubation for 30 minutes at room temperature. After addition of HTRF reagents diluted in lysis buffer (kit components), the plates were incubated for 1 hr, followed by measurement of the fluorescence ratio at 665/620 nm. In vitro potency of agonists was quantified by determining the concentrations that caused 50% activation of maximal response (EC50).


Example 28
Glucose Lowering in Female Diabetic Dbdb-Mice

Female diabetic dbdb-mice (BKS.CG-m+/+Lepr(db)/J) 24-27 weeks of age at study start were used. Mice arrived in the age of 10-13 weeks were habituated to feeding and housing conditions for at least 1 week, then used in a first study and after a washout period of at least 2 weeks finally reused for the present study. 19 days prior to study start, individual HbA1c values were determined to stratify and thereafter allocate the animals into 4 groups with an N=8 per group to provide groups with as equally as possible mean HbA1c values. Animals had access to food and water ad libitum throughout the entire study period. On the first day of the study blood glucose from tail tip incision was determined just before and 4 hours after single subcutaneous treatment (08:00-09:00 am) with either vehicle (sterile succinate buffer) or the HA-conjugate of GLP-1/Glucagon agonist Seq. ID 26 of example 23 in the doses 50, 100 and 200 nmol/kg diluted in vehicle. Thereafter daily blood glucose measurements were performed for the next 16 days all at a similar day time (08:00-09:00 am) with the exception of days 11 and 12 (weekend). In addition food intake and water consumption was monitored on a daily basis. Glucose data were analyzed by two-way-ANOVA on repeated measurements, followed by Dunnett's post-hoc test with a significance level of p<0.05. Glucose AUC analysis was done using a one-way-ANOVA followed by Dunnett's post-hoc test with a significance level of p<0.05.



FIG. 4 shows the blood glucose concentration relative to the baseline versus time after one injection at various doses of HA-GLP-1/Glucagon agonist conjugate with Seq. ID 26.


Example 28b Glucose Lowering Effects in Female Diabetic dbdb-Mice

Female, obese diabetic db/db-mice (BKS.CG-m+/+Lepr(db)/J and healthy, lean controls (BKS.Cg-m+/+Lepr(db)/J) arrived at the age of 10-11 weeks and were habituated to feeding and vivarium conditions. At the age of 12-13 weeks individual HbA1c values were determined to stratify and allocate animals into different groups with an N=8 per group. Goal wes to provide groups with as equally as possible mean HbA1c values. At the age of 13-14 weeks 2 groups of db/db mice received a s.c. 100 nmol/kg body mass single dose of a conjugate with Seq. ID 26 of either soluble or crosslinked HA. At the same time and a second time on day 7 a third group of db/db mice and the healthy lean references received a s.c. injection of succinate buffer and soluble HA in a 1:1 ratio. In all groups a total volume of 5 ml/kg body mass was injected. Morning-fed blood glucose concentrations were determined just before (between 08:00-09:00 am) and four hours after treatment and thereafter daily between 08:00-09:00 am. Blood was collected from tail tip incisions and concentrations determined via a handheld glucometer (Accu Check). Throughout the entire study period animals had access to food and water ad libitum.



FIG. 4b shows the blood glucose concentration relative to the baseline versus time after one injection of cross-linked and soluble HA-GLP-1/Glucagon agonist conjugate with Seq. ID 26.


Example 28c
Glucose Lowering Effects in Female Diabetic Db/Db-Mice

After arrival female, obese diabetic db/db-mice (BKS.CG-m+/+Lepr(db)/J) were habituated to feeding and vivarium conditions and were 12 weeks old at study start. Individual HbA1c values were determined on day 10 of the predose phase to stratify and allocate animals into different groups with an N=8 per group. Goal was to provide groups with as equally as possible mean HbA1c values. On day 1 of the dosing phase db/db mice received a s.c., single dose of 12.75 mg/kg body mass of either conjugate with Seq ID 45, 46, 48 and 49. Db/db animals of the Vehicle group received a s.c. PBS injection. In all groups a total volume of 10 ml/kg body mass was injected. Morning-fed blood glucose concentrations were determined just before (between 08:00-09:00 am) and four hours after treatment on day 1 of the dosing phase and thereafter daily between 08:00-09:00 am. Blood was collected from tail tip incisions and concentrations determined via a handheld glucometer (Accu Check). Throughout the entire study period animals had access to food and water ad libitum.



FIG. 4c shows the blood glucose concentration relative to the baseline versus time after one injection at various doses of HA-GLP-1/Glucagon agonist conjugate with Seq. ID 45 (triangles), 46 (squares), 48 (circles).



FIG. 4d shows the blood glucose concentration relative to the baseline versus time after one injection at various doses of HA-GLP-1/Glucagon agonist conjugate with Seq. ID 49 (circles).


Example 29
Injectability Study

A suspension of 3.75% (mg/mL) HA-peptide conjugate was prepared by dispersing appropriate amount of the peptide conjugate in 20 mM succinate buffered saline (SBS) at pH 4.5. In another vial, a 2% (mg/mL) solution of native HA polymer was prepared by dissolving lyophilized HA in 20 mM SBS at pH 4.5. Both the samples were allowed to hydrate for 24 to 48 hours at 2-8° C. The vials were brought to room temperature. After equilibration at room temperature, 1 mL of the peptide-HA hydrogel conjugate suspension and HA was taken in a 3 mL syringe. To this suspension was added 0.25 mL of the soluble HA solution. The resulting suspension was subjected to 20 times of back and forth mixing between two 3 mL syringes. Approximately 220 μl of this suspension was to a 1 ml syringe. To this syringe was attached a 30 g, ½ inch needle. It should be ensured that the needle is not primed with test material. The syringe containing the suspension was loaded into the syringe fixture of the Instron equipment and the crosshead of the Instron was aligned with the plunger of the syringe. The speed of the crosshead was adjusted to achieve target injection rate. The testing was initiated. For each sample, three measurements were performed and result presented is an average of these measurements. The results on injectability force for different peptide-HA hydrogel conjugates are shown in Table 4.









TABLE 4







Effect of HA Modification on Average Injectability Force*










Peptide content in
Average



the conjugate
Injectability Force










Compound type
(wt. %)
(mol %)
(N)













HA Conjugated Seq. ID No.
11.0
1.2
19.1 ± 0.6


26, Asn linker


HA Conjugated Seq. ID No.
15.9
2.18
16.8 ± 1.8


26, Asn linker


HA Conjugated Seq. ID No.
21.7
2.69
13.7 ± 1.2


26, Asn linker





*n = 3 per group; injection rate = 12 μl/s







FIG. 5 shows the average extrusion force by pressing the liquid through a 2.5 cm long 30G needle attached to an 1 mL syringe. It is clearly seen that a higher peptide loading on the HA hydrogel leads to lower extrusion forces and therefore enhances injectability.


Example 29b
Injectability Study of HA-Conjugate with Seq. ID No. 45, Aib-Linker

A 1 mL Luer-Lock syringe (BD syringe, inner diameter 4 mm) was filled with approximately 500 μL (460 μL on the syringe scale) of the sample solution and a 29G×12.7 mm needle was attached. The measurement was carried out at the LF Plus dynamometer from Lloyed Instruments. The plunger rod was pushed until a small droplet appeared at the tip of the 29G needle. The syringe was placed in the syringe holder and the dynamometer was moved so that it touches the plunger rod.


The measurement setting was: an abortion force of 50 N and an injection speed of 5.8 mm/s (which equals 100 μL/s in this case).


The measurement was started and was automatically aborted when the plunger rod reached the bottom of the syringe (syringe is empty) or a force higher than 50 N was reached during the measurement.


Table 5 shows the maximum injection forces for different conjugate concentrations with different mixing ratios of soluble HA (sHA).















TABLE 5





Conjugate








[mg/mL]
No sHA
50:50
40:60
30:70
20:80
10:90

















600 kD sHA:conjugate mixing ratio/



sHA 20 mg/mL













20
10.5 N
  10 N
11.7 N
11.9 N
11.2 N
10.3 N


25
13.6 N







30
16.6 N
12.3 N
12.1 N
12.4 N
12.6 N
12.6 N









2600 kD sHA:conjugate mixing ratio/



sHA 20 mg/mL













30

15.5 N
14.5 N
16.1 N
14.2 N
18.5 N









The injection forces of pure 20 mg/mL 600 kDa sHA solution was 13.3 N and of 2600 kDa sHA: 18.8 N.

Claims
  • 1. A prodrug or a pharmaceutically acceptable salt thereof comprising a drug linker conjugate of formula (I) Z-L1-L2-L-Y—R20  (I)wherein Y is a peptide moiety having the formula (II)
  • 2. A prodrug or a pharmaceutically acceptable salt thereof comprising a drug linker conjugate of formula (I) as claimed in claim 1Z-L1-L2-Y—R20  (I)wherein Y is a peptide moiety having the formula (II)
  • 3. The prodrug of claim 1, wherein L is a linker moiety of formula (Ib),
  • 4. The prodrug of claim 1, wherein L is a linker moiety -L of formula (Ic),
  • 5. The prodrug of claim 1, wherein L2 is a C1-6 alkyl chain, which is optionally interrupted by one group selected from —O— and C(O)N(R3aa) and, wherein R3aa is independently selected from the group consisting of H and C1-4 alkyl; andL2 is attached to L1 via a terminal group selected from the group consisting of
  • 6. The prodrug of claim 1, wherein Y is a peptide moiety having the formula (II)
  • 7. The prodrug of claim 1, wherein Y is a peptide moiety having the formula (IVa)
  • 8. The prodrug of claim 1, wherein Y is a peptide moiety having the formula (IVb)
  • 9. The prodrug of claim 1, wherein Y—R20 is a GLP-1/Glucagon agonist selected from the group consisting of SEQ ID NOs: 4 to 60.
  • 10. A pharmaceutical composition comprising a prodrug of claim 1 or a pharmaceutical salt thereof together with at least one pharmaceutically acceptable excipient.
  • 11. A pharmaceutical composition comprising a prodrug of claim 1 or a pharmaceutical salt thereof together with at least one pharmaceutically acceptable excipient and a viscosity modifier.
  • 12. A pharmaceutical composition as claimed in claim 11, wherein the viscosity modifier is hyaluronic acid.
  • 13. A pharmaceutical composition as claimed in claim 10 in the form of an injectable formulation.
  • 14. A pharmaceutical composition as claimed in claim 10 in the form of a suspension.
  • 15. A pharmaceutical composition as claimed in claim 10 in the form of a suspension, wherein the prodrug has a concentration of 0.5 to 8 weight/volume percent.
  • 16. A pharmaceutical composition as claimed in claim 10 in the form of a suspension, wherein the prodrug has a concentration of 1.5 to 3 weight/volume percent.
  • 17. A composition according to claim 10, wherein the prodrug is sufficiently dosed in the composition to provide a therapeutically effective amount of GLP1/Glucagon agonist for at least 6 days in one application.
  • 18. A composition according to claim 10, wherein the composition is a single dose composition.
  • 19. A method of treating diseases or disorders which can be treated by GLP-1/Glucagon agonist, comprising administering to a patient in need thereof an effective amount of the prodrug of claim 1.
  • 20. A method of treating diabetes, comprising administering to a patient in need thereof an effective amount of the prodrug of claim 1.
  • 21. A method of treating dyslipdemia, comprising administering to a patient in need thereof an effective amount of the prodrug of claim 1.
  • 22. A method of treating metabolic syndrome, comprising administering to a patient in need thereof an effective amount of the prodrug of claim 1.
  • 23. A method of treating hepatosteatosis, comprising administering to a patient in need thereof an effective amount of the prodrug of claim 1.
  • 24. A GLP-1/Glucagon agonist-linker conjugate intermediate of formula (VIII)
  • 25. An GLP-1/Glucagon agonist-linker conjugate intermediate of formula (IX)
  • 26. An GLP-1/Glucagon agonist-linker conjugate intermediate of formula (X)
  • 27. The composition of claim 18, wherein the prodrug is in the form of a suspension, and wherein the prodrug suspension can be administered by injection through a needle smaller than 0.26 mm inner diameter.
  • 28. A method of treating non-alcoholic liver-disease (NAFLD) or non-alcoholic steatohepatitis (NASH), comprising administering to a patient in need thereof an effective amount of the prodrug of claim 1.
Priority Claims (1)
Number Date Country Kind
15305858 Jun 2015 EP regional
PCT Information
Filing Document Filing Date Country Kind
PCT/EP2016/062496 6/2/2016 WO 00
Publishing Document Publishing Date Country Kind
WO2016/193371 12/8/2016 WO A
US Referenced Citations (652)
Number Name Date Kind
5424286 Eng Jun 1995 A
5641757 Bornstein et al. Jun 1997 A
6284727 Kim et al. Sep 2001 B1
6329336 Bridon et al. Dec 2001 B1
6344180 Holst et al. Feb 2002 B1
6410511 L'Italien et al. Jun 2002 B2
6429197 Coolidge et al. Aug 2002 B1
6451974 Hansen Sep 2002 B1
6458924 Knudsen et al. Oct 2002 B2
6482799 Tuseé et al. Nov 2002 B1
6506724 Hiles et al. Jan 2003 B1
6514500 Bridon et al. Feb 2003 B1
6528486 Larsen et al. Mar 2003 B1
6579851 Goeke et al. Jun 2003 B2
6593295 Bridon et al. Jul 2003 B2
6703359 Young et al. Mar 2004 B1
6706689 Coolidge et al. Mar 2004 B2
6723530 Drucker Apr 2004 B1
6821949 Bridon et al. Nov 2004 B2
6828303 Kim et al. Dec 2004 B2
6849714 Bridon et al. Feb 2005 B1
6858576 Young et al. Feb 2005 B1
6861236 Moll et al. Mar 2005 B2
6872700 Young et al. Mar 2005 B1
6884579 Holst et al. Apr 2005 B2
6887470 Bridon et al. May 2005 B1
6887849 Bridon et al. May 2005 B2
6894024 Coolidge et al. May 2005 B2
6902744 Kolterman et al. Jun 2005 B1
6924264 Prickett et al. Aug 2005 B1
6956026 Beeley et al. Oct 2005 B2
6969702 Bertilsson et al. Nov 2005 B2
6972319 Pan et al. Dec 2005 B1
6982248 Coolidge et al. Jan 2006 B2
6989366 Beeley et al. Jan 2006 B2
6998387 Goke et al. Feb 2006 B1
7056734 Egan et al. Jun 2006 B1
7056887 Coolidge et al. Jun 2006 B2
7105489 Hathaway et al. Sep 2006 B2
7105490 Beeley et al. Sep 2006 B2
7115569 Beeley et al. Oct 2006 B2
7138375 Beeley et al. Nov 2006 B2
7138546 Tang Nov 2006 B2
7141240 Perfetti et al. Nov 2006 B2
7141547 Rosen et al. Nov 2006 B2
7144863 Defelippis et al. Dec 2006 B2
7153825 Young et al. Dec 2006 B2
7157555 Beeley et al. Jan 2007 B1
7179788 Defelippis et al. Feb 2007 B2
7189690 Rosen et al. Mar 2007 B2
7220721 Beeley et al. May 2007 B1
7223725 Beeley et al. May 2007 B1
7256253 Bridon et al. Aug 2007 B2
7259136 Hathaway et al. Aug 2007 B2
7259233 Dodd Aug 2007 B2
7259234 Bachovchin et al. Aug 2007 B2
7265087 Göke et al. Sep 2007 B1
7271149 Glaesner et al. Sep 2007 B2
7297761 Beeley et al. Nov 2007 B2
7312196 L'Italien et al. Dec 2007 B2
7329646 Sun et al. Feb 2008 B2
7399489 Kolterman et al. Jul 2008 B2
7399744 Mack et al. Jul 2008 B2
7407932 Young et al. Aug 2008 B2
7407955 Himmelsbach et al. Aug 2008 B2
7414107 Larsen Aug 2008 B2
7419952 Beeley et al. Sep 2008 B2
7442680 Young et al. Oct 2008 B2
7442682 Kitaura et al. Oct 2008 B2
7452858 Hiles et al. Nov 2008 B2
7456254 Wright et al. Nov 2008 B2
7476652 Brunner-Schwarz et al. Jan 2009 B2
7507714 Pan et al. Mar 2009 B2
7521423 Young et al. Apr 2009 B2
7544657 Ebbehøj et al. Jun 2009 B2
7563871 Wright et al. Jul 2009 B2
7576050 Greig et al. Aug 2009 B2
7585837 Shechter et al. Sep 2009 B2
7592010 Rosen et al. Sep 2009 B2
7595293 Engelund et al. Sep 2009 B2
7595294 Nestor Sep 2009 B2
7608692 Prickett et al. Oct 2009 B2
7612176 Wright et al. Nov 2009 B2
7632806 Juul-Mortensen et al. Dec 2009 B2
7638299 Cho et al. Dec 2009 B2
7682356 Alessi et al. Mar 2010 B2
7683030 Prickett et al. Mar 2010 B2
7691963 Prickett et al. Apr 2010 B2
7696161 Beeley et al. Apr 2010 B2
7700549 Beeley et al. Apr 2010 B2
7704953 Herman et al. Apr 2010 B2
7713930 Brunner-Schwarz et al. May 2010 B2
7723471 Levy et al. May 2010 B2
7741269 Young et al. Jun 2010 B2
7749955 Hansen et al. Jul 2010 B2
7772189 Herman et al. Aug 2010 B2
7790681 Hathaway et al. Sep 2010 B2
7799344 Oberg Sep 2010 B2
7799759 Rosen et al. Sep 2010 B2
7803404 Hokenson et al. Sep 2010 B2
7829664 Tatake et al. Nov 2010 B2
7847079 Rosen et al. Dec 2010 B2
7858740 Beeley et al. Dec 2010 B2
7867972 Ballance et al. Jan 2011 B2
7879028 Alessi et al. Feb 2011 B2
7888314 Hathaway et al. Feb 2011 B2
7897560 Dorwald et al. Mar 2011 B2
7906146 Kolterman et al. Mar 2011 B2
7928065 Young et al. Apr 2011 B2
7928186 Chang Apr 2011 B2
7935786 Larsen May 2011 B2
7939494 Khan et al. May 2011 B2
7960341 Hathaway et al. Jun 2011 B2
7977306 Rosen et al. Jul 2011 B2
7981861 Coolidge et al. Jul 2011 B2
7989585 Dodd et al. Aug 2011 B2
7994121 Bachovchin et al. Aug 2011 B2
7994122 Riber et al. Aug 2011 B2
8008255 Ong et al. Aug 2011 B2
8012464 Rosen et al. Sep 2011 B2
8026210 Young et al. Sep 2011 B2
8030273 Lau et al. Oct 2011 B2
8039432 Bridon et al. Oct 2011 B2
8057822 Prickett et al. Nov 2011 B2
8071539 Rosen et al. Dec 2011 B2
8076288 Levy et al. Dec 2011 B2
8080516 Bridon et al. Dec 2011 B2
8084414 Bridon et al. Dec 2011 B2
8093206 Bridon et al. Jan 2012 B2
8097239 Johnsson et al. Jan 2012 B2
8097586 Lv et al. Jan 2012 B2
8114632 Melarkode et al. Feb 2012 B2
8114833 Pedersen et al. Feb 2012 B2
8114958 Soares et al. Feb 2012 B2
8114959 Juul-Mortensen Feb 2012 B2
8119648 Himmelsbach et al. Feb 2012 B2
8143217 Balkan et al. Mar 2012 B2
8158579 Ballance et al. Apr 2012 B2
8158583 Knudsen et al. Apr 2012 B2
8178495 Chilkoti May 2012 B2
8178541 Himmelsbach et al. May 2012 B2
8197450 Glejbol et al. Jun 2012 B2
8211439 Rosen et al. Jul 2012 B2
8232281 Dugi et al. Jul 2012 B2
8236760 Pimentel et al. Aug 2012 B2
8252739 Rosen et al. Aug 2012 B2
8263545 Levy et al. Sep 2012 B2
8263550 Beeley et al. Sep 2012 B2
8263554 Tatarkiewicz et al. Sep 2012 B2
8268781 Gotthardt et al. Sep 2012 B2
8278272 Greig et al. Oct 2012 B2
8278420 Wang et al. Oct 2012 B2
8288338 Young et al. Oct 2012 B2
8293726 Habib Oct 2012 B2
8293869 Bossard et al. Oct 2012 B2
8293871 Wright et al. Oct 2012 B2
8299024 Rabinovitch et al. Oct 2012 B2
8299025 Alessi et al. Oct 2012 B2
8329419 Nicolaou et al. Dec 2012 B2
8329648 Fineman et al. Dec 2012 B2
8338368 Dimarchi et al. Dec 2012 B2
8343910 Shechter et al. Jan 2013 B2
8372804 Richardson et al. Feb 2013 B2
8377869 Richardson et al. Feb 2013 B2
8389473 Hathaway et al. Mar 2013 B2
8404637 Levy et al. Mar 2013 B2
8410047 Bock et al. Apr 2013 B2
8420604 Hokenson et al. Apr 2013 B2
8424518 Smutney et al. Apr 2013 B2
8426361 Levy et al. Apr 2013 B2
8431685 Wright et al. Apr 2013 B2
8445647 Prickett et al. May 2013 B2
8450270 Dimarchi et al. May 2013 B2
8454971 Day et al. Jun 2013 B2
8461105 Wright et al. Jun 2013 B2
8481490 Tatarkiewicz et al. Jul 2013 B2
8485180 Smutney et al. Jul 2013 B2
8497240 Levy et al. Jul 2013 B2
8499757 Smutney et al. Aug 2013 B2
8546327 Dimarchi et al. Oct 2013 B2
8551946 Dimarchi et al. Oct 2013 B2
8551947 Coolidge et al. Oct 2013 B2
8557769 Coskun et al. Oct 2013 B2
8557771 Fan et al. Oct 2013 B2
8569481 Köster et al. Oct 2013 B2
8575097 Xu et al. Nov 2013 B2
8580919 Bossard et al. Nov 2013 B2
8598120 Soares et al. Dec 2013 B2
8603761 Nicolaou et al. Dec 2013 B2
8603969 Levy et al. Dec 2013 B2
8614181 Juul-Mortensen et al. Dec 2013 B2
8617613 Wright et al. Dec 2013 B2
8636001 Smutney et al. Jan 2014 B2
8641683 Glejbol et al. Feb 2014 B2
8642544 Alfaro-Lopez et al. Feb 2014 B2
8664232 Himmelsbach et al. Mar 2014 B2
8669228 Dimarchi et al. Mar 2014 B2
8673927 Dugi et al. Mar 2014 B2
8697647 Levy et al. Apr 2014 B2
8697838 Dimarchi et al. Apr 2014 B2
8710002 Rothkopf Apr 2014 B2
8710181 Christiansen et al. Apr 2014 B2
8716221 Lv et al. May 2014 B2
8729018 Chilkoti May 2014 B2
8729019 Oberg et al. May 2014 B2
8735350 Shechter et al. May 2014 B2
8748376 Ludvigsen et al. Jun 2014 B2
8759290 James Jun 2014 B2
8759295 Ghosh et al. Jun 2014 B2
8772232 Lau et al. Jul 2014 B2
8778872 Dimarchi et al. Jul 2014 B2
8785396 Leone-Bay et al. Jul 2014 B2
8801700 Alessi et al. Aug 2014 B2
8809499 Fan et al. Aug 2014 B2
8816047 Levetan et al. Aug 2014 B2
8841255 Chilkoti Sep 2014 B2
8853157 Knudsen et al. Oct 2014 B2
8853160 Greig et al. Oct 2014 B2
8877252 Wright et al. Nov 2014 B2
8877709 Shechter et al. Nov 2014 B2
8883449 Kjeldsen et al. Nov 2014 B2
8889619 Bai et al. Nov 2014 B2
8900593 Day et al. Dec 2014 B2
8969288 Dimarchi et al. Mar 2015 B2
8969294 Bianchi et al. Mar 2015 B2
8980830 Dimarchi et al. Mar 2015 B2
8981047 Dimarchi et al. Mar 2015 B2
9018164 Dimarchi et al. Apr 2015 B2
9181305 Haack et al. Nov 2015 B2
9365632 Haack et al. Jun 2016 B2
9457066 Rau et al. Oct 2016 B2
9670261 Haack et al. Jun 2017 B2
9694053 Haack et al. Jul 2017 B2
9745360 Haack et al. Aug 2017 B2
9750788 Kadereit et al. Sep 2017 B2
9751926 Kadereit et al. Sep 2017 B2
9758561 Bossart et al. Sep 2017 B2
9771406 Bossart et al. Sep 2017 B2
9775904 Bossart et al. Oct 2017 B2
9789165 Kadereit et al. Oct 2017 B2
20010011071 Knudsen et al. Aug 2001 A1
20010027180 Isaacs Oct 2001 A1
20010043934 L'Italien et al. Nov 2001 A1
20020061838 Holmquist et al. May 2002 A1
20020137666 Beeley et al. Sep 2002 A1
20020146405 Coolidge et al. Oct 2002 A1
20030036504 Kolterman et al. Feb 2003 A1
20030050237 Kim et al. Mar 2003 A1
20030069182 Rinella et al. Apr 2003 A1
20030087820 Young et al. May 2003 A1
20030087821 Beeley et al. May 2003 A1
20030092606 L'Italien et al. May 2003 A1
20030119021 Koster et al. Jun 2003 A1
20030119734 Flink et al. Jun 2003 A1
20030180287 Gombotz et al. Sep 2003 A1
20030216287 Tang Nov 2003 A1
20030220255 Knudsen et al. Nov 2003 A1
20040023871 Hiles et al. Feb 2004 A1
20040029784 Hathaway Feb 2004 A1
20040037826 Michelsen et al. Feb 2004 A1
20040038865 Gelber et al. Feb 2004 A1
20040048783 Brunner-Schwarz et al. Mar 2004 A1
20040097510 Himmelsbach et al. May 2004 A1
20040209255 Koster et al. Oct 2004 A1
20040209803 Baron et al. Oct 2004 A1
20040242853 Greig et al. Dec 2004 A1
20040266670 Hiles et al. Dec 2004 A9
20040266678 Beeley et al. Dec 2004 A1
20040266683 Hathaway et al. Dec 2004 A1
20040266692 Young et al. Dec 2004 A1
20050009742 Bertilsson et al. Jan 2005 A1
20050009847 Bertilsson et al. Jan 2005 A1
20050009988 Harris et al. Jan 2005 A1
20050043238 Young et al. Feb 2005 A1
20050059601 Beeley et al. Mar 2005 A1
20050096276 Coolidge et al. May 2005 A1
20050101537 Beeley et al. May 2005 A1
20050106214 Chen May 2005 A1
20050142152 Leshchiner et al. Jun 2005 A1
20050143303 Quay et al. Jun 2005 A1
20050171019 Young et al. Aug 2005 A1
20050186174 Bossard Aug 2005 A1
20050197287 Mack et al. Sep 2005 A1
20050209142 Bertilsson et al. Sep 2005 A1
20050215469 Beeley et al. Sep 2005 A1
20050215475 Ong et al. Sep 2005 A1
20050267034 Prickett et al. Dec 2005 A1
20050271702 Wright et al. Dec 2005 A1
20050281879 Chen et al. Dec 2005 A1
20060003918 Kim et al. Jan 2006 A1
20060057137 Steiness Mar 2006 A1
20060069029 Kolterman et al. Mar 2006 A1
20060073182 Wong et al. Apr 2006 A1
20060074012 Hiles et al. Apr 2006 A1
20060079448 Bertilsson et al. Apr 2006 A1
20060084605 Engelund et al. Apr 2006 A1
20060094652 Levy et al. May 2006 A1
20060094653 Levy et al. May 2006 A1
20060110423 Wright et al. May 2006 A1
20060135586 Kozlowski et al. Jun 2006 A1
20060135747 Levy et al. Jun 2006 A1
20060148713 Beeley et al. Jul 2006 A1
20060165733 Betz et al. Jul 2006 A1
20060171920 Shechter et al. Aug 2006 A1
20060172001 Ong et al. Aug 2006 A1
20060178304 Juul-Mortensen et al. Aug 2006 A1
20060183677 Young et al. Aug 2006 A1
20060183682 Juul-Mortensen Aug 2006 A1
20060210614 Quay et al. Sep 2006 A1
20060247167 Schlein et al. Nov 2006 A1
20060275252 Harris et al. Dec 2006 A1
20060287221 Knudsen et al. Dec 2006 A1
20060293232 Levy et al. Dec 2006 A1
20060293499 Bentley et al. Dec 2006 A1
20070010424 Pedersen et al. Jan 2007 A1
20070010656 Beeley et al. Jan 2007 A1
20070014818 Betz et al. Jan 2007 A1
20070021336 Anderson et al. Jan 2007 A1
20070037750 Young et al. Feb 2007 A1
20070049531 Knudsen et al. Mar 2007 A1
20070059373 Oberg Mar 2007 A1
20070059374 Hokenson et al. Mar 2007 A1
20070065469 Betz et al. Mar 2007 A1
20070066528 Beeley et al. Mar 2007 A1
20070092482 Bossard et al. Apr 2007 A1
20070129284 Kjeldsen et al. Jun 2007 A1
20070166352 Wright et al. Jul 2007 A1
20070196416 Li et al. Aug 2007 A1
20070281940 Dugi et al. Dec 2007 A1
20080071063 Allan et al. Mar 2008 A1
20080091176 Alessi et al. Apr 2008 A1
20080119393 Beeley et al. May 2008 A1
20080119569 Wright et al. May 2008 A1
20080125348 Wright et al. May 2008 A1
20080125349 Wright et al. May 2008 A1
20080125351 Wright et al. May 2008 A1
20080125353 Hiles et al. May 2008 A1
20080125361 Ludvigsen et al. May 2008 A1
20080171848 Christiansen et al. Jul 2008 A1
20080176802 Prickett et al. Jul 2008 A1
20080176804 Mack et al. Jul 2008 A1
20080200390 Prickett et al. Aug 2008 A1
20080213288 Michelsen et al. Sep 2008 A1
20080214467 Prickett et al. Sep 2008 A1
20080233053 Gross et al. Sep 2008 A1
20080249007 Lau et al. Oct 2008 A1
20080249018 Kolterman et al. Oct 2008 A1
20080249089 Himmelsbach et al. Oct 2008 A1
20080255159 Himmelsbach et al. Oct 2008 A1
20080260838 Hokenson et al. Oct 2008 A1
20080260847 Wright et al. Oct 2008 A1
20080274952 Soares et al. Nov 2008 A1
20080280814 Ludvigsen et al. Nov 2008 A1
20080300171 Balkan et al. Dec 2008 A1
20080312157 Levy et al. Dec 2008 A1
20080318865 Juul-Mortensen Dec 2008 A1
20090011976 Ludvigsen et al. Jan 2009 A1
20090018053 L'Italien et al. Jan 2009 A1
20090029913 Beeley et al. Jan 2009 A1
20090035253 Wright et al. Feb 2009 A1
20090036364 Levy et al. Feb 2009 A1
20090043264 Glejbol et al. Feb 2009 A1
20090054315 Bock et al. Feb 2009 A1
20090069226 Ong et al. Mar 2009 A1
20090082255 Brunner-Schwarz et al. Mar 2009 A1
20090088369 Steiness Apr 2009 A1
20090098130 Bradshaw et al. Apr 2009 A1
20090110647 Richardson et al. Apr 2009 A1
20090111749 Richardson et al. Apr 2009 A1
20090137456 Dimarchi et al. May 2009 A1
20090137466 Anderson et al. May 2009 A1
20090148534 Yasugi et al. Jun 2009 A1
20090163423 Young et al. Jun 2009 A1
20090170750 Kjeldsen et al. Jul 2009 A1
20090176704 Beeley et al. Jul 2009 A1
20090180953 Gotthardt et al. Jul 2009 A1
20090186817 Ghosh et al. Jul 2009 A1
20090186819 Carrier et al. Jul 2009 A1
20090203597 Rabinovitch et al. Aug 2009 A1
20090203603 Baron et al. Aug 2009 A1
20090215688 Knudsen et al. Aug 2009 A1
20090215694 Kolterman et al. Aug 2009 A1
20090221485 James Sep 2009 A1
20090226431 Habib Sep 2009 A1
20090232775 Bertilsson et al. Sep 2009 A1
20090232807 Glaesner et al. Sep 2009 A1
20090232891 Gelber et al. Sep 2009 A1
20090239796 Fineman et al. Sep 2009 A1
20090247463 Wright et al. Oct 2009 A1
20090253625 Greig et al. Oct 2009 A1
20090258818 Surolia et al. Oct 2009 A1
20090264352 Anderson et al. Oct 2009 A1
20090280169 Leonard Nov 2009 A1
20090280170 Lee et al. Nov 2009 A1
20090286716 Knudsen et al. Nov 2009 A1
20090286723 Levy et al. Nov 2009 A1
20090291886 Ong et al. Nov 2009 A1
20090298757 Bloom Dec 2009 A1
20090308390 Smutney et al. Dec 2009 A1
20090308391 Smutney et al. Dec 2009 A1
20090308392 Smutney et al. Dec 2009 A1
20090325860 Constantino et al. Dec 2009 A1
20100009904 Lv et al. Jan 2010 A1
20100016806 Glejbol et al. Jan 2010 A1
20100022455 Chilkoti Jan 2010 A1
20100029554 Ghosh et al. Feb 2010 A1
20100041867 Shechter et al. Feb 2010 A1
20100056451 Juul-Mortensen et al. Mar 2010 A1
20100087365 Cherif-Cheikh et al. Apr 2010 A1
20100099619 Levy et al. Apr 2010 A1
20100137558 Lee et al. Jun 2010 A1
20100152097 Wright et al. Jun 2010 A1
20100152111 Wright et al. Jun 2010 A1
20100168011 Jennings, Jr. Jul 2010 A1
20100173844 Ludvigsen et al. Jul 2010 A1
20100185184 Alessi et al. Jul 2010 A1
20100190699 Dimarchi et al. Jul 2010 A1
20100190701 Day et al. Jul 2010 A1
20100190715 Schlein et al. Jul 2010 A1
20100196405 Ng et al. Aug 2010 A1
20100197565 Smutney et al. Aug 2010 A1
20100210505 Bossard et al. Aug 2010 A1
20100216692 Brunner-Schwarz et al. Aug 2010 A1
20100240586 Bao et al. Sep 2010 A1
20100247661 Hokenson et al. Sep 2010 A1
20100261637 Spetzler et al. Oct 2010 A1
20100278924 Oberg et al. Nov 2010 A1
20100292172 Ghosh et al. Nov 2010 A1
20100317056 Tiwari et al. Dec 2010 A1
20100317576 Rothkopf Dec 2010 A1
20100331246 Dimarchi et al. Dec 2010 A1
20110003004 Hokenson et al. Jan 2011 A1
20110034373 Coskun et al. Feb 2011 A1
20110034377 Young et al. Feb 2011 A1
20110059181 Hu et al. Mar 2011 A1
20110065633 Dimarchi et al. Mar 2011 A1
20110065731 Dugi et al. Mar 2011 A1
20110071076 Beeley et al. Mar 2011 A1
20110091420 Liu et al. Apr 2011 A1
20110097386 Steiner et al. Apr 2011 A1
20110097751 Nicolaou et al. Apr 2011 A1
20110098217 Dimarchi et al. Apr 2011 A1
20110112277 Kozlowski et al. May 2011 A1
20110118136 Köster et al. May 2011 A1
20110123487 Chilkoti May 2011 A1
20110129522 Mevorat-Kaplan et al. Jun 2011 A1
20110136737 Levy et al. Jun 2011 A1
20110152181 Alsina-Fernandez et al. Jun 2011 A1
20110152182 Alsina-Fernandez et al. Jun 2011 A1
20110152185 Plum et al. Jun 2011 A1
20110166062 Dimarchi et al. Jul 2011 A1
20110166554 Alessi et al. Jul 2011 A1
20110171178 Levetan et al. Jul 2011 A1
20110178014 Hathaway et al. Jul 2011 A1
20110178242 Harris et al. Jul 2011 A1
20110190200 Dimarchi et al. Aug 2011 A1
20110195897 Kajihara et al. Aug 2011 A1
20110230409 Knudsen et al. Sep 2011 A1
20110237503 Alsina-Fernandez et al. Sep 2011 A1
20110237510 Steiner et al. Sep 2011 A1
20110245162 Fineman et al. Oct 2011 A1
20110257092 Dimarchi et al. Oct 2011 A1
20110263496 Fineman et al. Oct 2011 A1
20110281798 Kolterman et al. Nov 2011 A1
20110288003 Dimarchi et al. Nov 2011 A1
20110301080 Bush et al. Dec 2011 A1
20110301081 Becker et al. Dec 2011 A1
20110301084 Lau et al. Dec 2011 A1
20110306549 Tatarkiewicz et al. Dec 2011 A1
20120004168 Young et al. Jan 2012 A1
20120021978 Werner et al. Jan 2012 A1
20120040899 Costello et al. Feb 2012 A1
20120046222 Alfaro-Lopez et al. Feb 2012 A1
20120071510 Leone-Bay et al. Mar 2012 A1
20120071817 Ward et al. Mar 2012 A1
20120094356 Chung et al. Apr 2012 A1
20120100070 Ahn et al. Apr 2012 A1
20120122783 Dimarchi et al. May 2012 A1
20120135922 Prickett et al. May 2012 A1
20120136318 Lanin et al. May 2012 A1
20120148586 Chou et al. Jun 2012 A1
20120149639 Balkan et al. Jun 2012 A1
20120157932 Glejbol et al. Jun 2012 A1
20120172295 Dimarchi et al. Jul 2012 A1
20120177697 Chen Jul 2012 A1
20120196795 Xu et al. Aug 2012 A1
20120196796 Soares et al. Aug 2012 A1
20120196802 Lv et al. Aug 2012 A1
20120196804 Dimarchi et al. Aug 2012 A1
20120208755 Leung et al. Aug 2012 A1
20120208831 Himmelsbach et al. Aug 2012 A1
20120209213 Theucher Aug 2012 A1
20120225810 Pedersen et al. Sep 2012 A1
20120231022 Bass et al. Sep 2012 A1
20120238493 Dimarchi et al. Sep 2012 A1
20120238496 Fan et al. Sep 2012 A1
20120253023 Levy et al. Oct 2012 A1
20120258912 Bentley et al. Oct 2012 A1
20120258985 Kozlowski et al. Oct 2012 A1
20120264683 Coskun et al. Oct 2012 A1
20120264684 Kajihara et al. Oct 2012 A1
20120276098 Hamilton et al. Nov 2012 A1
20120277154 Fan et al. Nov 2012 A1
20120283179 Brunner-Schwarz et al. Nov 2012 A1
20120294855 Van Cauter et al. Nov 2012 A1
20120295836 Knudsen et al. Nov 2012 A1
20120295846 Hagendorf et al. Nov 2012 A1
20120295850 Tatarkiewicz et al. Nov 2012 A1
20120302501 Coolidge et al. Nov 2012 A1
20120309975 Colca et al. Dec 2012 A1
20120316108 Chen et al. Dec 2012 A1
20120316138 Colca et al. Dec 2012 A1
20120322725 Dimarchi et al. Dec 2012 A1
20120322728 Colca et al. Dec 2012 A1
20120329715 Greig et al. Dec 2012 A1
20130005664 Chilkoti Jan 2013 A1
20130023470 Young et al. Jan 2013 A1
20130023471 Rabinovitch et al. Jan 2013 A1
20130046245 Raab et al. Feb 2013 A1
20130053350 Colca et al. Feb 2013 A1
20130065826 Soula et al. Mar 2013 A1
20130079277 Chilkoti Mar 2013 A1
20130079278 Lau et al. Mar 2013 A1
20130084277 Arnold et al. Apr 2013 A1
20130085099 Chilkoti Apr 2013 A1
20130085104 Chilkoti Apr 2013 A1
20130089878 Nicolaou et al. Apr 2013 A1
20130090286 Dimarchi et al. Apr 2013 A1
20130095037 Gotthardt et al. Apr 2013 A1
20130096258 Bossard et al. Apr 2013 A1
20130104887 Smutney et al. May 2013 A1
20130116172 Dimarchi et al. May 2013 A1
20130116175 Shechter et al. May 2013 A1
20130118491 Richardson et al. May 2013 A1
20130123178 Dimarchi et al. May 2013 A1
20130123462 Dimarchi et al. May 2013 A1
20130125886 Richardson et al. May 2013 A1
20130130977 Wright et al. May 2013 A1
20130137631 Levy et al. May 2013 A1
20130137645 Rosendahl May 2013 A1
20130142795 Bai et al. Jun 2013 A1
20130156849 De Fougerolles et al. Jun 2013 A1
20130157934 Dimarchi et al. Jun 2013 A1
20130157953 Petersen et al. Jun 2013 A1
20130164310 Annathur et al. Jun 2013 A1
20130165370 Bock et al. Jun 2013 A1
20130165379 Kolterman et al. Jun 2013 A1
20130172274 Chilkoti Jul 2013 A1
20130178411 Chilkoti Jul 2013 A1
20130178415 Soula et al. Jul 2013 A1
20130184203 Alfaro-Lopez et al. Jul 2013 A1
20130184443 Bentley et al. Jul 2013 A1
20130189328 Cleeman et al. Jul 2013 A1
20130189365 Hokenson et al. Jul 2013 A1
20130199527 Smutney et al. Aug 2013 A1
20130203660 Day et al. Aug 2013 A1
20130209586 Hathaway et al. Aug 2013 A1
20130217622 Lee et al. Aug 2013 A1
20130236974 De Fougerolles Sep 2013 A1
20130237592 De Fougerolles et al. Sep 2013 A1
20130237593 De Fougerolles et al. Sep 2013 A1
20130237594 De Fougerolles et al. Sep 2013 A1
20130244278 De Fougerolles et al. Sep 2013 A1
20130244279 De Fougerolles et al. Sep 2013 A1
20130245104 De Fougerolles et al. Sep 2013 A1
20130245105 De Fougerolles et al. Sep 2013 A1
20130245106 De Fougerolles et al. Sep 2013 A1
20130245107 De Fougerolles et al. Sep 2013 A1
20130252281 De Fougerolles et al. Sep 2013 A1
20130253043 De Fougerolles et al. Sep 2013 A1
20130259923 Bancel et al. Oct 2013 A1
20130259924 Bancel et al. Oct 2013 A1
20130266640 De Fougerolles et al. Oct 2013 A1
20130280206 Kozlowski et al. Oct 2013 A1
20130281368 Bilsky et al. Oct 2013 A1
20130281374 Levy et al. Oct 2013 A1
20130284912 Vogel et al. Oct 2013 A1
20130288958 Lau et al. Oct 2013 A1
20130289241 Bai et al. Oct 2013 A1
20130291866 Smutney et al. Nov 2013 A1
20130291867 Smutney et al. Nov 2013 A1
20130296236 Silvestre et al. Nov 2013 A1
20130303442 Levy et al. Nov 2013 A1
20130310310 Liu et al. Nov 2013 A1
20130310538 Chilkoti Nov 2013 A1
20130331322 Young et al. Dec 2013 A1
20130336893 Haack et al. Dec 2013 A1
20130338065 Smutney et al. Dec 2013 A1
20130338071 Knudsen et al. Dec 2013 A1
20130345134 Sauerberg et al. Dec 2013 A1
20140007873 Smutney et al. Jan 2014 A1
20140011732 Spetzler et al. Jan 2014 A1
20140014106 Smutney et al. Jan 2014 A1
20140017208 Osei Jan 2014 A1
20140031281 Wright et al. Jan 2014 A1
20140038891 Prickett et al. Feb 2014 A1
20140056924 Van Cauter Feb 2014 A1
20140066368 Mack et al. Mar 2014 A1
20140083421 Smutney et al. Mar 2014 A1
20140088003 Wright et al. Mar 2014 A1
20140100156 Haack et al. Apr 2014 A1
20140107019 Erickson et al. Apr 2014 A1
20140107021 Dimarchi et al. Apr 2014 A1
20140120120 Woo et al. May 2014 A1
20140121352 Shechter et al. May 2014 A1
20140128318 Jung et al. May 2014 A1
20140128604 Himmelsbach et al. May 2014 A1
20140135348 Dugi et al. May 2014 A1
20140141467 Tiwari et al. May 2014 A1
20140142037 Yue May 2014 A1
20140162943 Alfaro-Lopez et al. Jun 2014 A1
20140187483 Steiness Jul 2014 A1
20140200183 Hathaway et al. Jul 2014 A1
20140206608 Haack et al. Jul 2014 A1
20140206609 Haack et al. Jul 2014 A1
20140206613 Rabinovitch et al. Jul 2014 A1
20140206615 Knudsen et al. Jul 2014 A1
20140212419 Dimarchi et al. Jul 2014 A1
20140212440 Jung et al. Jul 2014 A1
20140213513 Haack et al. Jul 2014 A1
20140213516 Chilkoti Jul 2014 A1
20140220029 Michelsen et al. Aug 2014 A1
20140220134 Zierhut et al. Aug 2014 A1
20140221280 Bloom Aug 2014 A1
20140221281 Haack et al. Aug 2014 A1
20140221282 Sun et al. Aug 2014 A1
20140227264 Hamilton et al. Aug 2014 A1
20140235535 Erickson et al. Aug 2014 A1
20140243263 Rothkopf Aug 2014 A1
20140249299 Levy et al. Sep 2014 A1
20140308358 Oberg et al. Oct 2014 A1
20140309168 Rosendahl Oct 2014 A1
20140315953 Leone-Bay et al. Oct 2014 A1
20150011467 Bloom et al. Jan 2015 A1
20150126440 Day et al. May 2015 A1
20150164995 Kadereit et al. Jun 2015 A1
20150164996 Kadereit et al. Jun 2015 A1
20150164997 Haack et al. Jun 2015 A1
20150166625 Haack Jun 2015 A1
20150166627 Kadereit et al. Jun 2015 A1
20150216941 Bley et al. Aug 2015 A1
20150232527 Gong et al. Aug 2015 A1
20150315260 Bossart et al. Nov 2015 A1
20150322128 Bossart et al. Nov 2015 A1
20150322129 Bossart et al. Nov 2015 A1
20150368311 Haack et al. Dec 2015 A1
20160168225 Haack et al. Jun 2016 A1
20160220643 Haack et al. Aug 2016 A1
20160235855 Xiong et al. Aug 2016 A1
20170008944 Bossart et al. Jan 2017 A1
20170216406 Haack et al. Aug 2017 A1
20180154005 Kadereit et al. Jun 2018 A1
Foreign Referenced Citations (345)
Number Date Country
101538323 Sep 2009 CN
101559041 Oct 2009 CN
101663317 Mar 2010 CN
101798588 Aug 2010 CN
101870728 Oct 2010 CN
101601646 Mar 2011 CN
102100906 Jun 2011 CN
102363633 Feb 2012 CN
102421796 Apr 2012 CN
101444618 Jun 2012 CN
102532301 Jul 2012 CN
102649947 Aug 2012 CN
102816244 Dec 2012 CN
102827270 Dec 2012 CN
101670096 Jan 2013 CN
103304660 Sep 2013 CN
103421094 Dec 2013 CN
103665148 Mar 2014 CN
103833841 Jun 2014 CN
103908657 Jul 2014 CN
102766204 Oct 2014 CN
104926934 Sep 2015 CN
1 140 145 Jul 2005 EP
0 619 322 Dec 2005 EP
1 609 478 Dec 2005 EP
1 143 989 Dec 2006 EP
1 790 665 May 2007 EP
1 658 856 Mar 2010 EP
1 684 793 Sep 2011 EP
1 633 391 Oct 2011 EP
2 387 989 Nov 2011 EP
1 633 390 Jan 2012 EP
2 494 983 Sep 2012 EP
2 626 368 Aug 2013 EP
2 664 374 Nov 2013 EP
1 817 048 Feb 2014 EP
2 769 990 Aug 2014 EP
2014-227368 Dec 2014 JP
10-2012-0137271 Dec 2012 KR
10-2012-0139579 Dec 2012 KR
10-2014-0018462 Feb 2014 KR
10-2014-0058104 May 2014 KR
10-2014-0058387 May 2014 KR
10-2014-0130659 Nov 2014 KR
10-2014-0133493 Nov 2014 KR
2009121626 Feb 2011 RU
1996019229 Jun 1996 WO
1998005351 Feb 1998 WO
1998008871 Mar 1998 WO
1998030231 Jul 1998 WO
1999007404 Feb 1999 WO
1999025727 May 1999 WO
1999025728 May 1999 WO
1999034822 Jul 1999 WO
1999043708 Sep 1999 WO
1999047160 Sep 1999 WO
1999064061 Dec 1999 WO
2000015224 Mar 2000 WO
2000037098 Jun 2000 WO
2000041546 Jul 2000 WO
2000041548 Jul 2000 WO
2000055119 Sep 2000 WO
2000066629 Nov 2000 WO
2000071175 Nov 2000 WO
2000073331 Dec 2000 WO
2001051078 Jul 2001 WO
2002016309 Feb 2002 WO
2002034285 May 2002 WO
2002067989 Sep 2002 WO
2003011892 Feb 2003 WO
2003020201 Mar 2003 WO
2003061362 Jul 2003 WO
2003077851 Sep 2003 WO
2003084563 Oct 2003 WO
2003092581 Nov 2003 WO
2003099314 Dec 2003 WO
2003101395 Dec 2003 WO
2003105888 Dec 2003 WO
2003105897 Dec 2003 WO
2004004779 Jan 2004 WO
2004004780 Jan 2004 WO
2004004781 Jan 2004 WO
2004005342 Jan 2004 WO
2004012672 Feb 2004 WO
2004018468 Mar 2004 WO
2004035623 Apr 2004 WO
2004045592 Jun 2004 WO
2004056313 Jul 2004 WO
2004056317 Jul 2004 WO
2004089280 Oct 2004 WO
2004089985 Oct 2004 WO
2004105781 Dec 2004 WO
2004105790 Dec 2004 WO
2005000222 Jan 2005 WO
2005000360 Jan 2005 WO
2005012347 Feb 2005 WO
2005021022 Mar 2005 WO
2005046716 May 2005 WO
2005048989 Jun 2005 WO
2005049061 Jun 2005 WO
2005049069 Jun 2005 WO
2005054291 Jun 2005 WO
2005077072 Aug 2005 WO
2005077094 Aug 2005 WO
2005081619 Sep 2005 WO
2005102293 Nov 2005 WO
2005110425 Nov 2005 WO
2005115437 Dec 2005 WO
2005117584 Dec 2005 WO
2005120492 Dec 2005 WO
2006017688 Feb 2006 WO
2006024275 Mar 2006 WO
2006024631 Mar 2006 WO
2006029634 Mar 2006 WO
2006037811 Apr 2006 WO
2006044531 Apr 2006 WO
2006051103 May 2006 WO
2006051110 May 2006 WO
2006066024 Jun 2006 WO
2006069388 Jun 2006 WO
2006073890 Jul 2006 WO
2006074600 Jul 2006 WO
2006083254 Aug 2006 WO
2006086769 Aug 2006 WO
2006097535 Sep 2006 WO
2006110887 Oct 2006 WO
2006114396 Nov 2006 WO
2006125763 Nov 2006 WO
2006134340 Dec 2006 WO
2006138572 Dec 2006 WO
2007019331 Feb 2007 WO
2007022123 Mar 2007 WO
2007024700 Mar 2007 WO
2007033316 Mar 2007 WO
2007033372 Mar 2007 WO
2007035665 Mar 2007 WO
2007047834 Apr 2007 WO
2007047922 Apr 2007 WO
2007056362 May 2007 WO
2007064691 Jun 2007 WO
2007065156 Jun 2007 WO
2007067964 Jun 2007 WO
2007075534 Jul 2007 WO
2007109354 Sep 2007 WO
2007120899 Oct 2007 WO
2007121411 Oct 2007 WO
2007128761 Nov 2007 WO
2007133778 Nov 2007 WO
2007139941 Dec 2007 WO
2007140284 Dec 2007 WO
2008021133 Feb 2008 WO
2008021560 Feb 2008 WO
2008023050 Feb 2008 WO
2008038147 Apr 2008 WO
2008058461 May 2008 WO
2008071972 Jun 2008 WO
2008073448 Jun 2008 WO
2008081418 Jul 2008 WO
2008086086 Jul 2008 WO
2008098212 Aug 2008 WO
2008101017 Aug 2008 WO
2008148839 Dec 2008 WO
2008152403 Dec 2008 WO
2009020802 Feb 2009 WO
2009024015 Feb 2009 WO
2009029847 Mar 2009 WO
2009030771 Mar 2009 WO
2009035540 Mar 2009 WO
2009055740 Apr 2009 WO
2009055742 Apr 2009 WO
2009058662 May 2009 WO
2009058734 May 2009 WO
2009063072 May 2009 WO
2009067268 May 2009 WO
2009095479 Aug 2009 WO
2009099763 Aug 2009 WO
WO2009095479 Aug 2009 WO
WO-2009095479 Aug 2009 WO
2009113099 Sep 2009 WO
2009137078 Nov 2009 WO
2009137080 Nov 2009 WO
2009143014 Nov 2009 WO
2009143285 Nov 2009 WO
2009152477 Dec 2009 WO
2009153960 Dec 2009 WO
2009155257 Dec 2009 WO
2009155258 Dec 2009 WO
2009158704 Dec 2009 WO
2010011439 Jan 2010 WO
2010013012 Feb 2010 WO
2010043566 Apr 2010 WO
2010070251 Jun 2010 WO
2010070252 Jun 2010 WO
2010070253 Jun 2010 WO
2010070255 Jun 2010 WO
2010071807 Jun 2010 WO
2010096052 Aug 2010 WO
2010096142 Aug 2010 WO
2010102148 Sep 2010 WO
2010120476 Oct 2010 WO
2010121559 Oct 2010 WO
2010123290 Oct 2010 WO
2010133675 Nov 2010 WO
2010133676 Nov 2010 WO
2010138671 Dec 2010 WO
2010142665 Dec 2010 WO
2010148089 Dec 2010 WO
2011000095 Jan 2011 WO
2011006497 Jan 2011 WO
2011011675 Jan 2011 WO
2011012718 Feb 2011 WO
2011020319 Feb 2011 WO
2011020320 Feb 2011 WO
2011024110 Mar 2011 WO
2011039096 Apr 2011 WO
2011049713 Apr 2011 WO
2011052523 May 2011 WO
2011056713 May 2011 WO
2011058082 May 2011 WO
2011058083 May 2011 WO
2011075393 Jun 2011 WO
2011075514 Jun 2011 WO
2011075623 Jun 2011 WO
2011080103 Jul 2011 WO
2011084453 Jul 2011 WO
2011084456 Jul 2011 WO
2011084459 Jul 2011 WO
2011087671 Jul 2011 WO
2011087672 Jul 2011 WO
2011088837 Jul 2011 WO
2011094337 Aug 2011 WO
2011109784 Sep 2011 WO
2011117415 Sep 2011 WO
2011117416 Sep 2011 WO
2011119657 Sep 2011 WO
2011143208 Nov 2011 WO
2011143209 Nov 2011 WO
2011144751 Nov 2011 WO
2011153965 Dec 2011 WO
2011156407 Dec 2011 WO
2011160630 Dec 2011 WO
2011162830 Dec 2011 WO
2011163012 Dec 2011 WO
2011163272 Dec 2011 WO
2011163473 Dec 2011 WO
2012012352 Jan 2012 WO
2012012460 Jan 2012 WO
2012015975 Feb 2012 WO
2012031518 Mar 2012 WO
2012035139 Mar 2012 WO
2012050923 Apr 2012 WO
2012059762 May 2012 WO
2012064892 May 2012 WO
2012080471 Jun 2012 WO
2012088116 Jun 2012 WO
2012088157 Jun 2012 WO
2012122535 Sep 2012 WO
2012130015 Oct 2012 WO
2012138941 Oct 2012 WO
2012140647 Oct 2012 WO
2012150503 Nov 2012 WO
2012158965 Nov 2012 WO
2012162547 Nov 2012 WO
2012167744 Dec 2012 WO
2012169798 Dec 2012 WO
2012173422 Dec 2012 WO
2012177443 Dec 2012 WO
2012177444 Dec 2012 WO
2012177929 Dec 2012 WO
2013002580 Jan 2013 WO
2013004983 Jan 2013 WO
2013009545 Jan 2013 WO
2013029279 Mar 2013 WO
2013041678 Mar 2013 WO
2012174478 May 2013 WO
2013060850 May 2013 WO
2013074910 May 2013 WO
2013078500 Jun 2013 WO
2013090648 Jun 2013 WO
2013092703 Jun 2013 WO
2013093720 Jun 2013 WO
2013101749 Jul 2013 WO
2013104861 Jul 2013 WO
2013148871 Oct 2013 WO
2013148966 Oct 2013 WO
2013151663 Oct 2013 WO
2013151664 Oct 2013 WO
2013151665 Oct 2013 WO
2013151666 Oct 2013 WO
2013151667 Oct 2013 WO
2013151668 Oct 2013 WO
2013151669 Oct 2013 WO
2013151670 Oct 2013 WO
2013151671 Oct 2013 WO
2013151672 Oct 2013 WO
2013151736 Oct 2013 WO
2013160397 Oct 2013 WO
2013163162 Oct 2013 WO
2013164484 Nov 2013 WO
2013171135 Nov 2013 WO
2013177565 Nov 2013 WO
2013186240 Dec 2013 WO
2013192129 Dec 2013 WO
2013192130 Dec 2013 WO
2014012069 Jan 2014 WO
2014016300 Jan 2014 WO
2014017843 Jan 2014 WO
2014017845 Jan 2014 WO
2014017849 Jan 2014 WO
2014027253 Feb 2014 WO
2014027254 Feb 2014 WO
2014041195 Mar 2014 WO
2014041375 Mar 2014 WO
2014049610 Apr 2014 WO
2014056872 Apr 2014 WO
2014073842 May 2014 WO
2014073845 May 2014 WO
2014081872 May 2014 WO
2014091316 Jun 2014 WO
2014096145 Jun 2014 WO
WO 2014096148 Jun 2014 WO
WO 2014096149 Jun 2014 WO
WO 2014096150 Jun 2014 WO
2014140222 Sep 2014 WO
2014152460 Sep 2014 WO
2014158900 Oct 2014 WO
2014170496 Oct 2014 WO
2015055801 Apr 2015 WO
2015055802 Apr 2015 WO
2015067716 May 2015 WO
2015086728 Jun 2015 WO
2015086729 Jun 2015 WO
2015086730 Jun 2015 WO
2015086731 Jun 2015 WO
2015086732 Jun 2015 WO
2015086733 Jun 2015 WO
WO 2015086731 Jun 2015 WO
2015100876 Jul 2015 WO
2015104314 Jul 2015 WO
2015132599 Sep 2015 WO
2016055610 Apr 2016 WO
2016065090 Apr 2016 WO
2016198604 Dec 2016 WO
2016198624 Dec 2016 WO
WO 2018100174 Jun 2018 WO
Non-Patent Literature Citations (192)
Entry
US 8,729,011 B2, 05/2014, Dimarchi et al. (withdrawn)
Xu, Cancer Res Prey Treat, vol. 42, No. 4, 2013 (Year: 2013).
Xiaoyu, Progress in Pharmaceutical Sciences, vol. 31, 2007 (Year: 2007).
Aramadhaka et al. (Apr. 18, 2013) “Connectivity maps for biosimilar drug discovery in venoms: The case of Gila Monster Venom and the anti-diabetes drug Byetta®,” Toxicon. 69:160-167.
Bhavsar et al. (Mar. 2013) “Evolution of exenatide as a diabetes therapeutic,” Curr. Diabetes Rev. 9(2):161-193.
Gao et al. (Jun. 4, 2012) “A site-specific PEGylated analog of exendin-4 with improved pharmacokinetics and pharmacodynamics in vivo,” J. Pharm. Pharmacol. 64(11):1646-1653.
Gupta (May 2013) “Glucagon-like peptide-1 analogues: An overview,” Indian J. Endocrinol. Metab. 17(3):413-421.
Hou et al. (Jan. 23, 2013) “Long-term treatment with EXf, a peptide analog of Exendin-4, improves β-cell function and survival in diabetic KKAy mice,” Peptides. 40:123-132.
Kim et al. (Nov. 9, 2012) “Site-specific PEGylated Exendin-4 modified with a high molecular weight trimeric PEG reduces steric hindrance and increases type 2 antidiabetic therapeutic effects,” Bioconjug. Chem. 23(11):2214-2220.
Lee et al. (Oct. 17, 2013) “Decanoic acid-modified glycol chitosan hydrogels containing tightly adsorbed palmityl-acylated exendin-4 as a long-acting sustained-release anti-diabetic system,” Acta Biomater. 10(2):812-820.
Parkes et al. (Dec. 12, 2012) “Discovery and development of exenatide: the first antidiabetic agent to leverage the multiple benefits of the incretin hormone, GLP-1,” Expert Opin. Drug Discov. 8(2):219-244.
Qian et al. (Jul. 1, 2013) “Characterization of a site-specific PEGylated analog of exendin-4 and determination of the PEGylation site,” Int. J. Pharm. 454(1):553-558.
Simonsen et al. (Jan. 11, 2013) “The C-terminal extension of exendin-4 provides additional metabolic stability when added to GLP-1, while there is minimal effect of truncating exendin-4 in anaesthetized pigs,” Regul. Pept. 181:17-21.
Sun et al. (Nov. 6, 2013) “Bifunctional PEGylated exenatide-amylinomimetic hybrids to treat metabolic disorders: an example of long-acting dual hormonal therapeutics,” J. Med. Chem. 56(22):9328-9341.
Yim et al. (Aug. 8, 2013) “Synthesis and preclinical characterization of [64Cu]NODAGA-MAL-exendin-4 with a Nϵ-maleoyl-L-lysyl-glycine linkage,” Nucl. Med. Biol. 40(8):1006-1012.
Yue et al. (Jan. 28, 2013) “Development of a new thiol site-specific prosthetic group and its conjugation with [Cys(40)]-exendin-4 for in vivo targeting of insulinomas,” Bioconjug. Chem. 24(7):1191-1200.
Guryanov et al. (May 30, 2016) “Innovative chemical synthesis and conformational hints on the lipopeptide liraglutide,” J. Pept. Sci. 22:471-479.
Lau et al. (Aug. 26, 2015) “Discovery of the once-weekly Glucagon-like Peptide-1 (GLP-1) analogue Semaglutide,” Journal of Medicinal Chemistry. 58:7370-7380.
International Search Report with Written Opinion corresponding to International Patent Application No. PCT/EP2016/063305, dated Oct. 4, 2016.
International Search Report with Written Opinion corresponding to International Patent Application No. PCT/EP2016/066299, dated Jan. 10, 2017.
Shendi et al. (Mar. 14, 2016) “Tunable, bioactive protein conjugated hyaluronic acid hydrogel for neural engineering applications,” J. Mater. Chem. B. 4:2803-2818.
Amylin Pharmaceuticals, Inc. (2007) “Byetta: Exenatide Injection,” Product Information. Accessible on the Internet at URL: http://www.accessdata.fda.gov/drugsaffda_docs/label/2008/021773s012lbl.pdf. [Last Accessed Jun. 2, 2014].
Baggio et al. (2007) “Biology of incretins: GLP-1 and GIP,” Gastroenterology. 132:2131-2157.
Bhat et al. (Jun. 1, 2013) “A novel GIP-oxyntomodulin hybrid peptide acting through GIP, glucagon and GLP-1 receptors exhibits weight reducing and anti-diabetic properties,” Biochem. Pharmacol. 85:1655-1662.
Ghat et al. (Mar. 17, 2013) “A DPP-IV-resistant triple-acting agonist of GIP, GLP-1 and glucagon receptors with potent glucose-lowering and insulinotropic actions in high-fat-fed mice,” Diabetologia. 56:1417-1424.
Biron et al. (2006) “Optimized selective N-methylation of peptides on solid support,” J. Peptide Sci. 12:213-219.
Bis et al. (Jun. 27, 2014) “Antimicrobial preservatives induce aggregation of interferon alpha-2a: the order in which preservatives induce protein aggregation is independent of the protein,” Int. J. Pharm. 472:356-361.
Braga et al. (2005) “Making Crystals from Crystals: a green route to crystal engineering and polymorphism,” Chem. Commun. 2005:3635-3645.
Bromer (1983) “Chemical Characteristics of Glucagon,” Handbook of Experimental Pharmacology. 66:1-22.
Bunck et al. (Sep. 2011) “Effects of Exenatide on Measures of B-Cell Function After 3 Years in Metformin-Treated Patients with Type 2 Diabetes,” Diabetes Care. 34:2041-2047.
Buse et al. (2009) “Liraglutide once a day versus exenatide twice a day for type 2 diabetes: a 26-week randomised, parallel group, multinational, open-label trial (LEAD-6),” The Lancet. 374:39-47.
Chae et al. (2010) “The fatty acid conjugated exendin-4 analogs for type 2 antidiabetic therapeutics,” Journal of Controlled Release. 144:10-16.
Chen et al. (Jan. 2014) “Hyaluronic acid-based drug conjugates: state-of-the-art and perspectives,” J. Biomed. Nanotechnol. 10(1):4-16.
Chhabra et al. (1998) “An Appraisal of New Variants of Dde Amine Protecting Group for Solid Phase Peptide Synthesis,” Tetrahedron Letters. 39:1603-1606.
Creutzfeld et al. (1978) “Gastric inhibitory polypeptide (GIP) and insulin in obesity: increased response to stimulation and defective feedback control of serum levels,” Diabetologia. 14:15-24.
Day et al. (2009) “A New Glucagon and GLP-1 co-agonist Eliminates Obesity in Rodents,” Nature Chemical Biology. 5 (10):749-757.
Deacon (2004) “Circulation and degradation of GIP and GLP-1,” Horm. Metab. Res. 36:761-765.
Donnelly (May 2012) “The structure and function of the glucagon-like peptide-1 receptor and its ligands,” Br. J. Pharmacol. 166(1):27-41.
Druce et al. (2009) “Investigation of structure-activity relationships of Oxyntomodulin (Oxm) using Oxm analogs,” Endocrinology. 150(4):1712-1722.
Drucker et al. (2010) “Liraglutide,” New Reviews—Drug Discovery. 9(4):267-268.
Eng et al. (1990) “Purification and structure of exendin-3, a new pancreatic secretagogue isolated from Heloderma horridum venom,” J. Biol. Chem. 265:20259-20262.
Eng et al. (1992) “Isolation and Characterization of Exendin-4, an Exendin-3 Analogue, from Heloderma Suspectum Venom,” The Journal of Biological Chemistry. 267(11):7402-7405.
Eng et al. (1996) “Prolonged Effect of Exendin-4 on Hyperglycemia of db/db Mice,” Diabetes. 45:152A. Abstract 554.
Ferry, Jr. “Diabetes Health (cont.),” MedicineNet. Accessible on the Internet at URL: http://www.onhealth.com/diabetes_health/page3.htm. [Last Accessed Aug. 22, 2013].
Ficht et al. (2008) “Solid-phase Synthesis of Peptide and Glycopeptide Thioesters through Side-Chain-Anchoring Strategies,” Chem. Eur. J. 14:3620-3629.
Finan et al. (Dec. 8, 2014) “A rationally designed monomeric peptide triagonist corrects obesity and diabetes in rodents,” Nat. Med. 21(1):27-36.—with supplementary information.
Finan et al. (Oct. 30, 2013) “Unimolecular Dual Incretins Maximize Metabolic Benefits in Rodents, Monkeys, and Humans,” Sci. Trans. Med. 5:209RA151.
Furman (Mar. 15, 2012) “The development of Byetta (exenatide) from the venom of the Gilo monster as an anti-diabetic agent,” Toxicon. 59:464-471.
Gault et al. (2007) “Chemical gastric inhibitory polypeptide receptor antagonism protects against obesity, insulin resistance, glucose intolerance and associated disturbances in mice fed high-fat and cafeteria diets,” Diabetologia. 50:1752-1762.
Gault et al. (Aug. 1, 2011) “Administration of an acylated GLP-1 and GIP preparation provides added beneficial glucose-lowering and insulinotropic actions over single incretins in mice with Type 2 diabetes and obesity,” Clin Sci (Lond). 121:107-117.
Gentilella et al. (2009) “Exenatide: A Review from Pharmacology to Clinical Practice,” Diabetes, Obesity, and Metabolism. 11:544-556.
Göke et al. (1993) “Exendin-4 is a high potency agonist and truncated exendin-(9-39)-amide an antagonist at the glucagon-like peptide 1-(7-36)-amide receptor of insulin-secreting beta-cells,” J. Biol. Chem. 268:19650-19655.
Hadji-Georgopoulos et al. (1983) “Increased gastric inhibitory polypeptide levels in patients with symptomatic postprandial hypoglycemia,” J. Endocrinol. Metabol. 56(4):648-652.
Hargrove et al. (2007) “Biological Activity of AC3174, A Peptide Analog of Exendin-4,” Regulatory Peptides. 141:113-119.
Heppner et al. (2010) “Glucagon regulation of energy metabolism,” Physiol. Behav. 100:545-548.
Herling et al. (1998) “Pharmacodynamic profile of a novel inhibitor of the hepatic glucose-6-phosphatase system,” Am. J. Physiol. 274(6 Pt 1):G1087-G1093.
Hjorth et al. (1994) “Glucagon and Glucagon-like Peptide 1: Selective Receptor Recognition via Distinct Peptide Epitopes,” The Journal of Biological Chemistry. 269(48):30121-30124.
Holst (2007) “The physiology of glucagon-like peptide 1,” Physiol. Rev. 87(4):1409-1439.
Joshi et al. (2000) “The degradation pathways of glucagon in acidic solutions,” Int. J. Pharm. 203(1-2):115-125.
Kaiser et al. (1970) “Color test for detection of free terminal amino groups in the solid-phase synthesis of peptides.” Anal. Biochem. 34:595-598.
Kamerzell et al. (2011) “Protein—excipient interactions: Mechanisms and biophysical characterization applied to protein formulation development,” Adv. Drug Deliv. Rev. 63:1118-1159.
Kazakos et al. (2011) “Incretin effect: GLP-1, GIP, DPP4,” Diabetes Res Clin Pract. 93(Suppl 1):S32-536. et al. (2011) “Incretin effect: GLP-1, GIP, DPP4,” Diabetes Res Clin Pract. 93(Suppl 1):S32-S36.
King et al. (1990) “A Cleavage Method which Minimizes Side Reactions Following Fmoc Solid Phase Peptide Synthesis,” International Journal of Peptide Protein Research. 36:255-266.
Knudsen et al. (2000) “Potent derivatives of glucagon-like peptide-1 with pharmacokinetic properties suitable for once daily administration” J. Med. Chem. 43(9):1664-1669.
Kong et al. (2010) “Long acting hyaluronate—exendin 4 conjugate for the treatment of type 2 diabetes,” Biomaterials. 31:4121-4128.
Korczyn et al. (2002) “Emerging Therapies in the Pharmacological Treatment of Parkinson's Disease,” Drugs. 62:775-786.
Kosinski et al. (Mar. 16, 2012) “The glucagon receptor is involved in mediating the body weight-lowering effects of oxyntomodulin,” Obesity (Silver Spring). 20:1566-1571.
Krstenansky et al. (1986) “Importance of the 10-13 Region of Glucagon for Its Receptor Interaction and Activation of Adenylate Cyclase,” Biochemistry. 25(13):3833-3839.
Lee et al. (May 10, 2013) “Hormonal Response to a Mixed-Meal Challenge After Reversal of Gastric Bypass for Hypoglycemia,” J. Clin. Endocrinol. Metab. 98(7):E1208-E1212.
Li et al. (Jul. 25, 2012) “Cloning, expressing of Exendin-4 analogue and bioactivity analysis in vivo,” Chinese Journal of Biotechnology. 28(7):877-886.
Liu et al. (2011) “Solid phase peptide synthesis and analysis for exendin-4,” China Biotechnology. 31(2):69-73.—English abstract and drawings.
Bayram et al. (Sep. 2014) “Effects of glucagon-like peptide-1 in diabetic rat small resistance arteries,” Journal of Cardiovascular Pharmacology. 64(3):277-84.
Brom et al. (Feb. 1, 2014) “Non-invasive quantification of the beta cell mass by SPECT with 111In-labelled exendin,” Diabetologia. 57(5):950-959.
Cai et al. (Dec. 2014) “Rb and p107 are required for alpha cell survival, beta cell cycle control and glucagon-like peptide-1 action,” Diabetologia. 57(12):2555-2565.
Charokopou et al. (Nov. 2014) “Cost-effectiveness of saxagliptin compared to GLP-1 analogues as an add-on to insulin in the treatment of type 2 diabetes mellitus from a UK health care perspective,” Value in Health. 17(7):A347. Abstract No. PDB89.
Chen et al. (Dec. 14, 2013) “Exendin-4 is effective against metabolic disorders induced by intrauterine and postnatal overnutrition in rodents,” Diabetologia. 57(3):614-622.
Choi et al. (Jun. 2014) “A long-acting exendin-4 analog conjugate to the human fc fragment reveals low immunogenic potential,” Diabetes. 63(Suppl 1):A259-A260. Abstract No. 1009-P.
Clemmensen et al. (Dec. 30, 2013) “GLP-1/glucagon coagonism restores leptin responsiveness in obese mice chronically maintained on an obesogenic diet,” Diabetes. 63(4):1422-1427.
De Marinis et al. (Jun. 2014) “Differential action of GLP-1 and GIP on human pancreatic islet function and viability,” Diabetes. 63(Suppl 1):A52. Abstract No. 196-OR.
De Marinis et al. (Sep. 2014) “Differential action of GLP-1 and GIP on human pancreatic islet function and viability,” Diabetologia. 57(Suppl 1):S171. Abstract No. 401.
Eriksson et al. (Feb. 10, 2014) “Detection of metastatic insulinoma by positron emission tomography with [(68)ga] exendin-4—a case report,” J. Clin. Endocrinol. Metab. 99(5):1519-1524.
Eriksson et al. (May 2014) “Effects of the glucagon-like peptide-1 analog exendin-4 on reendothelialization and intimal hyperplasia formation in an animal model of vascular injury,” Arteriosclerosis, Thrombosis, and Vascular Biology. 34(Suppl 1): Abstract No. 515.
Gong et al. (Apr. 18, 2014) “Geniposide and its iridoid analogs exhibit antinociception by acting at the spinal GLP-1 receptors,” Neuropharmacology. 84:31-45.
Gupta et al. (Sep. 25, 2014) “Mitigation of autophagy ameliorates hepatocellular damage following ischemia reperfusion injury in murine steatotic liver,” Am. J. Physiol. Gastrointest. Liver Physiol. 307(11):G1088-G1099.
Jerlhag et al. (Jun. 2014) “A glucagon like peptide-1 analogue reduces alcohol intake and prevents relapse drinking,” Alcoholism: Clinical and Experimental Research. 38(Suppl 1):85A. Abstract No. 0339.
Jin et al. (Jun. 24, 2014) “Dipeptidyl peptidase IV inhibitor MK-0626 attenuates pancreatic islet injury in tacrolimus-induced diabetic rats,” PloS one. 9(6):e100798. pp. 1-10.
Johnson et al. (Sep. 5, 2014) “A Potent α/β-Peptide Analogue of GLP-1 with Prolonged Action in Vivo,” Journal of the American Chemical Society. 136(37):12848-12851.
Kwon et al. (Sep. 2014) “Pharmacological evaluation of once-weekly potentials by combination of long-acting insulin with long-acting exendin4 in an animal model,” Diabetologia. 57(Suppl 1):S398-S399. Abstract No. 972.
Li et al. (Apr. 2014) “Vascular protective effect of exendin-4 in experimental models of oxidative stress,” Cytotherapy. 16(4 Suppl):S37-S38. Abstract No. 115.
Li et al. (Nov. 5, 2014) “Exendin-4 promotes endothelial barrier enhancement via PKA-and Epac1-dependent Rac1 activation,” American Journal of Physiology. 308(2):C164-C175.
Lim et al. (Nov. 18, 2014) “Evaluation of PEGylated Exendin-4 Released from Poly (Lactic-co-Glycolic Acid) Microspheres for Antidiabetic Therapy,” Journal of Pharmaceutical Sciences. 104(1):72-80.
Lovshin et al. (Oct. 2014) “Blood pressure-lowering effects of incretin-based diabetes therapies,” Canadian Journal of Diabetes. 38(5)364-71.
Lynch et al. (Jun. 24, 2014) “A novel DPP IV-resistant C-terminally extended glucagon analogue exhibits weight-lowering and diabetes-protective effects in high-fat-fed mice mediated through glucagon and GLP-1 receptor activation,” Diabetologia. 57(9):1927-1936.
Maas et al. (Oct. 2014) “Impact of the mTOR inhibitor Everolimus on peptide receptor radionuclide therapy in a transgenic neuroendocrine tumor mouse model,” European Journal of Nuclear Medicine and Molecular Imaging. 41 (Suppl 2):S529. Abstract No. P593.
Masjkur et al. (Nov. 4, 2014) “Hes3 is Expressed in the Adult Pancreatic Islet and Regulates Gene Expression, Cell Growth, and Insulin Release,” The Journal of Biological Chemistry. 289(51):35503-35516.
Mondragon et al. (Aug. 13, 2014) “Divergent effects of liraglutide, exendin-4, and sitagliptin on beta-cell mass and indicators of pancreatitis in a mouse model of hyperglycaemia,” PloS one. 9(8):e104873. pp. 1-9.
Nagai et al. (Sep. 2014) “Effects of sitagliptin on body fat and intrahepatic lipid content in Japanese overweight patients with type 2 diabetes,” Diabetologia. 57(Suppl 1):S356. Abstract No. 876.
Patel et al. (Sep. 29, 2014) “Cannabinoid receptor 1 antagonist treatment induces glucagon release and shows an additive therapeutic effect with GLP-1 agonist in diet-induced obese mice,” Canadian Journal of Physiology and Pharmacology. 92(12):975-983.
Pathak et al. (Nov. 6, 2014) “Antagonism of gastric inhibitory polypeptide (GIP) by palmitoylation of GIP analogues with N- and C-terminal modifications improves obesity and metabolic control in high fat fed mice”; Molecular and Cellular Endocrinology. 401:120-129.
Pi et al. (2014) “ [Clinical research progresses on glucagon-like peptide-1 analogs in treatment of diabetes mellitus],” [Jianyan Yixue Yu Linchuang]. 11(6):830-832.—with English machine translation.
Qian et al. (Jun. 19, 2014) “Analysis of the interferences in quantitation of a site-specifically PEGylated exendin-4 analog by the Bradford method,” Analytical Biochemistry. 465C:50-52.
Roed et al. (Nov. 22, 2013) “Real-time trafficking and signaling of the glucagon-like peptide-1 receptor,” Mol. Cell Endocrinol. 382(2):938-949.
Russell et al. (Jun. 2014) “The novel GLP-1-GLP-2 dual agonist ZP-GG-72 increases intestinal growth and improves insulin sensitivity in DIO mice,” Diabetes. 63(Suppl 1):A98. Abstract No. 374-OR.
Schattauer Gmbh (Jun. 12, 2014) Meeting Abstracts of the Swiss Society of Radiology and the Swiss Society of Nuclear Medicine 2014. Nuklearmedizin. 53(2):A111-A126.
Tashiro et al. (Jan. 10, 2014) “A glucagon-like peptide-1 analog liraglutide suppresses macrophage foam cell formation and atherosclerosis,” Peptides. 54:19-26.
Tweedie et al. (May 2014) “Exendin-4, a candidate treatment for the clinical management of traumatic brain injury,” Brain Injury. 28(5-6):549-550. Abstract No. 0101.
Vioix et al. (Nov. 2014) “Cost-minimisation analysis of dapagliflozin compared to lixisenatide as an add-on to insulin in the treatment of type 2 diabetes mellitus from a UK health care perspective,” Value in Health. 17(7):A348. Abstract No. PDB95.
Wang et al. (Jun. 2014) “Microfluidic multiplexer perifusion device for studying islet immunotoxicity,” Diabetes. 63 (Suppl 1):A555. Abstract No. 2181-P.
Wu et al. (May 24, 2014) “(64)Cu labeled sarcophagine exendin-4 for microPET imaging of glucagon like peptide-1 receptor expression,” Theranostics. 4(8):770-777.
Xu et al. (Feb. 11, 2014) “Exendin-4 alleviates high glucose-induced rat mesangial cell dysfunction through the AMPK pathway,” Cell. Physiol. Biochem. 33(2):423-432.
Xu et al. (Sep. 2014) “Insulinoma imaging with glucagon-like peptide-1 receptor targeting probe (18)F-FBEM-Cys (39)-exendin-4,” Journal of Cancer Research and Clinical Oncology. 140(9):1479-1488.
Yang et al. (2014) “Design, synthesis and biological evaluation of novel peptide MC62 analogues as potential antihyperglycemic agents,” European Journal of Medicinal Chemistry. 73:105-111.
Yang et al. (Jun. 2014) “Exendin-4, an analogue of glucagon-like peptide-1, attenuates hyperalgesia through serotonergic pathways in rats with neonatal colonic sensitivity,” J. Physiol. Pharmacol. 65(3)349-357.
Yosida et al. (May 13, 2014) “Involvement of cAMP/EPAC/TRPM2 activation in glucose- and incretin-induced insulin secretion,” Diabetes. 63(10):3394-3403.
Zhang et al. (Aug. 2014) “GLP-1 ameliorates the proliferation activity of INS-1 cells inhibited by intermittent high glucose concentrations through the regulation of cyclins,” Molecular Medicine Reports. 10(2):683-688.
Stoessl et al. (2008) “Potential therapeutic targets for Parkinson's disease,” Expert Opinion on Therapeutic Targets. 12(4):425-436.
International Preliminary Report on Patentability corresponding to International Patent Application No. PCT/EP2013/077313, dated Feb. 12, 2015.
International Preliminary Report on Patentability corresponding to International Patent Application No. PCT/EP2014/077336, dated Feb. 26, 2016.
International Preliminary Report on Patentability corresponding to International Patent Application No. PCT/EP2014/077337, dated Jun. 14, 2016.
International Preliminary Report on Patentability corresponding to International Patent Application No. PCT/EP2014/077338, dated Jun. 14, 2016.
International Preliminary Report on Patentability corresponding to International Patent Application No. PCT/EP2014/077339, dated Jun. 14, 2016.
International Preliminary Report on Patentability corresponding to International Patent Application No. PCT/EP2014/077340, dated Jun. 14, 2016.
International Preliminary Report on Patentability corresponding to International Patent Application No. PCT/EP2014/077341, dated Jun. 14, 2016.
International Search Report with Written Opinion corresponding to International Patent Application No. PCT/EP2013/062090, dated Feb. 7, 2014.
International Search Report with Written Opinion corresponding to International Patent Application No. PCT/EP2013/070882, dated Dec. 5, 2013.
International Search Report with Written Opinion corresponding to International Patent Application No. PCT/EP2013/077307, dated Feb. 18, 2014.
International Search Report with Written Opinion corresponding to International Patent Application No. PCT/EP2013/077310, dated Feb. 18, 2014.
International Search Report with Written Opinion corresponding to International Patent Application No. PCT/EP2013/077312, dated Feb. 18, 2014.
International Search Report with Written Opinion corresponding to International Patent Application No. PCT/EP2013/077313, dated Feb. 18, 2014.
International Search Report with Written Opinion corresponding to International Patent Application No. PCT/EP2014/077336, dated Mar. 18, 2015.
International Search Report with Written Opinion corresponding to International Patent Application No. PCT/EP2014/077337, dated Apr. 1, 2015.
International Search Report with Written Opinion corresponding to International Patent Application No. PCT/EP2014/077338, dated Mar. 26, 2015.
International Search Report with Written Opinion corresponding to International Patent Application No. PCT/EP2014/077339, dated May 11, 2015.
International Search Report with Written Opinion corresponding to International Patent Application No. PCT/EP2014/077340, dated Mar. 18, 2015.
International Search Report with Written Opinion corresponding to International Patent Application No. PCT/EP2014/077341, dated Mar. 18, 2015.
International Search Report with Written Opinion corresponding to International Patent Application No. PCT/EP2015/057416, dated Jun. 22, 2015.
International Search Report with Written Opinion corresponding to International Patent Application No. PCT/EP2015/057417, dated Jun. 17, 2015.
International Search Report with Written Opinion corresponding to International Patent Application No. PCT/EP2015/057418, dated Jun. 19, 2015.
International Search Report with Written Opinion corresponding to International Patent Application No. PCT/EP2015/063607, dated Sep. 23, 2015.
International Search Report with Written Opinion corresponding to International Patent Application No. PCT/EP2016/062496, dated Aug. 3, 2016.
International Search Report with Written Opinion corresponding to International Patent Application No. PCT/EP2016/063332, dated Aug. 10, 2016.
International Search Report with Written Opinion corresponding to International Patent Application No. PCT/EP2016/063339, dated Aug. 8, 2016.
Lorenz et al. (2013) “Recent progress and future options in the development of GLP-1 receptor agonists for the reatment of diabesity” Bioorg. Med. Chem. Lett. 23(14):4011-4018.
Lozano et al. (2013) “Polyarginine nanocapsules: a new platform for intracellular drug delivery,” Journal of Nanoparticle Research. 15:1515. pp. 1-14.
Margolis (2004) “Diagnosis of Huntington Disease,” Clin. Chem. 49:1726-1732.
Martin et al. (1998) “Neurodegeneration in excitotoxicity, global cerebral ischemia, and target deprivation: A perspective on the contributions of apoptosis and necrosis,” Brain Res. Bull. 46:281-309.
McLaughlin et al. (2010) “Reversible Hyperinsulinemic Hypoglycemia after Gastric Bypass: A Consequence of Altered Nutrient Delivery,” J. Clin. Endocrinol. Metabol. 95(4):1851-1855.
Medline Plus “Obesity,” National Insitute of Health. Accessible on the Internet at URL: http://www.nlm.nih.gov/medlineplus/obesity.html. [Last Accessed Aug. 22, 2013].
Meier (Sep. 4, 2012) “GLP-1 receptor agonists for individualized treatment of type 2 diabetes mellitus,” Nat. Rev. Endocrinol. 8:728-742.
Meier et al. (May 21, 2015) “Incretin-based therapies: where will we be 50 years from now?” Diabetologia. 58:1745-1750.
Miyawaki et al. (2002) “Inhibition of gastric inhibitory polypeptide signaling prevents obesity,” Nat. Med. 8(7):738-742.
Murage et al. (2008) “Search for alpha-helical propensity in the receptor-bound conformation of glucagon-like peptide-1,” Bioorg. Med. Chem. 16:10106-10112.
Nauck et al. (1993) “Additive insulinotropic effects of exogenous synthetic human gastric inhibitory polypeptide and glucagon-like peptide-1-(7-36) amide infused at near-physiological insulinotropic hormone and glucose concentrations,” J. Clin. Endocrinol. Metab. 76:912-917.
Norris et al. (2009) “Exenatide Efficacy and Safety: A Systematic Review,” Diabetic Medicine. 26:837-846.
Norwegian Institute of Public Health (Dec. 19, 2013) ATC/DDD Index for Cardiovascular System.
Oh et al. (2010) “Target specific and long-acting delivery of protein, peptide, and nucleotide therapeutics using hyaluronic acid derivatives,” Journal of Controlled Release. 141:2-12.
Pan et al. (2006) “Design of a long acting peptide functioning as both a glucagon-like peptide-1 receptor agonist and a glucagon receptor antagonist.” Journal of Biological Chemistry. 281(18):12506-12515.
Pedersen et al. (2006) “N- and C-terminal hydrophobic patches are involved in fibrillation of glucagon,” Biochemistry. 45:14503-14512.
Pocai (2009) “Glucagon-like peptide 1/glucagon receptor dual agonism reverses obesity in mice,” Diabetes. 58 (10):2258-2266.
Pocai (Dec. 14, 2013) “Action and therapeutic potential of oxyntomodulin,” Molecular Metabolism 3:2412-51.
Rentier et al. (Mar. 26, 2015) “Synthesis of diastereomerically pure Lys(Nϵ-lipoyl) building blocks and their use in Fmoc/tBu solid phase synthesis of lipoyl-containing peptides for diagnosis of primary biliary cirrhosis,” Journal of Peptide Science. 21(5):408-414.
Robberecht et al. (1986) “Comparative efficacy of seven synthetic glucagon analogs, modified in position 1, 2 and/or 12, on liver and heart adenylate cyclase from rat,” Peptides. 7(1):109-112.
Rovo et al. (May 2014) “Rational design of a-helix-stabilized exendin-4 analogues,” Biochemistry. 53(22):3540-3552.
Seddon (2004) “Pseudopolymorph: A polemic,” Crystal Growth and Design. 4(6):1087.
Shiau et al. (1998) “The structural basis of estrogen receptor/coactivator recognition and the antagonism of this interaction by tamoxifen,” Cell. 95(7):927-937.
St. John Providence Health System “Preventing Obesity in Children,” St. John Providence Health System. Accessible on the Internet at URL: http://www.stjohnprovidence.org/HealthInfoLib/swarticle.aspx?type=85&id=P07863. [Last Accessed Aug. 22, 2013].
Tasyurek et al. (Jul. 2014) “Incretins: Their physiology and application in the treatment of diabetes mellitus,” Diabetes Metab. Res. Rev. 30(5):354-371.
Ueda et al. (2010) “Identification of glycosylated exendin-4 analogue with prolonged blood glucose-lowering activity through glycosylation scanning substitution,” Bioorg. Med. Chem. Lett. 20(15):4631-4634.
United Healthcare “Diabetes,” United Healthcare. Accessible on the Internet at URL: http://www.uhc.com/source4women/health_topics/diabetes/relatedinformation/d0f0417b073bf110VgnVCM1000002f10b10a.htm. [Last Accessed Aug. 22, 2013].
Unison et al. (1993) “The role of histidine-1 in glucagon action,” Arch. Biochem. Biophys. 300(2):747-750.
Vippagunta et al. (2001) “Crystalline Solids,” Advanced Drug Delivery Reviews. 48:3-26.
Vojkovsky (1995) “Detection of secondary amines on solid phase,” Peptide Research 8:236-237.
Ward et al. (Nov. 2013) “Peptide lipidation stabilizes structure to enhance biological function,” Mol. Metabol. 2 (4):468-479.
World Health Organization (2007) “Prevention of Cardiovascular Disease,” WorldHealth Organization. pp. 1-86.
Yun et al. (Feb. 2012) “Solution Structure of LXXLL-related Cofactor Peptide of Orphan Nuclear Receptor FTZ-F1.” Bulletin of the Korean Chemical Society, 33(2):583-588.
European Search Report corresponding to European Patent Application No. 12172010, dated Apr. 19, 2013.
European Search Report corresponding to European Patent Application No. 12306232, dated Apr. 19, 2013.
European Search Report corresponding to European Patent Application No. 12306647, dated May 22, 2013.
European Search Report corresponding to European Patent Application No. 13306712, dated May 27, 2014.
European Search Report corresponding to European Patent Application No. 13306713, dated Jun. 12, 2014.
European Search Report corresponding to European Patent Application No. 13306714, dated May 28, 2014.
European Search Report corresponding to European Patent Application No. 13306715, dated Jun. 12, 2014.
European Search Report corresponding to European Patent Application No. 13306716, dated May 27, 2014.
European Search Report corresponding to European Patent Application No. 13306717, dated Jun. 3, 2014.
European Search Report corresponding to European Patent Application No. 13305222, dated Jul. 15, 2013.
European Search Report corresponding to European Patent Application No. 14305501, dated Sep. 23, 2014.
European Search Report corresponding to European Patent Application No. 14305503, dated Sep. 23, 2014.
International Preliminary Report on Patentability corresponding to International Patent Application No. PCT/EP2013/062090, dated Nov. 24, 2014.
International Preliminary Report on Patentability corresponding to International Patent Application No. PCT/EP2013/070882, dated Dec. 1, 2014.
International Preliminary Report on Patentability corresponding to International Patent Application No. PCT/EP2013/077307, dated Feb. 12, 2015.
International Preliminary Report on Patentability corresponding to International Patent Application No. PCT/EP2013/077310, dated Feb. 2, 2015.
International Preliminary Report on Patentability corresponding to International Patent Application No. PCT/EP2013/077312, dated Feb. 13, 2015.
Related Publications (1)
Number Date Country
20180289822 A1 Oct 2018 US