throbber
Diabetologia (1998) 41: 271±278
`
`Ó Springer-Verlag 1998
`
`Dipeptidyl peptidase IV resistant analogues of glucagon-like
`peptide-1 which have extended metabolic stability and improved
`biological activity
`
`C. F. Deacon1, L. B. Knudsen2, K. Madsen2, F. C. Wiberg2, O. Jacobsen1, J. J. Holst1
`
`1 Department of Medical Physiology, The Panum Institute, University of Copenhagen, Denmark
`2 Novo Nordisk A/S, MaÊ lùv, Denmark
`
`Summary Glucagon-like peptide 1 (GLP-1) has great
`potential in diabetes therapy due to its glucose-de-
`pendent stimulation of insulin secretion, but this is
`limited by its rapid degradation, primarily by dipepti-
`dyl peptidase IV. Four analogues, N-terminally sub-
`stituted with threonine, glycine, serine or a-ami-
`noisobutyric acid, were synthesised and tested for
`metabolic stability. All were more resistant to dipep-
`tidyl peptidase IV in porcine plasma in vitro, ranging
`from a t1/2 of 159 min (Gly8 analogue) to undetectable
`degradation after 6 h (Aib8 analogue; t1/2 for GLP-1
`(7±36) amide, 28 min). During i. v. infusion in anaes-
`thetised pigs, over 50 % of each analogue remained
`undegraded compared to 22.7 % for GLP-1 (7±
`36) amide. In vivo, analogues had longer N-terminal
`t1/2 (intact peptides: means, 3.3±3.9 min) than GLP-1
`(7±36) amide (0.9 min; p < 0.01), but these did not ex-
`ceed the C-terminal t1/2 (intact plus metabolite: ana-
`logues, 3.5±4.4 min; GLP-1 (7±36) amide, 4.1 min).
`Analogues were assessed for receptor binding using
`
`a cell line expressing the cloned receptor, and for
`ability to stimulate insulin or inhibit glucagon secre-
`tion from the isolated perfused porcine pancreas. All
`bound to the receptor, but only the Aib8 and Gly8 an-
`alogues had similar affinities to GLP-1 (7±36) amide
`(IC50; Aib8 = 0.45 nmol/l; Gly8 = 2.8 nmol/l; GLP-1
`(7±36) amide = 0.78 nmol/l). All analogues were ac-
`tive in the isolated pancreas, with the potency order
`(Aib8 > Gly8 >
`reflecting
`receptor
`affinities
`Ser8 > Thr8). N-terminal modification of GLP-1 con-
`fers resistance to dipeptidyl peptidase IV degrada-
`tion. Such analogues are biologically active and have
`prolonged metabolic stability in vivo, which, if associ-
`ated with greater potency and duration of action, may
`help to realise the potential of GLP-1 in diabetes
`therapy. [Diabetologia (1998) 41: 271±278]
`
`Keywords Glucagon-like peptide-1, analogue, dipep-
`tidyl peptidase IV, non-insulin-dependent diabetes
`mellitus, therapy.
`
`The insulinotropic hormone glucagon-like peptide-1
`(GLP-1) is the product of tissue-specific post-transla-
`tional processing of the glucagon precursor, progluca-
`
`Received: 9 September 1997 and in revised form: 20 October
`1997
`
`Corresponding author: Dr. C. F. Deacon, Department of Medi-
`cal Physiology, The Panum Institute, Blegdamsvej 3, DK-2200
`Copenhagen N, Denmark
`Abbreviations: GLP-1, Glucagon-like peptide-1; DPP IV,
`dipeptidyl peptidase IV; PG, proglucagon; GRF, growth hor-
`mone-releasing factor; TFA, trifluoroacetic acid; ANOVA,
`analysis of variance; NIDDM, non-insulin-dependent diabetes
`mellitus; Aib, a-aminoisobutyric acid; HSA, human serum al-
`bumin; BHK, baby hamster kidney.
`
`gon (PG) in the L-cells of the gastrointestinal mucosa
`[1, 2]. This results in the formation of GLP-1 (7±
`36) amide (corresponding to PG (78±107) amide),
`which is the predominant form in humans, although
`small amounts of non-amidated glycine-extended
`GLP-1 (7±37) are also produced [3]. GLP-1 is one of
`the most potent insulin secretagogues identified [4]
`and this, together with the glucose-dependency of its
`actions [5±7], has focussed interest on its role as a reg-
`ulator of blood glucose and its potential as a thera-
`peutic agent in the treatment of non-insulin-depen-
`dent diabetes mellitus (NIDDM) [6, 8±11].
`Recent studies have shown that GLP-1 itself is the
`subject of further enzyme cleavage. In particular,
`dipeptidyl peptidase IV (DPP IV; EC 3.4.14.5) is im-
`
`MPI EXHIBIT 1072 PAGE 1
`
`MPI EXHIBIT 1072 PAGE 1
`
`

`

`272
`
`C. F. Deacon et al.: Dipeptidyl peptidase IV resistant GLP-1 analogues
`
`portant, resulting in a metabolite which is N-termi-
`nally truncated by 2 amino acids [12±14]. The result-
`ing peptide, GLP-1 (9±36) amide, is an endogenous
`metabolite [13] which is an antagonist in vitro [15,
`16]. Moreover, exogenously administered GLP-1 is
`also rapidly degraded in both diabetic and non-dia-
`betic subjects, with GLP-1 (9±36) amide being the
`major metabolite [17]. In a strain of rats lacking
`DPP IV, this metabolite is not formed [14]. DPP IV
`is highly specific and has strict substrate requirements
`[18, 19], raising the possibility of developing ana-
`logues which the enzyme is unable to cleave. Studies
`with another peptide substrate of DPP IV, growth
`hormone-releasing factor (GRF), have shown that
`analogues with N-terminal amino acid substitutions
`have some resistance to the enzyme's action [20].
`The present study was undertaken to examine wheth-
`er small modifications of the N-terminus of GLP-1
`would also confer resistance to degradation by DPP
`IV, while retaining the peptide's biological activity.
`
`Materials and methods
`
`Peptide synthesis. Peptides were synthesised on an Applied
`Biosystems 431A peptide synthesiser (Foster City, Calif.,
`USA), according to the Fmoc strategy, using the following pro-
`tected amino acid derivatives: Fmoc-Arg(Pmc), Fmoc-
`Trp(Boc), Fmoc-Glu(OBut), Fmoc-Lys(Boc), Fmoc-Gln(Trt),
`Fmoc-Tyr(But), Fmoc-Ser(But), Fmoc-Thr(But), Fmoc-
`His(Trt)
`and
`Fmoc-Asp(OBut)
`(Calbiochem-Novabio-
`chem AG, LaÈ ufelingen, Switzerland). The peptides were de-
`protected and cleaved from the resin by treatment with trifluo-
`roacetic acid (TFA)/phenol/thioanisole/water/ethanedithiol
`(83.25:6.25:4.25:2.00) for 180 min. After evaporation of the
`TFA, the crude peptide was precipitated with diethyl ether
`and purified by semipreparative HPLC on a C18 reversed-
`phase column eluted with a gradient of acetonitrile in
`0.05 mol/l (NH4)2SO4, pH 2.5. Peptide-containing fractions
`were applied to a Sep Pak C18 cartridge (Waters-Millipore, Mil-
`ford, Mass., USA), eluted with 70 % acetonitrile/0.1 % TFA
`and lyophilised. The final products were characterised by ami-
`no acid analysis, analytical reversed-phase HPLC and by plas-
`ma desorption mass spectrometry. Purity was more than 95 %
`by HPLC with detection at 214 nm. Analogues, substituted at
`position 8 of GLP-1 with either threonine (Thr8-GLP-1 (7±
`37)), glycine (Gly8-GLP-1 (7±37)), serine (Ser8-GLP-1 (7±
`36) amide) or a-aminoisobutyric acid (Aib8-GLP-1 (7±
`36) amide and Aib8-GLP-1 (7±37)) were prepared.
`
`Peptide stability in porcine plasma in vitro. The stability of each
`peptide in porcine plasma was determined by incubation at
`37 °C with 300 pmol/l of GLP-1 (7±36) amide or each analogue
`for up to 6 h. This was followed by reversed-phase HPLC and
`RIA according to a previously published method [13], using
`antiserum 2135 as described below.
`
`Peptide pharmacokinetics in vivo. Danish LYY strain pigs (33±
`40 kg) were used. Food was withdrawn 24 h before surgery, but
`animals had free access to drinking water. After premedication
`with ketamine chloride (Ketaminol, 10 mg/kg; Veterinaria AG,
`Zurich, Switzerland), animals were anaesthetised with 1 % ha-
`lothane (Halocarbon Laboratories, River Edge, NJ, USA), and
`
`anaesthesia was maintained with intermittent positive pressure
`ventilation using an anaesthesia ventilator in a semi-open sys-
`tem. Catheters were placed in the right carotid artery for sam-
`pling of arterial blood, and into a vein of the left ear for peptide
`infusion. After surgical preparation, animals were heparinised,
`an infusion (0.9 % NaCl) was set up and given via the ear vein
`catheter (5 ml/min), and the animals were left undisturbed for
`30 min.
`Four groups of four animals were used. Each group receiv-
`ed separate i. v. infusions of GLP-1 (7±36) amide and one ana-
`logue in a cross-over design with 80 min between each infu-
`sion. Synthetic GLP-1 (7±36) amide or analogues were dis-
`solved in saline containing 1 % human serum albumin (HSA;
`Behringwerke, Marburg, Germany), and infused at a rate of
`5 pmol × kg-1 × min-1 for 30 min using a syringe pump. Arterial
`blood samples (4 ml) were taken at 0, 5, 10, 15, 20, 22, 25, 27
`and 30 min from the start of the infusion. After 30 min, the in-
`fusion was stopped, and further blood samples were taken at
`1, 2, 4, 6, 10, 15, 20, 30, 40, 50 and 65 min. The amount of blood
`taken over the entire procedure was 160 ml, which, for a 40 kg
`pig, is equivalent to 4 % of the total blood volume.
`Blood samples were collected into chilled tubes containing
`EDTA (7.4 mmol/l final concentration; Merck, Darmstadt,
`Germany), aprotinin (500 kallikrein inhibitory equivalents/ml
`blood; Novo Nordisk, Bagsvñrd, Denmark) and diprotin A
`(0.1 mmol/l final concentration; Bachem, Bubendorf, Switzer-
`land), and kept on ice until centrifugation at 4 °C. Plasma was
`separated and stored at -20 °C until analysis with the RIAs de-
`scribed below. In addition, plasma collected during mins 22±30
`of each infusion was separately pooled for each group of four
`animals, extracted on Sep Pak C18 cartridges and analysed by
`reversed-phase HPLC and RIA as before [13], using antiserum
`2135, described below.
`
`Expression of the cloned human GLP-1 receptor. The human
`GLP-1 receptor cDNA was obtained from Dr. B. Thorens.
`The cDNA was subcloned into the pcDNA 1 vector (Invitro-
`gen Corporation, San Diego, Calif., USA) using the Hind III
`± EcoRI sites, and was then called pAH 260. Baby hamster kid-
`ney (BHK) cells were co-transfected with 20 mg pAH 260 and
`0.6 mg pSV 2 neo vector [21] using the methods described by
`Chen and Okayama [22], and grown in Dulbecco's modified
`Eagle's medium, 10 % fetal calf serum, 100 IU penicillin,
`100 mg/ml streptomycin and 1 mmol/l Na-pyruvate (all from
`Gibco, Life Technologies, Roskilde, Denmark). Stable clones
`were selected in medium containing 1 mg/ml Geneticin G-418
`(Gibco) and maintained at 37 °C, in an atmosphere containing
`5 % CO2. Stable clones were screened in a receptor binding as-
`say, and those expressing high levels of GLP-1 receptor were
`then recloned and screened in an adenylate cyclase assay to
`find clones with functional receptors. These clones were select-
`ed for further studies.
`
`Receptor binding. Receptor binding was carried out as previ-
`ously described [16], using BHK cells expressing the human
`pancreatic GLP-1 receptor. In brief, plasma membranes were
`prepared by homogenisation with two 10 s bursts using a Poly-
`tron PT 10±35 homogeniser (Kinematica, Lucerne, Switzer-
`land),
`in a buffer consisting of 10 mmol/l Tris-HCl with
`30 mmol/l NaCl, pH 7.4, containing in addition, 1 mmol/l dithi-
`othreitol, 5 mg/l leupeptin, 5 mg/l pepstatin, 100 mg/l bacitra-
`cin (all from Sigma, St. Louis, Mo, USA) and 16 mg/l aprotinin,
`and centrifuged on top of a layer of 41 % (wt/vol) sucrose at
`95 000 ´ g for 75 min. The white band between the two layers
`was diluted in buffer and centrifuged at 40 000 ´ g for 45 min.
`The precipitate containing the plasma membranes was sus-
`pended in buffer, and stored at -80 °C until use.
`
`MPI EXHIBIT 1072 PAGE 2
`
`MPI EXHIBIT 1072 PAGE 2
`
`

`

`C. F. Deacon et al.: Dipeptidyl peptidase IV resistant GLP-1 analogues
`
`273
`
`The binding assay was performed in 96-well filter microtitre
`plates. The buffer used was 50 mmol/l HEPES, pH 7.4, with the
`addition of 2.5 % (wt/vol) HSA grade V (Sigma). Peptide, trac-
`er and plasma membranes were incubated for 30 min at 30 °C.
`The tracer was prepared by iodination of GLP-1 (7±36) amide
`using the lactoperoxidase method [23]. Purification by HPLC
`as previously described [24] yielded 125I-Tyr19-GLP-1 (7±
`36) amide with a specific activity of 80 kBq/pmol.
`
`Isolated perfused pancreas. Danish LYY strain pigs (14±
`16 kg) were fasted overnight and anaesthetised with chloral-
`ose (100 mg/kg; Merck). The pancreases were isolated as pre-
`viously described [25] and perfused in a single-pass system,
`using a gassed (5 % CO2 in O2) Krebs-Ringer-bicarbonate
`perfusion medium containing, in addition, 0.1 % HSA, 5 %
`dextran T 70 (Pharmacia, Uppsala, Sweden) and 5 mmol/l
`glucose. The venous effluent was collected for 1-min intervals,
`centrifuged at 4 °C, and stored at -20 °C until analysis. Syn-
`thetic GLP-1 (7±36) amide or peptide analogues were dis-
`solved in 0.04 mol/l phosphate buffer, pH 7.4, containing 1 %
`HSA, and infused into the arterial line using a syringe pump,
`to give final concentrations of 1 nmol/l in the perfusate. Pep-
`tides were infused for 10 min periods, separated by 10±
`15 min rest periods, during which time endocrine secretion re-
`turned to basal levels. Insulin and glucagon concentrations in
`the venous effluent were measured by RIA as described be-
`low.
`
`Hormonal analysis. HPLC fractions were analysed using anti-
`serum 2135 [26, 27], which is ªside-viewingº, and which rec-
`ognises all molecules containing the central sequence of
`GLP-1 regardless of C- or N-terminal truncations or exten-
`sions. It cross-reacts fully with GLP-1 (7±37), 79 % with
`GLP-1 (9±36) amide, and has a detection limit of 5 pmol/l.
`Plasma samples were assayed for GLP-1 immunoreactivity
`using RIAs which are specific for each terminus of the mole-
`cule. N-terminal immunoreactivity was measured using the
`newly described antiserum 93242 [28], which has a cross-reac-
`tivity of approximately 10 % with GLP-1 (1±36) amide, and
`less than 0.1 % with GLP-1 (8±36) amide and GLP-1 (9±
`36) amide, and has a detection limit of 5 pmol/l. C-terminal
`immunoreactivity was measured using antiserum 89390 or
`92071 as appropriate. Antiserum 89390 [3, 29] has an abso-
`lute requirement
`for the intact amidated C-terminus of
`GLP-1 (7±36) amide, and cross-reacts less than 0.01 % with
`C-terminally truncated fragments, and 83 % with GLP-1 (9±
`36) amide. The detection limit is 1 pmol/l. Antiserum 92071
`[3] is specific for the C-terminus of GLP-1 (7±37) and cross-
`reacts less than 0.1 % with GLP-1 (7±36) amide. It has a de-
`tection limit of 4 pmol/l. For all assays, the intra-assay coeffi-
`cient of variation was less than 6 %. GLP-1 (7±36) amide,
`GLP-1 (7±37) or appropriate analogue were used as standard,
`and 125I-labelled GLP-1 (7±36) amide or GLP-1 (7±37) were
`used as tracer. Separation of antibody-bound from free pep-
`tide was achieved using plasma-coated charcoal [26, 29]. Plas-
`ma samples were extracted with 70 % ethanol (vol/vol, final
`concentration) before assay, giving recoveries of 75 % [27].
`The cross-reactivity of each analogue was determined for
`each antiserum.
`Venous effluent from the perfused pancreas was assayed
`using antiserum 2004 for insulin [27], and antiserum 4305 for
`glucagon [30].
`
`Calculations. During the peptide infusions, stable arterial pep-
`tide levels were achieved after 20 min, so the plateau concen-
`tration was defined as the mean of the last four measurements
`during the infusion. The plasma t1/2 was calculated by ln-linear
`
`regression analysis of peptide concentrations in samples col-
`lected after termination of the infusion, after subtraction of en-
`dogenous arterial GLP-1 concentrations. Insulin and glucagon
`output from the perfused pancreas are expressed as percentag-
`es of basal secretion, which is defined as 100 %.
`
`Statistical analysis. Data are expressed as means ± SEM, and
`were analysed using GraphPAD InStat software, version 1.13
`(San Diego, Calif, USA). In vitro data were analysed using
`one-sample t-tests, analysis of variance (ANOVA) and 2-tailed
`t-tests with correction for multiple comparisons as appropriate.
`In vivo data were analysed using repeated measures ANOVA
`and 2-tailed t-tests for paired or unpaired data as appropriate.
`Values of p less than 0.05 were considered to be significant.
`
`Results
`
`Analogue cross-reactivity. All analogues cross-react-
`ed more than 80 % with the appropriate C-terminal
`RIA; however, the cross-reactivity with the N-termi-
`nally directed 93242 assay varied considerably (Aib8-
`GLP-1, < 5 %; Gly8-GLP, 17 %; Ser8-GLP-1, 21 %;
`Thr8-GLP-1, 34 %). In practice, this meant that C-ter-
`minal assays (with appropriate standard) could be
`used for each analogue (detection limit, 5 pmol/l),
`but N-terminal immunoreactivity, using the appropri-
`ate analogue as standard, could be measured for only
`three analogues (Gly8-GLP, Ser8-GLP-1 and Thr8-
`GLP-1; detection limit, 20 pmol/l).
`
`Peptide stability in porcine plasma in vitro. GLP-1 (7±
`36) amide was degraded by porcine plasma in vitro at
`37°C, with a t1/2 of 28.1 ± 1.2 min (n = 12). HPLC
`analysis revealed the time-dependent generation of
`a second peak corresponding to GLP-1 (9±36) amide,
`and no other immunoreactive peaks were detected.
`Incubation of the GLP-1 analogues revealed a signif-
`icantly (p < 0.0001) prolonged t1/2 compared to GLP-
`(Gly8-GLP-1, 159 ± 12 min,
`1 (7±36) amide itself
`n = 3; Ser8-GLP-1, 174 ± 12 min, n = 9; Thr8-GLP-1,
`197 ± 14 min, n = 3), and again, HPLC analysis
`showed the formation of only one other peak, in addi-
`tion to the intact peptide. Degradation of Aib8-GLP-
`1 (n = 9) was undetectable after 6 h.
`
`Peptide pharmacokinetics in vivo. Stable arterial pep-
`tide concentrations were reached after 20 min of infu-
`sion. In all groups, concentrations determined by the
`C-terminal RIA (which measures both intact and N-
`terminally degraded peptide) exceeded those deter-
`mined by the N-terminal assay (illustrated for Thr8
`analogue group in Figure 1), while the ratio of N-ter-
`minal to C-terminal immunoreactivity was greater
`for each analogue than for GLP-1 (7±36) amide (Ta-
`ble 1). HPLC analysis of plasma pooled from each
`group during each infusion revealed two immunore-
`active peaks, corresponding to the intact peptide and
`the N-terminally truncated metabolite.
`In each
`
`MPI EXHIBIT 1072 PAGE 3
`
`MPI EXHIBIT 1072 PAGE 3
`
`

`

`274
`
`C. F. Deacon et al.: Dipeptidyl peptidase IV resistant GLP-1 analogues
`
`Fig. 1. Plasma GLP-1 immunoreactivity in the carotid artery,
`measured with C-terminally ( T ) and N-terminally ( X ) di-
`rected RIAs. Anaesthetised pigs received separate i. v. infu-
`sions (5 pmol × kg-1 × min-1) of GLP-1 (7±36) amide and Thr8-
`GLP-1 (7±37) in a cross-over design with 80 min between
`each infusion. Data are mean ± SEM; n = 4. The horizontal ar-
`row indicates the period of the infusion
`
`group, a greater percentage of the analogue remained
`undegraded compared to GLP-1 (7±36) amide (Ta-
`ble 1).
`For all four groups, there was no significant differ-
`ence between the C-terminal t1/2 for GLP-1 (7±
`36) amide or the analogues, but the N-terminal t1/2
`for each analogue was significantly prolonged (Ta-
`ble 2). For all four groups, the N-terminal t1/2 was
`t1/2 for GLP-1 (7±
`shorter than the C-terminal
`36) amide, but there was no significant difference be-
`
`Fig. 2. Binding affinity of GLP-1 analogues using the cloned
`human GLP-1 receptor. Individual curves are from one repre-
`sentative experiment where the data points are expressed as
`the mean of duplicate samples
`
`tween N- and C-terminal t1/2 for the analogues (Ta-
`ble 2).
`
`Receptor binding. All the analogues bound to the
`cloned human pancreatic GLP-1 receptor (Fig. 2),
`but with widely differing binding affinities (IC50; Ta-
`ble 3). The Aib8 and Gly8 analogues had similar affin-
`ities compared to GLP-1 (7±36) amide, while the oth-
`er two analogues had lower receptor affinities than
`GLP-1 (7±36) amide.
`
`Isolated perfused pancreas. Perfusion of the pancreas
`with 1 nmol/l GLP-1 (7±36) amide or the analogues
`increased insulin and decreased glucagon release rel-
`ative to basal secretion (Fig. 3). Of the analogues,
`Aib8-GLP-1 (7±36) amide was at least as potent as
`GLP-1 (7±36) amide in stimulating insulin and inhib-
`iting glucagon secretion, and was
`significantly
`(p < 0.05) more potent than the Ser8 and Thr8 ana-
`
`Table 1. Plasma concentrations of GLP-1 peptides attained during infusion of GLP-1 (7±36) amide and N-terminally modified an-
`alogues calculated using N- and C-terminally directed RIAs
`
`Infusion
`
`Plateau concentrations (pmol/l)
`
`N-terminal
`76.0 ± 12.2a
`GLP-1 (7±36) amide
`437.8 ± 49.1a
`Thr8-GLP-1 (7±37)
`108.0 ± 8.6a
`GLP-1 (7±36) amide
`336.0 ± 10.4a
`Gly8-GLP-1 (7±37)
`104.7 ± 22.8a
`GLP-1 (7±36) amide
`331.0 ± 59.7b
`Ser8-GLP-1 (7±36) amide
`80.3 ± 7.4a
`GLP-1 (7±36) amide
`Aib8-GLP-1 (7±36) amide
`NDL
`Values are mean ± SEM; n = 4, except for the % intact peptide
`after HPLC, which was calculated after HPLC analysis of a
`single sample of plasma pooled from four animals.
`
`N-terminal/C-terminal
`immunoreactivity
`
`% intact peptide
`after HPLC
`
`C-terminal
`16.4 ± 1.6c
`484.3 ± 103.1
`24.9
`62.2 ± 0.6
`689.3 ± 84.4
`62.5
`22.4 ± 2.1c
`500.3 ± 67.7
`23.8
`53.8 ± 2.4
`627.0 ± 24.0
`ND
`26.4 ± 2.1c
`387.0 ± 59.5
`24.8
`55.7 ± 3.9
`629.7 ± 137.1
`52.2
`31.7 ± 2.1
`267.8 ± 22.6
`17.2
`530.3 ± 16.2
`67.1
`NDL
`a p < 0.05 vs C-terminal concentration; b NS, p > 0.05 vs C-ter-
`minal concentration; c p < 0.005 vs analogue; ND, not deter-
`mined; NDL, not determined due to lack of cross-reactivity
`
`MPI EXHIBIT 1072 PAGE 4
`
`MPI EXHIBIT 1072 PAGE 4
`
`

`

`C. F. Deacon et al.: Dipeptidyl peptidase IV resistant GLP-1 analogues
`
`275
`
`Table 2. In vivo plasma t1/2 for GLP-1 (7±36) amide and N-terminally modified analogues calculated using N- and C-terminally di-
`rected RIAs
`
`Group
`
`Thr8-GLP-1 (7±37)
`Gly8-GLP-1 (7±37)
`Ser8-GLP-1 (7±36) amide
`Aib8-GLP-1 (7±36) amide
`Values are mean ± SEM; n = 4
`a NS, p > 0.05 vs C-terminal t1/2 for analogue; b p < 0.01 vs N-
`terminal t1/2 for GLP-1 (7±36) amide; c NS, p > 0.05 vs C-termi-
`
`GLP-1 (7±36) amide t1/2 (min)
`Analogue t1/2 (min)
`N-terminal
`C-terminal
`N-terminal
`C-terminal
`0.7 ± 0.05d
`4.0 ± 0.1
`3.9 ± 0.2ab
`4.2 ± 0.4c
`0.9 ± 0.03d
`4.3 ± 0.5
`3.3 ± 0.4ab
`3.5 ± 0.7c
`0.9 ± 0.06d
`4.1 ± 0.2
`3.7 ± 0.4ab
`4.2 ± 0.2c
`1.1 ± 0.07d
`4.3 ± 0.3
`4.4 ± 0.2c
`NDL
`nal t1/2 for GLP-1 (7±36) amide; d p < 0.01 vs C-terminal t1/2 for
`GLP-1 (7±36) amide; NDL, not determined due to lack of
`cross-reactivity
`
`Table 3. Receptor binding affinities of GLP-1 (7±36) amide
`and N-terminally modified analogues in baby hamster kidney
`cells expressing the human pancreatic GLP-1 receptor
`
`Peptide
`
`Binding affinity
`(IC50, nmol/l)
`0.78 ± 0.29
`GLP-1 (7±36) amide
`49 ± 3.7a
`Thr8-GLP-1 (7±37)
`2.8 ± 0.42b
`Gly8-GLP-1 (7±37)
`9.0 ± 1.9a
`Ser8-GLP-1 (7±36) amide
`0.45 ± 0.05b
`Aib8-GLP-1 (7±37)
`Values are mean ± SD of separate triplicate experiments.
`a p < 0.001 vs GLP-1 (7±36) amide; b NS, p > 0.05 vs GLP-1 (7±
`36) amide
`
`n
`
`8
`4
`4
`3
`3
`
`logues in raising insulin output. It was also the most
`potent (p < 0.05) of all the analogues in reducing glu-
`cagon output. The Gly8 analogue was not significantly
`different to GLP-1 (7±36) amide in stimulating insu-
`lin or inhibiting glucagon secretion, but was more po-
`tent (p < 0.05) than the Ser8 and Thr8 analogues in in-
`hibiting glucagon release, while the Thr8 analogue
`was the least potent of the analogues tested.
`
`Discussion
`
`This study has demonstrated that small alterations in
`the N-terminus of GLP-1 confer resistance to the ac-
`tion of the enzyme DPP IV. Such analogues retain bi-
`ological activity, and have an improved metabolic sta-
`bility.
`In incubations with human plasma in vitro, DPP
`IV is the main enzyme responsible for GLP-1 degra-
`dation [12, 13], and a thorough study by Pauly et al.
`[31] concluded that only minor secondary degrada-
`tion could be attributed to other serum proteases. Ac-
`cordingly, the in vitro t1/2 of the GLP-1 analogues
`were considerably extended relative to GLP-1 (7±
`36) amide. There was no detectable degradation of
`the Aib8 analogue, and the slow degradation of the
`other three analogues could reflect the substrate
`specificity of DPP IV, as was found for GRF; substitu-
`tion of the alanine in position 2 of modified GRF an-
`alogues with either serine or threonine reduced the
`
`Fig. 3. Insulin and glucagon secretion by the isolated perfused
`porcine pancreas during infusion of 1 nmol/l GLP-1 (7±
`36) amide or N-terminally modified analogues. Data are ex-
`pressed as a percentage of basal output (defined as 100 %),
`and are mean ± SEM of 4 experiments, except for the glucagon
`output during GLP-1 (7±36) amide infusion where n = 3.
`* p < 0.05; ** p < 0.01 compared to basal output
`
`cleavage rates to less than 5 %, of that of the Ala2 an-
`alogue [20]. That DPP IV is also the main enzyme re-
`sponsible for in vivo N-terminal degradation of GLP-
`1 is illustrated by the increased proportions of N-ter-
`minal immunoreactivity (reflecting intact peptide)
`seen during i. v. infusion of each analogue relative to
`GLP-1 itself, and confirmed by HPLC analysis which
`revealed only two immunoreactive peaks corre-
`sponding to the intact peptide and the N-terminally
`truncated metabolite. In these in vivo studies, ana-
`logue N-terminal t1/2 were prolonged and equalled
`the C-terminal t1/2, but it is noteworthy that they
`could not be extended beyond those determined by
`
`MPI EXHIBIT 1072 PAGE 5
`
`MPI EXHIBIT 1072 PAGE 5
`
`

`

`276
`
`C. F. Deacon et al.: Dipeptidyl peptidase IV resistant GLP-1 analogues
`
`the C-terminal assay, presumably due to the presence
`of other, non-DPP IV-mediated degradation path-
`ways which become relevant in the intact animal. In
`studies of GLP-1 metabolism in the rat [14], dog [16,
`32] and pig [33], N-terminal degradation was shown
`to be particularly important, but the involvement of
`other enzymes was also indicated. Renal metabolism
`leading to substantial degradation to small, undetect-
`able fragments or complete cleavage was indicated to
`be a major route of GLP-1 elimination [33], and may
`explain why no other metabolites were detected after
`HPLC analysis in the present study.
`The binding affinity (IC50) for GLP-1 in the pre-
`sent study correlates well with the previously pub-
`lished Kd of 0.5 nmol/l [34]. All four analogues also
`bound to the cloned GLP-1 receptor, with the Aib8
`and Gly8 analogues having similar affinities to GLP-
`1 itself. It may be that, for the other analogues, steric
`hindrances cause the reduction in binding affinity,
`with the polar hydroxyl group in the serine and threo-
`nine residues impairing binding due to spatial con-
`straints.
`In the isolated perfused pancreas, all the GLP-1
`analogues were, to varying extents, capable of releas-
`ing insulin and/or inhibiting glucagon secretion, with
`potency orders reflecting receptor affinities. Similar
`findings were seen for GRF; serine, threonine and
`glycine substituted analogues were stabilised against
`proteolysis in plasma, but had low inherent growth
`hormone releasing activity when tested in vitro [35].
`However, the potency order in vivo changed, with
`the Thr2 analogue becoming more potent than native
`GRF, which may be explained by the improved meta-
`bolic stability of the analogue in vivo compensating
`for its lower in vitro potency.
`GLP-1 has been suggested to be a useful new ther-
`apy in the treatment of NIDDM [6, 8±11]. However,
`the therapeutic potential of native GLP-1 is limited
`by its susceptibility to degradation by DPP IV [12±
`14], with the concomitant formation of a metabolite
`which may act as an antagonist in its own right [15,
`16]. The short metabolic stability of the intact pep-
`tide can be offset by sustained i. v. infusion or by de-
`velopment of a protracted formulation, but this does
`not overcome the effect of DPP IV action. In recent
`studies in patients with NIDDM in whom GLP-1
`was infused overnight, Rachman et al. [36, 37] ob-
`served that, despite an initial fall, blood glucose con-
`centrations gradually increased during the night, al-
`though they remained lower than in the saline-treat-
`ed group. These authors concluded that ªGLP-1
`may have been losing its efficacy overnightº. Anoth-
`er explanation could be that concentrations of GLP-
`1 (9±36) amide increase during the overnight GLP-1
`infusion, so that the gradual rise in blood glucose
`may not be due to a post-receptor down-regulation,
`but rather to a local accumulation of metabolite an-
`tagonising the action of the residual intact peptide.
`
`In these studies [36, 37], the relative concentrations
`of intact GLP-1 and metabolite were not assessed,
`but this was addressed in another study, where GLP-
`1 (9±36) amide was found to reach 80 % of the total
`plasma immunoreactivity during an i. v. infusion of
`GLP-1 in NIDDM and control subjects [17]. Howev-
`er, marked desensitisation appears not to occur, since
`when GLP-1 was infused in NIDDM patients contin-
`uously i. v. over a 7-day period, both fasting and post-
`prandial glucose levels were significantly lower on
`the 7th day of treatment compared to pre-treatment
`values [38]. This study [38] was the first to show that
`continuous GLP-1 infusion is capable of improving
`glycaemic control in NIDDM patients for a pro-
`longed period, although it should be borne in mind
`that neither this [38] nor any other study has un-
`equivocally excluded the possibility that some loss
`of efficacy occurs over the course of the treatment
`period.
`It appears that in order to maintain its effect,
`GLP-1 must be continuously present. Thus, in NID-
`DM patients, when the peptide was infused for only
`16 h instead of continuously over 24 h, the beneficial
`effect on fasting blood glucose was reduced [39].
`Similar results are seen when GLP-1 is given to
`NIDDM patients as repeated s. c. injections. When
`administered 3 times daily before meals, it retained
`its ability to reduce postprandial increases in blood
`glucose throughout the 1 week course of the study
`[40], but the effect was lost between meals, suggest-
`ing that GLP-1 concentrations fell below the thresh-
`old for activity between successive injections. An
`earlier study be the same group [41] followed the ki-
`netics, and showed that total GLP-1 immunoreactiv-
`ity returned to basal levels by 215 min after the pep-
`tide injection. Similar findings were reported when
`immunoreactive GLP-1 was characterised by HPLC
`after s. c. administration to diabetic and non-diabetic
`subjects [17]. By 30 min after administration, the
`metabolite accounted for around 80 % of the plasma
`immunoreactivity, and concentrations of both GLP-
`1 (7±36) amide and GLP-1 (9±36) amide returned to
`basal values within 4 h. The effect of repeated s. c.
`injections of GLP-1 given before meals was con-
`firmed in another study using poorly controlled
`NIDDM patients on sulphonylurea therapy [42].
`Here, the peptide maintained its beneficial effect
`on postprandial glucose levels over the 3-week study
`period, although, again, it had no effect on fasting
`glucose levels. Thus, it appears that when GLP-1 is
`given as repeated injections before meals, full 24 h
`control of blood glucose is not attained. Tachyphy-
`laxis appears not to occur, possibly because both in-
`tact peptide and the metabolite are eliminated be-
`tween successive injections, preventing accumulation
`of a potential antagonist, however, leaving a period
`when there is insufficient intact GLP-1 left to main-
`tain an effect.
`
`MPI EXHIBIT 1072 PAGE 6
`
`MPI EXHIBIT 1072 PAGE 6
`
`

`

`C. F. Deacon et al.: Dipeptidyl peptidase IV resistant GLP-1 analogues
`
`277
`
`The kinetic data reported here for the GLP-1 ana-
`logues indicate that resistance to degradation by
`DPP IV is associated with prolonged metabolic sta-
`bility in vivo, raising the possibility that such ana-
`logues may have greater potency and duration of ac-
`tion. Clearly, further dynamic studies in vivo are re-
`quired to test whether this hypothesis is valid, and to
`see whether the 3-fold improvement in plasma t1/2 of
`the analogues in itself is sufficient to maintain an ef-
`fect or requires development of a protracted formula-
`tion. Nonetheless, DPP IV-resistant analogues may
`be one method of realising GLP-1's potential in dia-
`betes therapy, by extending its duration of action
`while, at the same time, minimising the build up of
`undesirable metabolites.
`
`Acknowledgements. This work was supported by grants from
`the Danish Medical Research Council and the Danish Biotech-
`nology Programme. The technical assistance of Letty Klarskov,
`Mette Olesen and Mette Frost is gratefully acknowledged. Siv
`Hjort is thanked for kindly providing the pAH 260 vector.
`
`References
`
`1. Holst JJ (1983) Gut glucagon, enteroglucagon, gut gluca-
`gonlike immunoreactivity, glicentin: current status. Gastro-
`enterology 84: 1602±1613
`2. érskov C (1992) Glucagon-like peptide-1, a new hormone
`of the entero-insular axis. Diabetologia 35: 701±711
`3. érskov C, Rabenhùj L, Wettergren A, Kofod H, Holst JJ
`(1994) Tissue and plasma concentrations of amidated and
`glycine-extended glucagon-like peptide-1 in humans. Dia-
`betes 43: 535±539
`4. érskov C, Holst JJ, Nielsen OV (1988) Effect of truncated
`glucagon-like peptide-1 [proglucagon-(78±107) amide] on
`endocrine secretion from pig pancreas, antrum and non-an-
`tral stomach. Endocrinology 123: 2009±2013
`5. Kreymann B, Williams G, Ghatei MA, Bloom SR (1987)
`Glucagon-like peptide 17±36: a physiological incretin in
`man. Lancet ii: 1300±1304
`6. Nathan DM, Schreiber E, Fogel H, Mojso

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