throbber
Bioorganic & Medicinal Chemistry Letters 23 (2013) 4011–4018
`
`Contents lists available at SciVerse ScienceDirect
`
`Bioorganic & Medicinal Chemistry Letters
`
`j o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m / l o c a t e / b m c l
`
`BMCL Digest
`Recent progress and future options in the development of GLP-1
`receptor agonists for the treatment of diabesity
`
`Martin Lorenz a,⇑, Andreas Evers b, Michael Wagner a,⇑
`
`a Diabetes Division/Res. & Transl. Med, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, 65926 Frankfurt am Main, Germany
`b LGCR/Struct., Design & Informatics, Sanofi Deutschland GmbH, 65926 Frankfurt am Main, Germany
`
`a r t i c l e
`
`i n f o
`
`a b s t r a c t
`
`Article history:
`Received 16 March 2013
`Revised 6 May 2013
`Accepted 7 May 2013
`Available online 16 May 2013
`
`Keywords:
`GLP-1 receptor agonists
`Gastro-intestinal hormones
`Diabesity
`Delivery technologies
`
`The dramatic rise of the twin epidemics, type 2 diabetes and obesity is associated with increased mortal-
`ity and morbidity worldwide. Based on this global development there is clinical need for anti-diabetic
`therapies with accompanied weight reduction. From the approved therapies, the injectable glucagon-like
`peptide-1 receptor agonists (GLP-1 RAs) are the only class of agents which are associated with a modest
`weight reduction. Physiological effects of the gastro-intestinal hormone GLP-1 are improvement of gly-
`cemic control as well as a reduction in appetite and food intake. Different approaches are currently under
`clinical evaluation to optimize the therapeutic potential of GLP-1 RAs directed to once-weekly up to once-
`monthly administration. The next generation of peptidic co-agonists comprises the activity of GLP-1 plus
`additional gastro-intestinal hormones with the potential for increased therapeutic benefits compared to
`GLP-1 RAs.
`
`Ó 2013 Elsevier Ltd. All rights reserved.
`
`Introduction. Type 2 diabetes mellitus (T2DM) is a progressive
`chronic disease characterized by hyperglycemia due to defective
`insulin secretion and resistance to insulin action. The disease is
`associated with morbidity and mortality in patients, and is a lead-
`ing cause for cardiovascular (CV) disease, renal failure, blindness,
`amputations and hospitalizations.1,2 T2DM has recently reached
`epidemic proportions in both developed and developing countries
`in conjunction with a substantial increase in obesity.3 Up to 80% of
`people with T2DM are overweight or obese, whereas obesity is
`considered as a major risk factor for T2DM.4 ‘Diabesity’ is a new
`term to characterize this phenomenon of obesity-dependent diabe-
`tes associated with multiple comorbidities (mainly CV disease) and
`in addition describes a rising epidemic.5 An effective approach to
`the management of diabesity is a modest reduction in body weight
`(4–5 kg) resulting in highly beneficial effects on glycemic control
`as well as reduced morbidity and mortality.4 According to the new
`guidelines of the American Diabetes Association (ADA) and the
`European Association for the Study of Diabetes (EASD) out of avail-
`able classes of glucose-lowering agents, the only class associated
`with modest body weight reduction are peptidic glucagon-like
`peptide-1 receptor agonists (GLP-1 RAs). All other agents are either
`weight neutral (Metformin, DPP4-inhibitors) or associated with
`weight gain (basal insulin, thiazolidinediones, sulfonylureas).6
`
`⇑ Corresponding authors. Tel.: +49 69 305 46875 (M.W.); tel.: +49 69 305 12041
`
`(M.L.).
`E-mail addresses: Martin.Lorenz@sanofi.com (M. Lorenz), Andreas.Evers@sanofi.
`com (A. Evers), Michael.Wagner@sanofi.com (M. Wagner).
`
`0960-894X/$ - see front matter Ó 2013 Elsevier Ltd. All rights reserved.
`http://dx.doi.org/10.1016/j.bmcl.2013.05.022
`
`Therapy with peptidic GLP-1 RAs is based on self-administra-
`tion by subcutaneous injection using a pen device (or a syringe).
`The following GLP-1 RAs are approved for the treatment of diabe-
`tes: twice-daily exenatide BID (ByettaÒ, BMS), once-daily lixisena-
`tide (LyxumiaÒ, Sanofi/Zealand), once-daily liraglutide (VictozaÒ,
`Novo Nordisk) and once-weekly exenatide LAR (BydureonÒ, BMS).
`Current optimization of peptidic GLP-1 RAs aims to improve
`efficacy, tolerability and compliance such as frequency and ease
`of administration. Furthermore, significant efforts are currently
`undertaken to identify therapeutic peptide approaches with the
`potential for greater body weight reduction compared to existing
`GLP-1 RAs. The present article reviews key data on marketed and
`novel long-acting GLP-1 RAs currently in late clinical development,
`describes novel techniques suitable for once-weekly up to once-
`monthly administration and discusses potential novel peptide ap-
`proaches based on further gut hormones. However, recent research
`activities have resulted also in peptidic and small molecule GLP-1
`RAs which might be suited for oral application7–among those
`TTP054 from Transtech Pharma as most advanced in phase 2.8
`Those still early developments in the field of orally available
`GLP-1 RAs are not covered by this review.
`GLP-1 RAs: overview. Natural human glucagon-like peptide-1
`(GLP-1) is an incretin hormone derived from the transcription
`product of the proglucagon gene. It is produced by intestinal L-cells
`and released in response to meal intake to induce insulin secretion
`from pancreatic b-cells. The biologically active forms of GLP-1 are
`GLP-1(7–37) and GLP-1(7–36)NH2, which exert their action by
`activation of the GLP-1 receptor, a class B G-protein-coupled
`receptor. GLP-1 shows a helical character in solution and in the
`
`MPI EXHIBIT 1139 PAGE 1
`
`

`

`4012
`
`M. Lorenz et al. / Bioorg. Med. Chem. Lett. 23 (2013) 4011–4018
`
`receptor-bound conformation (see Fig. 1).9,10 Exendin-4 is another
`potent peptidic GLP-1 RA, which was isolated from the venom of
`the Gila monster. Although it shares only 53% sequence identity
`with native GLP-1, both peptides show a helical character with
`many of the conserved amino acids facing the interaction site of
`the receptor (see Fig. 1).11 All GLP-1 RAs on the market or in clinical
`development are either derived from the natural GLP-1 or exendin-
`4 peptide. Based on their PK-profile, marketed GLP-1 RAs exenatide
`BID and lixisenatide could be classified as shorter-acting agents
`compared with longer-acting agents liraglutide and exenatide LAR.
`GLP-1 and GLP-1 RAs have 3 major pharmacological activi-
`ties12,13 to improve glycemic control in patients with T2DM by
`reducing fasting and postprandial glucose (FPG and PPG): (i) in-
`creased glucose-dependent insulin secretion (improved first- and
`second-phase), (ii) glucagon suppressing activity under hypergly-
`cemic conditions, (iii) delay of gastric emptying rate resulting in re-
`tarded absorption of meal-derived glucose. Treatment with GLP-1
`RAs for 12–52 weeks leads to reduction in glycosylated hemoglo-
`bin (HbA1c), a diagnostic marker reflecting the average glucose
`levels of the last 2–3 months (and serving as primary efficacy end-
`point in phase 2b–3 studies) of 1.1–1.6% due to reduced postpran-
`dial and fasting blood glucose.14 Unlike insulin or
`insulin
`secretagogues (sulfonylureas) the insulinotropic effect of GLP-1
`RAs is strictly glucose-dependent resulting in a generally low risk
`for hypoglycemia. Beyond anti-hyperglycemic actions of GLP-1
`RAs, central and possibly peripheral effects (e.g. vagal afferents)
`which modulate feelings of appetite and satiety are considered to
`reduce food intake and finally account for a moderate body weight
`reduction in the range of 1–3 kg.15
`In addition, beneficial reduction in systolic and diastolic blood
`pressure as well as reduced cholesterol plasma levels were ob-
`served in clinical trials.16 Preclinical data illustrate different cardio-
`protective effects of GLP-1 RAs (e.g. on cardiomyocytes, blood
`vessels, adipocytes and lipids).17 These effects might impair the on-
`set or progression of atherosclerotic disease. Furthermore, animal
`and initial clinical studies have shown that GLP-1 RAs could reduce
`ischemia–reperfusion injury. However, treatment with longer-
`
`acting GLP-1 RAs (liraglutide and exenatide LAR) is accompanied
`in humans by a slight increase in heart rate of 2–4 beats per min-
`ute. Ongoing cardiovascular outcome studies for the marketed
`agents will reveal if the potential harm of an increased heart rate
`will be outweighed by the reported cardiovascular benefits.17
`The most common side effects of GLP-RAs are nausea, vomiting
`and diarrhea, which are mostly transient and can be at least partly
`reduced by gradual uptitration of the dose. Beyond gastro-intesti-
`nal side effects an association of long-term GLP-1RA therapy with
`an increased risk for pancreatitis is under discussion. Some rare
`cases of acute pancreatitis have been reported as postmarketing
`surveillance activities for GLP-1 RAs. It is not clear if such cases
`are therapy-related or due to a general 2–3-fold increased risk
`for acute pancreatitis associated with T2DM.18 In rodents malig-
`nant thyroid C-cell carcinomas were observed following treatment
`with liraglutide. The human relevance of these findings in rodents
`could not be determined by clinical or nonclinical studies.18
`As peptide based drugs, GLP-1 RAs have the potential to induce
`an immune response and antibody formation was reported for all
`marketed GLP-1 RAs but does not appear to impact efficacy or
`safety of these agents. The level of glycemic control (HbA1c) is
`overall reported similar regardless of the antibody status, although
`for a rare number of patients high-titer antibody formation might
`be associated with attenuated glycemic response (reported for 1–
`4% of total patients treated with exenatide BID).19 There seems to
`be slightly increased antibody development and injection site reac-
`tions with exendin-4 compared to the GLP-1 analog liraglutide,20
`presumably due to the greater differences in sequence identity
`compared to native GLP-1.
`A differentiating characteristic within the GLP-1 RA class impor-
`tant for glycemic control is the propensity to slow gastric empty-
`ing, which is dependent on the pharmacokinetics of these agents.
`For longer-acting agents such as liraglutide and exenatide LAR, de-
`layed gastric emptying fades following the first dose probably
`reflecting tachyphylaxis.21,22 These agents strongly reduce fasting
`blood glucose probably via continuous increased insulin levels in
`the fasting state and cause an apparently better efficacy than
`
`Figure 1. Binding hypotheses of GLP-1 RAs at the GLP-1R. Amino acid changes versus native GLP-1 are shown in orange. Binding mode of GLP-1 from RCSB PDB (PDB ID 3iol),
`binding modes of the remaining peptides are computational models.
`
`MPI EXHIBIT 1139 PAGE 2
`
`

`

`M. Lorenz et al. / Bioorg. Med. Chem. Lett. 23 (2013) 4011–4018
`
`4013
`
`shorter-acting GLP-1 RAs.14 In contrast, the shorter-acting agents,
`exenatide BID and lixisenatide are both proven to strongly slow
`gastric emptying in patients with T2DM without any signs for
`desensitization with continuous treatment.23,24 As consequence,
`shorter-acting GLP-1 RAs are associated with a pronounced reduc-
`tion in PPG. As HbA1c levels decrease under therapy, the contribu-
`tion of postprandial glucose to HbA1c increases over that of fasting
`plasma glucose.25 For patients with improved HbA1c under ther-
`apy (including those on insulin treatment), who have a greater
`dependency on postprandial glucose, shorter-acting agents may
`be the better choice. This greater PPG-lowering effect of shorter-
`acting GLP-1 RAs would be complementary to the glucose lowering
`effects of basal insulin, which primarily targets fasting glucose.
`Hence, a combination therapy of shorter-acting GLP-1 RAs with ba-
`sal insulin may offer the advantage to lower basal insulin require-
`ments and result in beneficial weight loss effects.26
`GLP-1 agonists on the market and in clinical development. Thera-
`peutic utility of natural GLP-1 is limited by its rapid degradation by
`serum proteases, predominantly dipeptidyl peptidase IV (DPPIV),
`but also other enzymes such as neutral endopeptidase (NEP), plas-
`ma kallikrein or plasmin.27 Therefore, GLP-1 has a very short half-
`life of 2 min following intravenous administration. A variety of
`strategies have been applied to provide novel GLP-1 RAs with
`longer-term in vivo activity following subcutaneous administra-
`tion, which are described below.
`Peptides stabilized against DPPIV by amino acid exchange. One
`strategy to prolong in vivo half-life is stabilization towards degra-
`dation by DPPIV, which preferably cleaves N-terminal Xaa–Pro or
`Xaa–Ala dipeptide sequences. Alteration of that N-terminal se-
`quence, especially the second amino acid, has proven to reduce
`degradation by DPPIV.28,29
`Exenatide BID. In exendin-4, the second amino acid is a Gly ren-
`dering it resistant to DPPIV mediated degradation. Furthermore,
`the Leu21–Ser39 span of exendin-4 forms a compact tertiary fold
`(the Trp-cage) which shields the side chain of Trp25 from solvent
`exposure, leading to enhanced helicity and stability of the peptide
`(see Fig. 1).9 Exenatide BID (Amylin, now BMS), a synthetic version
`of exendin-4, represents the first GLP-1 RA approved in 2005 as
`antidiabetic therapy for the treatment of T2DM. It has a terminal
`half-life of 2.4 h after subcutaneous administration and is applied
`twice daily (10 lg). With this dosing regimen as monotherapy or in
`combination with oral antidiabetics, HbA1c reductions of 0.8–0.9%
`are typically observed accompanied by a body weight reduction in
`the range of 1.8–2.6 kg following a treatment duration of 30 weeks.
`In an open label extension the combination of exenatide BID and
`metformin showed a HbA1c reduction of 1.1% and a body weight
`reduction of 4.4 kg after 82 weeks.30 Exenatide BID and insulin
`glargine did not demonstrate a significant difference in HbA1c
`reduction (1.25% vs 1.26%) after 26 weeks treatment. However,
`insulin glargine resulted in a weight gain of 3 kg compared to a
`reduction of 2.7 kg seen with exenatide BID.31
`Lixisenatide. Lixisenatide, developed by Sanofi/Zealand, is a syn-
`thetic analog of exendin-4 (Fig. 1). Compared to exendin-4, six Lys
`residues have been added to the C-terminus (also amidated), while
`one Pro in the C-terminal region has been deleted. Lixisenatide has
`a mean terminal half-life of approximately 3 h in humans. In a
`dose-ranging study in patients with T2DM, inadequately con-
`trolled with metformin, lixisenatide at a dose of 20 lg once-daily
`demonstrated the best efficacy-to-tolerability ratio.32 Lixisenatide
`was applied as once-daily administration (20 lg) in phase 3 trials
`and has demonstrated efficacy as monotherapy, in combination
`with oral antidiabetic drugs, and as add-on to basal insulin, with
`particular efficacy in reducing postprandial glucose excursion
`(HbA1c reduction up to 0.9%). In combination with oral antidia-
`betic drugs, lixisenatide resulted in a sustained body weight reduc-
`tion from baseline in controlled studies (24 weeks) in a range from
`
`1.8–3 kg. Comparison of lixisenatide with exenatide BID as add-on
`to metformin demonstrated noninferior improvements in HbA1c,
`but with less hypoglycemia, slightly less weight loss and a more
`favorable gastro-intestinal tolerability profile after 24 weeks.33 Lix-
`isenatide significantly delays gastric emptying, a process which is
`accompanied by strong post-prandial glucose lowering.34 These
`attributes are very attractive with regard to a combination therapy
`with basal insulins, for example, insulin glargine, where Lixisena-
`tide demonstrated significantly reduced HbA1C (0.7–0.8%) and
`body weight (1.8 kg).34 In February 2013, the EMA granted market-
`ing authorization in Europe for lixisenatide for the treatment of
`adults with T2DM.35
`Sustained release of DPPIV stabilized peptides. DPPIV-resistant
`peptidic GLP-1 RAs are still subjected to renal clearance as illus-
`trated by the rather moderate half-lives of exenatide and lixisena-
`tide. Therefore, several approaches have been explored to further
`prolong the duration of action by either creating a depot (e.g. in
`the subcutaneous space), from which the peptide is slowly re-
`leased, or by conjugating peptidic GLP-1 agonists to carrier mole-
`cules in order to reduce renal clearance.
`Taspoglutide. Taspoglutide developed by Roche (in collaboration
`with Ipsen) is a close analog of natural GLP-1(7–36) in which the
`unnatural amino acid aminoisobutyric acid (Aib) has been intro-
`duced in position 8 and 35 in order to avoid degradation by DPPIV,
`but also by other serine proteases such as plasma kallikrein and
`plasmin.36 NMR-studies with taspoglutide showed a similar sec-
`ondary structure compared to native GLP-1, but clearly an in-
`creased a-helicity in the C-terminal part of
`the peptide.
`Taspoglutide was developed as a sustained release formulation
`containing zinc chloride suitable for once-weekly administration.
`When injected into the subcutaneous tissue, taspoglutide precipi-
`tates and forms a depot. Taspoglutide was studied in several phase
`3 studies with a 10 and 20 mg once-weekly dosing regimen, and
`showed consistent HbA1c and weight reductions.37–41 Significant
`weight reductions were only observed with the 20 mg dose.42
`Once-weekly taspoglutide was superior to twice-daily exenatide
`with respect to glycemic control, while resulting in a similar
`weight loss after treatment for 24 and 52 weeks. The overall safety
`profile comprising gastro-intestinal tolerability, systemic allergic
`reactions and injection-site reactions was clearly worse, especially
`the gastro-intestinal side effects, which resulted in a roughly dou-
`bled discontinuation rate in patients treated with taspoglutide
`compared to exenatide (34 vs 16%). Based on this phase 3 data
`Roche decided to stop the development of taspoglutide.43
`Exenatide LAR–Bydureon. Further strategies to create a subcuta-
`neous depot from which the peptidic GLP-1 RA of choice can be
`slowly released utilize polymeric matrices. Exenatide LAR (devel-
`oped by Amylin/Lilly/Alkermes–now BMS) is a once-weekly for-
`mulation of exenatide,
`in which exenatide is noncovalently
`entrapped into a biodegradable polymeric matrix consisting of
`poly(D,L-lactide-co-glycolide) (PLG) forming microspheres.44 Slow
`release from the polymeric matrix takes place through diffusion
`and microsphere breakdown. Exenatide LAR has a half-life in hu-
`mans of 5–6 days. After a 2 mg s.c. injection, steady state plasma
`levels of exendin-4 are typically obtained after 6–10 weeks.13 At
`that dose HbA1c reductions of 1.3–1.9% were observed. A direct
`comparison with exenatide BID revealed a better reduction in
`HbA1c (1.9 vs 1.5%) with a similar reduction in body weight (3.6
`vs 3.7 kg).45 Different from this efficacy data, a head-to-head com-
`parison with liraglutide showed an improved glycemic control fol-
`lowing 26 weeks of treatment associated with increased weight
`reduction in favor of liraglutide (HbA1c reduction 1.5% vs 1.3%,
`body weight reduction 3.6 kg vs 2.7 kg).46 One drawback of exena-
`tide LAR is the relatively large needle size (23 gauge), which is used
`for administration due to the viscosity of the polymeric suspen-
`sion, as well as the quite laborious preparation prior to injection.
`
`MPI EXHIBIT 1139 PAGE 3
`
`

`

`4014
`
`M. Lorenz et al. / Bioorg. Med. Chem. Lett. 23 (2013) 4011–4018
`
`Fatty acid conjugation. The concept of fatty acid conjugation is a
`well established strategy to prolong the action of peptides by facil-
`itating binding to serum albumin thereby reducing their renal
`clearance. Landmark achievements in this area have been the
`development of long acting insulin analogs.47,48
`Liraglutide. Liraglutide (VictozaÒ) developed by Novo Nordisk is
`a close structural homolog to GLP-1(7–37) with 97% sequence
`identity to the native hormone. Lys in position 34 is substituted
`by Arg and a palmitic acid is conjugated to Lys in position 26 via
`a glutamate spacer (see Fig. 2a).20 This combination of spacer
`and fatty acid length turned out to be optimal
`in terms of
`in vitro activity and in vivo prolonged duration of action in
`pigs.49,50 The mechanisms for prolongation of action are manifold.
`Following subcutaneous injection, the peptide is slowly released
`from the injection site due to self-association.51 Once it enters
`the bloodstream, liraglutide is extensively bound to serum albu-
`min (99%), which leads to an increased enzymatic stability to-
`wards DPPIV and NEP while reducing renal clearance. The plasma
`half-life in humans is 11–13 h.20 Liraglutide was approved in 2009
`for the treatment of T2DM. The therapeutic standard dose is 1.2 mg
`once-daily with possible increase to 1.8 mg to further improve gly-
`cemic control. The HbA1c reductions observed with these dosing
`regimens in phase 3 trials were 1.1–1.8% accompanied by a weight
`loss of around 2–3 kg (26 week treatment).52 In a direct compari-
`son liraglutide once-daily showed significant improvements in gly-
`cemic control compared with exenatide twice-daily (HbA1c
`reduction of 1.12% vs 0.79%).53 Using higher doses (3 mg) liraglu-
`tide is also under development (phase 3) for the treatment of obes-
`ity54 as well as of obese patients with T2DM.55 In a phase 3a study
`liraglutide 3 mg treatment resulted in 6% weight reduction in
`overweight to obese people compared to placebo.56 Among the
`class of GLP-1 RAs, liraglutide currently seems to be the most effec-
`tive agent for the treatment of obesity.
`Semaglutide. Semaglutide is a next generation GLP-1 analog
`from Novo Nordisk, currently in phase 3 clinical development for
`
`T2DM as a once-weekly injection. Semaglutide’s structure is based
`on liraglutide, with two further modifications: Gly in position 8 is
`replaced by Aib, a beneficial modification already applied in taspo-
`glutide. Furthermore, the fatty acid side chain has been modified
`towards a N6-[N-(17-carboxy-1-oxoheptadecyl-L-c-glutamyl[2-
`(2-aminoethoxy)ethoxy]acetyl[2-(2-aminoethoxy)ethoxy]acetyl]
`residue (see Fig. 2a). The human half-life is 160 h. In a 12 week
`phase 2 trial in T2DM patients, semaglutide was tested at 5 doses
`(0.1–1.6 mg) once-weekly. Semaglutide P0.2 mg dose depen-
`dently reduced HbA1c from baseline up to 1.7% (vs 0.5% reduction
`for placebo) and for doses P0.8 mg also body weight by up to
`4.8 kg (vs 1.2 kg reduction for placebo).57
`Conjugation to albumin. Another half-life prolonging principle is
`the (genetic) fusion to recombinant albumin. Human serum albu-
`min (HSA) has a molecular weight of 67 kDa.58 The half-life of
`albumin in humans is 19 days. PH-dependent recycling mediated
`by the neonatal FC receptor (FcRN) has been shown to contribute
`to that long half-life.59 When albumin is fused to therapeutic pep-
`tides such as GLP-1 RAs, both FcRN mediated recycling as well as
`reduced clearance due to the increased molecular weight are
`responsible for half-life prolongation.
`Albiglutide. In albiglutide, developed by GlaxoSmithKline (GSK),
`two copies of GLP-1 are fused as tandem repeat to the N-terminus
`of albumin. DPPIV-resistance is achieved by a single substitution,
`Ala for Gly, at the DPPIV cleavage site (see Fig. 2b). The tandem re-
`peat unit was developed to overcome reduced potency as was seen
`when only one GLP-1 moiety was directly fused to albumin, mean-
`ing that one GLP-1 moiety of the tandem repeat serves as a spacer
`to allow potent binding to the GLP-1 receptor.60 Albiglutide has a
`half-life of 6–8 days in humans and is currently in phase 3 for
`the treatment of T2DM as once-weekly injection. In a 32-week
`head-to-head study comparing albiglutide (50 mg) to once-daily
`liraglutide (1.8 mg) (HARMONY 7), albiglutide demonstrated a sta-
`tistically significant reduction in HbA1c (0.78%) from baseline.61
`The rather moderate body weight loss of 0.6 kg seen for
`
`Figure 2. Approaches for half-life extension of GLP-1 agonists. (a) Liraglutide and semaglutide carry fatty acids, which facilitate binding to serum albumin thereby reducing
`their renal clearance. (b) In albiglutide, two copies of modified GLP-1 are fused as tandem repeat to the N-terminus of albumin. (c) Similar to albumin fusion, peptides can be
`linked to the Fc region of immunoglobulin G (IgG) as applied in dulaglutide.
`
`MPI EXHIBIT 1139 PAGE 4
`
`

`

`M. Lorenz et al. / Bioorg. Med. Chem. Lett. 23 (2013) 4011–4018
`
`4015
`
`albiglutide is thought to be linked to limited actions in the CNS due
`to its large molecular size.13 A regulatory application for albiglu-
`tide was submitted in the US in January 2013.62
`CJC-1134-PC. Likewise, in ConjuChem’s CJC-1134-PC, exendin-4
`is coupled via its C-terminus to albumin by a chemical linker bear-
`ing a terminal maleimide which is used for the chemical conjuga-
`tion to a single cystein residue on albumin (see Fig. 2b).63 CJC-
`1134-PC has a half-life of 8 days in humans and is currently in
`phase 2 trials for the treatment of T2DM as once-weekly injection.
`CJC-1134-PC seems to be more potent compared to albiglutide. In
`phase 2 trials the once-weekly application of 2 mg led to a HbA1c
`reduction of 1.4%. As observed for albiglutide, the effects on weight
`were only moderate.64
`FC fusion. Similar to albumin fusion, peptides can be linked to
`the constant region of immunoglobulin G (IgG), the Fc region
`(see Fig. 2c).65 The Fc region of IgG has a half-life of 22 days.66
`Likewise, when fusing peptides to the FC region of IgG the protrac-
`tion principle is based on reduced renal clearance and FcRN med-
`iated receptor recycling.
`Dulaglutide. Dulaglutide (Eli Lilly) is a recombinant fusion pro-
`tein, which consists of two GLP-1 peptides covalently linked by a
`small peptide [tetraglycyl-L-seryltetraglycyl-L-seryltetraglycyl-L-
`seryl-L-alanyl] to a human IgG4-Fc heavy chain variant. Compared
`to natural GLP-1, the GLP-1 moieties contain amino acid substitu-
`tions (Ala8?Gly, Gly26?Glu, Arg36?Gly) to ensure protection
`from DPPIV cleavage as well as maintenance of the potency of
`the construct. Dulaglutide has a half-life of 4 days and is in ad-
`vanced clinical trials as once-weekly injection for the treatment
`of T2DM.67 In phase 2 clinical trials dulaglutide showed significant
`dose-dependent reduction in HbA1c (1.5% for the 1.5 mg dose after
`12 weeks) and dose-dependent reductions in body weight. How-
`ever, the decrease in body weight was not statistically significant
`when compared to the placebo group.68 Currently, a large phase
`3 program (AWARD studies Nos. 1–7) is ongoing, in which dulaglu-
`tide has shown a superior glycemic control compared to exenatide
`BID after 6 month of treatment.69 Detailed reports regarding the
`efficacy in weight loss in longer studies are not yet available.
`Langlenatide (HM11260C). In langlenatide, developed by Hanmi
`Pharmaceuticals, an exendin-4 analog is fused via a short polyethyl-
`eneglycol (PEG) linker to a nonglycosylated Fc region (‘LAPS-carrier’).
`In contrast to dulaglutide only one peptide copy is fused to the Fc-car-
`rier. Such monomeric peptide-Fc-fusion constructs hold promise to
`show even higher in vivo efficacy and longer duration of action. In
`fact, the half-life of langlenatide has been reported to be 150 h.70
`Langlenatide is currently being investigated in phase 2 trials for the
`treatment of T2DM as once-weekly injection (1–4 mg doses), but also
`for its suitability as once-monthly application (8–16 mg).71
`Peptides coupled to biological polymers–Xtenylation. Xtenylation,
`which is the genetic fusion of an unstructured recombinant poly-
`peptide to a peptide or protein, is a further generic approach to ex-
`tend plasma half-life of peptides, again by reducing renal
`clearance. The Xten protein is a nonrepetitive amino acid polymer,
`which comprises 864 amino acids selected from Ser, Ala, Pro, Thr,
`Glu and Gly, with a huge hydrodynamic volume and good serum
`stability.72 In contrast to polyethyleneglycol (PEG), Xten is highly
`biodegradable which might be beneficial with respect to safety.
`VRS-859: In VRS-859, Xten is genetically fused to exenatide. The
`product shows a half-life in cynomolgous monkeys of 3 days.
`VRS-859 is currently being developed by Diartis Pharmaceuticals
`as once-monthly treatment for patients with T2DM. In October
`2012, phase 1 results in T2DM patients were disclosed showing
`statistically significant reductions in HbA1c levels at 30 days after
`only one 200 mg dose of VRS-859, supporting the once-monthly
`dosing regimen.73
`Novel approaches and combination of multiple hormone actions.
`Approximately 70–80% of severely obese patients with T2DM
`
`who undergo bariatric surgery show significant and fast improve-
`ment of glycemic control even up to a complete remission of dia-
`betes, often resulting in discontinuation of insulin or oral anti-
`diabetic therapy.74 The mechanism seems partly independent of
`weight reduction and related to alterations in the release of gas-
`tro-intestinal hormones, for example increased plasma levels of
`GLP-1, oxyntomodulin and peptide YY (PYY). In addition to the ef-
`fects of GLP-1, these peptides are known to be involved in the reg-
`ulation of glucose homeostasis,
`food intake
`and energy
`expenditure.75 Based on the impressive improvements of T2DM
`after bariatric surgery, trying to mimic some alterations in plasma
`levels of these peptide hormones is a highly attractive research ef-
`fort to identify novel therapeutic options for obese patients with
`T2DM. Combining dual or triple agonistic effects in a single peptide
`(feasible if peptides are sufficiently similar) or combining 2 distinct
`peptides in a formulation approach are possible options. Slow re-
`lease formulations/delivery approaches as mentioned above could
`prolong the duration of action from once-daily up to a once-weekly
`administration regimens.
`Below we discuss promising combinations of peptides, which
`have the potential for improved glycemic control associated with
`more effective weight reduction compared to pure GLP-1 RAs.
`GLP-1/glucagon co-agonists. Oxyntomodulin is a 37 amino acid
`peptide (comprising the entire 29 amino acid sequence from gluca-
`gon plus a C-terminal extension of 8 residues) secreted by intesti-
`nal L-cells (together with GLP-1 and PYY) following meal ingestion.
`The dual agonistic activity of oxyntomodulin is weaker for the GLP-
`1 and glucagon receptors when compared to the cognate native li-
`gands GLP-1 and glucagon.76 Originally glucagon was discovered as
`a hormone with effects counter to those of insulin, raising blood
`glucose levels by stimulating gluconeogenesis and glycogenolysis
`to circumvent a hypoglycemic state. However, more recent data
`in rodents and humans reveal that glucagon could have beneficial
`effects on energy balance, body fat mass and nutrient intake. Those
`effects seem to be mediated at least in part by FGF21-dependent
`pathways.77,78
`In overweight and obese people native oxyntomodulin was
`shown to significantly reduce body weight by 1.7 kg vs. placebo
`following three-times subcutaneous administration (to compen-
`sate for the short half-life of the native peptide) for 4 weeks.79
`Furthermore, oxyntomodulin was mechanistically proven to re-
`duce food intake after an ad libitum test meal and increase energy
`expenditure in humans.80 Thus, combining the actions of GLP-1
`and glucagon in one molecule like in oxyntomodulin might lead
`to a therapeutic principle with anti-diabetic action related to the
`GLP-1 component and a pronounced weight lowering effect related
`to glucagon receptor stimulation. Pioneering work in the field was
`carried out by Bloom and co-workers,81 DiMarchi and co-workers82
`and Merck Research Laboratories.83 Chemical strategies in order to
`identify such GLP-1/glucagon co-agonists started either from the
`sequence of oxyntomodulin or glucagon by carefully incorporating
`amino acids known to be relevant for GLP-1 agonism but also ele-
`ments to prolong half-life. A Tschöp, model peptide is shown in
`Figure 3, which is a hybrid peptide derived from native GLP-1
`and glucagon. Further chemical modifications to increase the sta-
`bility and in vivo half-life include a lactam bond between the
`side-chains of Glu in position 16 and Lys in position 20 (‘stapling’)
`and the addition of a linear 40 kDa PEG chain to Cys in position 24.
`For the design of novel stabilized peptides the selection of the
`activity-ratio for the GLP-1 and the glucagon receptor is important
`for adjusting the right balance between anti-diabetic and anti-
`obesity effects. High potency at the glucagon receptor can be
`anticipated to strongly increase weight loss but potentially at the
`expense of elevated glucose levels. Data in mice suggest that a
`balanced ratio of GLP-1/glucagon-receptor activation (1:1) is
`optimal to ensure glucose control associated with improved
`
`MPI EXHIBIT 1139 PAGE 5
`
`

`

`4016
`
`M. Lorenz et al. / Bioorg. Med. Chem. Lett. 23 (2013) 4011–4018
`
`Figure 3. Amino acid sequence of native GLP-1, native glucagon and

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket