throbber
Systemic Administration of the Long-Acting GLP-1
`Derivative NN2211 Induces Lasting and Reversible
`Weight Loss in Both Normal and Obese Rats
`
`Philip J. Larsen,1,2 Christian Fledelius,3 Lotte Bjerre Knudsen,4 and Mads Tang-Christensen1
`
`Postprandial release of the incretin glucagon-like pep-
`tide-1 (GLP-1) has been suggested to act as an endoge-
`nous satiety factor in humans. In rats, however, the
`evidence for this is equivocal probably because of very
`high endogenous activity of the GLP-1 degrading en-
`zyme dipeptidyl peptidase-IV. In the present study, we
`show that intravenously administered GLP-1 (100 and
`500 ␮g/kg) decreases food intake for 60 min in hungry
`rats. This effect is pharmacologically specific as it is
`inhibited by previous administration of 100 ␮g/kg ex-
`endin(9-39), and biologically inactive GLP-1(1-37) had
`no effect on food intake when administered alone (500
`␮g/kg). Acute intravenous administration of GLP-1 also
`caused dose-dependent inhibition of water intake, and
`this effect was equally well abolished by previous ad-
`ministration of exendin(9-39). A profound increase in
`diuresis was observed after intravenous administration
`of both 100 and 500 ␮g/kg GLP-1. Using a novel long-
`acting injectable GLP-1 derivative, NN2211, the acute
`and subchronic anorectic potentials of GLP-1 and deriv-
`atives were studied in both normal rats and rats made
`obese by neonatal monosodium glutamate treatment
`(MSG). We showed previously that MSG-treated ani-
`mals are insensitive to the anorectic effects of centrally
`administered GLP-1(7-37). Both normal and MSG-
`lesioned rats were randomly assigned to groups to
`receive NN2211 or vehicle. A single bolus injection of
`NN2211 caused profound dose-dependent inhibition of
`overnight food and water intake and increased diuresis
`in both normal and MSG-treated rats. Subchronic mul-
`tiple dosing of NN2211 (200 ␮g/kg) twice daily for 10
`days to normal and MSG-treated rats caused profound
`inhibition of food intake. The marked decrease in food
`intake was accompanied by reduced body weight in both
`groups, which at its lowest stabilized at ⬃85% of initial
`body weight. Initial excursions in water intake and
`diuresis were transient as they were normalized within
`a few days of treatment. Lowered plasma levels of
`triglycerides and leptin were observed during NN2211
`treatment in both normal and MSG-treated obese rats.
`
`From the 1Laboratory of Obesity Research, Center for Clinical and Basic
`Research, Ballerup, Denmark; and 2Neuroendocrine Pharmacology, 3Pharma-
`cological Research 2, and 4Molecular Pharmacology, Novo Nordisk A/S,
`Copenhagen, Denmark.
`Address correspondence and reprint requests to Philip J. Larsen, Dr, MSci,
`Laboratory of Obesity Research, Centre for Clinical and Basic Research,
`Ballerup Byvej 222, 2750 Ballerup, Denmark. E-mail: pjl@ccbr.dk.
`Received for publication 24 January 2001 and accepted in revised form 26
`July 2001.
`L.B.K and C.F. are employees of and hold stock in Novo Nordisk.
`ANOVA, analysis of variance; DEXA, dual energy X-ray absorptiometry;
`DPP-IV, dipeptidyl peptidase-IV; EE, energy expenditure; FFA, free fatty acids;
`GLP-1, glucagon-like peptide-1; MSG, monosodium glutamate; RER, respira-
`tory exchange ratio; TG, triacylglycerol.
`
`In a subsequent study, a 7-day NN2211 treatment period
`of normal rats ended with measurement of energy ex-
`penditure (EE) and body composition determined by
`indirect calorimetry and dual energy X-ray absorptiom-
`etry, respectively. Compared with vehicle-treated rats,
`NN2211 and pair-fed rats decreased their total EE
`corresponding to the observed weight loss, such that EE
`per weight unit of lean body mass was unaffected.
`Despite its initial impact on body fluid balance, NN2211
`had no debilitating effects on body water homeostasis as
`confirmed by analysis of body composition, plasma elec-
`trolytes, and hematocrit. This is in contrast to pair-fed
`animals, which displayed hemoconcentration and ten-
`dency toward increased percentage of fat mass. The
`present series of experiments show that GLP-1 is fully
`capable of inhibiting food intake in rats via a peripher-
`ally accessible site. The loss in body weight is accompa-
`nied by decreased levels of circulating leptin indicative
`of loss of body fat. The profound weight loss caused by
`NN2211 treatment was without detrimental effects on
`body water homeostasis. Thus, long-acting GLP-1 deriv-
`atives may prove efficient as weight-reducing therapeu-
`tic agents for overweight patients with type 2 diabetes.
`Diabetes 50:2530 –2539, 2001
`
`Peripheral administration of glucagon-like pep-
`
`tide-1 (GLP-1) acutely affects food intake in hu-
`mans, but
`the underlying mechanisms that
`decrease food intake concomitant with earlier
`onset of subjective sensation of fullness are not fully
`understood (1– 4). The anorectic effects of continuous
`intravenous administration of GLP-1 are present in indi-
`viduals who are lean or obese or have type 2 diabetes
`(1,3,4), suggesting that the observed effects are part of a
`physiologically relevant meal-terminating system. Results
`from similar experiments in rats have been ambiguous,
`probably because of high activity of GLP-1 degrading
`enzyme dipeptidyl peptidase-IV (DPP-IV) (5,6). Thus, early
`experiments were unable to demonstrate peripheral ef-
`fects of intraperitoneal GLP-1 injections on feeding be-
`havior (7,8), whereas later experiments have shown
`significant but short-lasting anorectic effects of subcuta-
`neous administration of GLP-1 (9). Anorexia induced by
`peripheral administration of GLP-1 involves vagal control
`of gastric motility (10,11).
`Central administration of 1–3 ␮g of GLP-1 specifically
`inhibits food intake in rats via a hypothalamic site that is
`sensitive to neonatal monosodium glutamate (MSG) le-
`sioning (12). However, central administration of slightly
`
`2530
`
`DIABETES, VOL. 50, NOVEMBER 2001
`
`MPI EXHIBIT 1086 PAGE 1
`
`

`

`higher doses of GLP-1 leads to taste aversion, but because
`this latter effect is unaffected by MSG treatment, it further
`stresses the specificity of the central GLP-1–induced an-
`orexia (12). In addition, it is possible to elicit anorexia
`without concomitant taste aversion if GLP-1 is injected
`directly into the hypothalamic paraventricular nucleus
`(13). Acute injections of both GLP-1 and NN2211 exerts
`profound adipsia and diuresis. These effects on body water
`homeostasis could potentially hamper long-term treatment
`of patients with type 2 diabetes with GLP-1 agonists, and
`the potential anorectic effects of these agonists may be
`accompanied with debilitating affects on body water ho-
`meostasis.
`Given that peripheral administration of GLP-1 affects
`food intake in humans, we decided to study further the
`anorectic potential of this peptide in the laboratory rat.
`Dose-response studies investigating the effect of intrave-
`nously administered GLP-1(7-37) or a novel long-acting
`acylated GLP-1 derivative NN2211 (14) on food intake
`were performed. In continuation of acute pharmacological
`studies, we examined the effect on food intake and body
`weight of twice daily subcutaneous administration of
`NN2211 for 10 days followed by a 5-day recovery period.
`To study the impact of 7 days of NN2211 treatment on
`energy expenditure (EE) and body composition, we stud-
`ied normal rats by indirect calorimetry and subsequently
`subjected them to dual energy X-ray absorptiometry
`(DEXA) scanning. Before, during, and after treatment,
`blood biochemical markers of energy and fluid homeosta-
`sis metabolic state were monitored.
`
`RESEARCH DESIGN AND METHODS
`Animals. All experiments were carried out on male Wistar rats. Normal male
`rats arrived at the animal facilities 2 weeks before experimentation, and until
`use, animals were housed three to four per cage under standard laboratory
`conditions (12:12 light:dark cycle, lights on 6:00 A.M.). Before experimentation,
`animals had free access to a standard rat diet (Altromin 1324) and water ad
`libitum. Hypothalamically obese rats were obtained by treating neonatal rats
`with MSG. Pregnant Wistar rats (Møllegården, Ll; Skensved, Denmark) arrived
`at the animal unit 1 day before timed labor (E21). Neonatal Wistar pups
`received a number of subcutaneous injections with MSG (L-glutamic acid,
`G-1626; 4 mg/g body wt; Sigma, Vallensbaek Strand, Denmark) dissolved in
`sterile distilled H2O. Injections were given at days P1, P3, P5, P7, and P9
`according to a previously published method (12). After weaning, pups were
`separated by sex and allowed to grow to the age of 12–14 weeks before
`experiments were initiated. Neonatal MSG treatment is accompanied by adult
`obesity and a number of neuroendocrine dysfunctions (hypothalamic hypo-
`gonadism, low fertility, stunted growth, chronic HPA-axis activation), and
`currently used MSG-treated rats all displayed characteristic stunted growth,
`adiposity, short tail caused by self-mutilation, and apparent blindness. All
`experiments were carried out in accordance with guidelines provided by the
`Danish Justice Department and approved by a personal license to P.J.L.
`Formulation of GLP-1(7-37), GLP-1(1-37), exendin(9-39), and NN2211.
`GLP-1(7-37) and GLP-1(1-37) were obtained from L. Thim (Novo Nordisk);
`exendin(9-39) was purchased from Bachem (Bissendorf Biochemicals, Ham-
`burg, Germany). High-performance liquid chromatography analyses of all
`peptides claim ⬎90% purity. Peptides were dissolved in sterile isotonic saline
`to which 1% bovine serum albumin (BSA) was added (Fraction V, #735 086;
`Boehringer Mannheim, Mannheim, Germany). The GLP-1 derivative NN2211
`(Arg34, Lys26-[N-⑀(␥-Glu[N-␣-hexadecanoyl])]-GLP-1[7-37]) was synthesized
`according to a previously described procedure (14). The compound NN2211 is
`a member of a group of pharmacologically active GLP-1 derivatives that have
`long plasma half-lives (14). Thus, NN2211 is an acylated GLP-1 derivative with
`a plasma half-life of ⬃14 h in pigs. Pharmacokinetic data from rats show that
`⫽ 4 h probably as a result of considerably higher endogenous DPP-IV
`t1/2
`activity in this species (L.B.K., unpublished observations). NN2211 was
`dissolved in sterile phosphate-buffered saline (50 mmol/l, pH 7.4) to a final
`concentration of either 0.1 or 1.0 mg/ml. Solutions were always made fresh ⬃1
`h before use and stored at 4°C in sterile tubes. Material from several batches
`
`P.J. LARSON AND ASSOCIATES
`
`of NN2211 was used, and corrections for impurity were always performed.
`Subcutaneous injections were administered using standard 1-ml syringes
`equipped with 25-G needles.
`Experiment 1: single dose of GLP-1. Sixteen adult male Wistar rats were
`equipped with jugular intravenous catheters (Department of Pharmacology,
`The Panum Institute, University of Copenhagen). Catheters were implanted
`under Avertin (tribromoethanol, 200 mg/kg) anesthesia in the right jugular
`vein with the tip aiming at the right atrium. After placement, the catheter was
`externalized via subcutaneous tunneling to the interscapular area. Catheter
`patency was secured by instillation of heparinized (1,000 IU/l) isotonic sterile
`saline into the catheter before closure with a metal rod. After 7 days of
`postoperative recovery, animals were housed individually in standard meta-
`bolic cages (Techniplast, Gazzoda, Italy) and acclimatized over a period of 7
`days to a 5-h restricted feeding scheme with access to food from 8:00 A.M. to
`1 P.M. and water ad libitum.
`Seven days after initiation of the restricted feeding scheme, animals were
`assigned to a random crossover dosing paradigm. Five minutes before
`presentation of food, animals received an intravenous injection of GLP-1 (5,
`100, or 500 ␮g/animal). Statistical analysis of intergroup treatment variation
`was carried out using factorial analysis of variance (ANOVA) followed by
`Scheffe post hoc analysis.
`Experiment 2: single dose of NN2211. Normal adult male Wistar rats (n ⫽
`16) and MSG-treated rats (n ⫽ 16) were housed individually in metabolic
`cages with free access to a rat diet and water for at least 1 week before
`experimentation. Animals were kept in a 12:12 light:dark cycle, and the effect
`of NN2211 on nighttime food intake was assessed by injecting animals
`subcutaneously 2–3 h before lights out. Animals were left without food and
`water in the period from dosing to the onset of darkness. Three doses of
`NN2211 and vehicle were tested in a random crossover experiment (10, 50,
`and 200 ␮g/kg). Food and water intake and diuresis were monitored every 30
`min for the first 2 h after onset of nighttime (lights out) and finally 12 h later
`at lights on (t720). All measurements were done in complete darkness with the
`assistance of night vision goggles (Bausch and Lomb, Rochester, NY). The
`minimum interval between experiments was 48 h. Statistical analysis of
`intergroup treatment variation was carried out using factorial ANOVA fol-
`lowed by Scheffe post hoc analysis.
`Experiment 3: continuous administration of NN2211. Beginning 1 week
`before the first dose was administered, normal adult male Wistar rats (n ⫽ 16)
`and MSG-treated rats (n ⫽ 16) were housed individually in metabolic cages
`with free access to a rat diet and water. Animals were kept on a 12:12
`light:dark cycle, and the subchronic effect of two daily subcutaneous injec-
`tions of NN2211 or vehicle on body weight, food and water intake, diuresis,
`and feces excretion was monitored. All parameters were measured between 9
`and 11 A.M. while animals received their morning dose. On the basis of daily
`food intake, animals were stratified to different treatment groups ensuring
`comparable baseline values for food and water intake. Animals received two
`daily injections of NN2211 (100 or 200 ␮g/kg b.i.d.) at 8:00 A.M. and 7:00 P.M.
`throughout a 10-day period followed by a 5-day drug-free recovery period.
`Animals were weighed every morning (between 9 and 11 A.M.) together
`with measurement of daily food and water intake as well as diuresis and feces
`excretion. At day 0, orbital blood samples were taken from all animals before
`the first dose was administered. Further orbital blood samples were taken at
`day 7 and 14 (i.e., during and after treatment with NN2211). At the final day of
`experimentation, animals were decapitated and trunk blood was collected as
`described. Different dose administrations were conducted in two separate
`experiments, each with its own vehicle-treated control groups. Data from
`these control groups were pooled.
`Statistical analysis of effect of treatment on body weight, food and water
`intake, diuresis, and feces excretion was carried out using factorial ANOVA
`followed by Bonferroni correction for multiple comparison. Biochemical data
`from plasma samples were analyzed using factorial ANOVA followed by
`Fisher’s or Scheffe’s post hoc analysis.
`Experiment 4: effect of subchronic NN2211 treatment on energy ex-
`penditure and body composition. Twenty-four 12-week-old male rats were
`used in this study. The rats were housed individually in a temperature- (20°C)
`and light-controlled environment (12:12 light:dark cycle; lights on from 7:00
`A.M.) with free access to food and water for at least 7 days before experimen-
`tation. The rats were stratified into three groups (G1, G2, and G3) according
`to weight 3 days before study start (n ⫽ 7 per group). The rats in G1 and G3
`were treated with vehicle, and the rats in G2 were treated with NN2211 (200
`␮g/kg b.i.d.). The rats in G1 and G2 had free access to food and water during
`the 7-day treatment period, whereas the rats in G3 were “single” pair fed after
`the rats in G2. Body weight and food and water intake were recorded daily. By
`the end of the study (day 7), oxygen consumption and body composition were
`determined by indirect calorimetry and DEXA, respectively. Likewise, blood
`samples were collected for determination of hematocrit as well as for plasma
`
`DIABETES, VOL. 50, NOVEMBER 2001
`
`2531
`
`MPI EXHIBIT 1086 PAGE 2
`
`

`

`WEIGHT LOSS THERAPY WITH GLP-1 DERIVATIVE
`
`FIG. 1. Single intravenous infusions of GLP-1(7-37) cause dose-depen-
`dent anorexia in rats that are kept on a restricted feeding schedule.
`Food intake over the initial 30 min of the feeding session was recorded
`for all doses tested (5, 100, and 500 ␮g/rat). *, statistically significant
`differences from vehicle-treated animals (P < 0.05 as determined by
`ANOVA followed by Scheffe’s post hoc analysis).
`
`levels of triacylglycerol (TG), glycerol, free fatty acids (FFAs), and total
`cholesterol.
`DEXA. Body composition was determined by DEXA (pDEXA Sabre, Stratec
`Medizintechnic; Norland Medical Systems, Pho¨ rzheim, Germany). The instru-
`ment settings used were as follows: a scan speed of 40 mm/s, a resolution of
`1.0 ⫻ 1.0 mm, and automatic/manual histogram width estimation. The
`coefficient of variation as assessed by 10 repeated measurements (with
`repositioning of the rat between each measurement) was 3.48, 3.17, and 3.73%
`for bone mineral content, lean tissue mass, and fat tissue mass, respectively.
`By the end of the study (day 7), the rats were killed. The carcasses were stored
`in plastic bags at ⫺20°C before determination of body composition; which
`was determined on defrosted carcasses.
`Indirect calorimetry. Oxygen consumption, CO2 production, EE, and the
`respiratory exchange ratio (RER) were determined by indirect calorimetry
`(Oxymax System; Columbus Instruments, Columbus, OH). The rats (n ⫽ 1 per
`chamber) were placed in airtight acrylic chambers (10.5 l). Oxygen and CO2
`concentrations in the chamber in- and outlet gas were determined simulta-
`neously every 20.25 min over a period of 4.4 h. Instrument settings used were
`as follows: a gas flow rate of 1.86 l/min, settle time of 90 s; measure time of
`40 s, and system recalibration for each eight-chamber measuring cycle. By the
`end of the study (day 7), the nonfasted rats were subjected to indirect
`calorimetry (from 8:00 A.M. 1:00 P.M.). In contrast to the previous 6 days, the
`nonfasted rats did not receive NN2211 between 7:30 and 8:30 before indirect
`calorimetry. On the day of indirect calorimetry, the rats received treatment at
`9:30 —after four pretreatment measurements. The rats had no access to food
`or water during their stay in the acrylic chamber. The indirect calorimetric
`measurements were performed over 3 days. As a “positive” instrument
`control, every day included a reference animal “treated” with the EE increas-
`ing compound 2,4-dinitrophenol (DNP, Sigma). On the basis of the measure-
`ments of O2 and CO2 in the chamber in- and outlet gas, estimates of O2
`consumption, CO2 production, EE, and RER were calculated.
`Blood sampling and biochemical assays
`Blood sampling. Orbital blood samples were obtained by puncture of the
`orbital venous plexus with glass capillary tubes. Samples were taken in
`standard heparinized EDTA (0.18 mol/l) glass tubes (Vacutainer) to which
`aprotinin (1,500 KIE/ml) and bacitracin (3%) were added. After sampling,
`tubes were kept on ice before being centrifuged (4°C at 5,000g for 10 min), and
`the resulting plasma was stored at ⫺80°C before being analyzed. Trunk blood
`was obtained by decapitating animals and sampling into heparinized (⬃500
`IU/tube) glass tubes to which aprotinin (1,500 KIE/ml) and bacitracin (3%)
`were added. Glycerol and FFA concentrations were determined in EDTA (0.18
`mol/l) plasma containing 1% NaF (wt/vol).
`Plasma glucose. Plasma glucose was measured on a standard COBAS
`analyzer (Toxicology Projects & Planning, Novo Nordisk, Copenhagen, Den-
`mark).
`Plasma leptin. Plasma leptin was measured using a commercially available
`
`mouse leptin enzyme-linked immunosorbent assay kit (Crystal Chemical,
`Chicago, IL), showing ⬎95% cross-reactivity to rat leptin.
`Plasma biochemistry. Orbital blood samples (days 0, 7, and 14) were taken
`from rats that were receiving 100 ␮g/kg b.i.d. NN2211 and a set of correspond-
`ing vehicle-treated animals. Plasma values of sodium, TG, cholesterol, creat-
`inine, carbamide, and total protein were measured on a standard COBAS
`analyzer. FFA and glycerol were measured on a standard Hitachi Automatic
`analyzer.
`Plasma potassium. Orbital blood samples (days 0, 7, and 14) were taken
`from rats that were receiving 200 ␮g/kg b.i.d. NN2211 and a set of correspond-
`ing vehicle-treated animals. Blood was collected in heparinized glass tubes
`(Vacutainer), and the potassium content in resulting plasma was measured
`potentiometrically with an ion selective probe on a standard COBAS analyzer.
`
`RESULTS
`Experiment 1
`Effects of a single dose of GLP-1 on feeding behav-
`ior. Acute intravenous administration of high doses of
`GLP-1 (100 and 500 ␮g/animal) decreased food intake at 30
`and 60 min after onset of feeding period in rats that were
`kept on a restricted feeding scheme (Fig. 1). The anorectic
`effect of the highest dose of GLP-1 (500 ␮g/animal) was
`completely abolished by previous administration of 100 ␮g
`of exendin(9-39) (6.1 ⫾ 0.4 vs. 5.9 ⫾ 0.3 g). Also, the
`biologically inactive peptide GLP-1(1-37) (500 ␮g/animal)
`had no acute effect on food intake (6.1 ⫾ 0.4 vs. 6.2 ⫾
`0.3 g). Water intake was similarly decreased in rats that
`were receiving an intravenous injection of GLP-1, and the
`pharmacological characteristics of water consumption
`mirrored that of feeding.
`Intravenous administration of GLP-1 also affected diure-
`sis. Water excretion was markedly and dose-dependently
`increased in rats that were receiving intravenous bolus
`injections of GLP-1 (100 and 500 ␮g/animal; Fig. 2). The
`effect displayed pharmacological specificity in as much it
`was not elicited by 500 ␮g/animal GLP-1(1-37). However,
`an attempt to antagonize the diuretic effect of 500 ␮g of
`GLP-1 with exendin(9-39) (100 ␮g/animal) was only partial-
`ly successful. Thus, exendin(9-39) completely abolished
`anorectic GLP-1 actions, whereas the diuretic actions of
`500 ␮g GLP-1 were only partially abolished (Fig. 2).
`Experiment 2
`Effects of a single dose of NN2211 on feeding behav-
`ior in rats. The acute effects of three doses of NN2211 on
`nighttime feeding were tested in a random crossover
`experiment (10, 50, and 200 ␮g/kg). The dose dependence
`of NN2211 on overnight food intake in normal rats is
`illustrated in Fig. 3. Two hours after onset of the feeding
`
`FIG. 2. Acute intravenous administration of GLP-1 causes profound
`diuresis. *, statistically significant difference from all other groups; a,
`statistically significant differences from vehicle-treated animals (P <
`0.05 as determined by ANOVA followed by Scheffe’s post hoc analysis).
`
`2532
`
`DIABETES, VOL. 50, NOVEMBER 2001
`
`MPI EXHIBIT 1086 PAGE 3
`
`

`

`P.J. LARSON AND ASSOCIATES
`
`FIG. 4. Subchronic administration of NN2211 dose-dependently de-
`creases body weight in both normal adult male Wistar rats and MSG-
`treated rats. Animals received two daily subcutaneous injections of
`NN2211 (100 or 200 ␮g/kg) or vehicle for 10 days followed by a 5-day
`recovery period. Body weight was monitored each morning. Values are
`mean ⴞ SE (n ⴝ 5– 8). *, significant difference from relevant vehicle-
`treated control as determined by ANOVA followed by Bonferroni
`multiple comparison post hoc analysis.
`
`ited food intake in normal rats (control: 22.4 ⫾ 0.7; 50
`␮g/kg: 16.3 ⫾ 0.7; 200 ␮g/kg: 8.7 ⫾ 1.3 g of food) as well as
`in MSG-treated rats (control: 12.5 ⫾ 1.9; 50 ␮g/kg: 10.0 ⫾
`1.0; 200 ␮g/kg: 5.8 ⫾ 0.7 g of food).
`Effects of a single dose of NN2211 on water intake
`and diuresis. In the same experiment, water intake and
`diuresis was monitored (Fig. 3). Both 50 and 200 ␮g/kg
`NN2211 significantly inhibited overnight (t720) water in-
`take in normal and MSG-treated rats (vehicle: 32.8 ⫾ 2.0;
`50 ␮g/kg: 21.1 ⫾ 1.1; 200 mg/kg: 11.5 ⫾ 2.5 g; vehicle-MSG:
`21.2 ⫾ 2.5; MSG-50 ␮g/kg: 17.1 ⫾ 1.9; MSG-200 ␮g/kg:
`11.5 ⫾ 1.3 ml; P ⬍ 0.05 as determined by ANOVA followed
`by post hoc Scheffe’s analysis). In contrast, the highest
`dose of NN2211 (200 ␮g/kg) significantly increased diure-
`sis in both normal and MSG-treated rats (vehicle: 7.6 ⫾ 0.3;
`200 ␮g/kg: 13.7 ⫾ 1.7; vehicle-MSG: 9.0 ⫾ 1.2; MSG-200
`␮g/kg: 12.3 ⫾ 0.9 ml; P ⬍ 0.05 as determined by ANOVA
`followed by post hoc Scheffe’s analysis).
`Experiment 3
`Effect of subchronic administration of NN2211 on
`food intake and body weight. Two daily injections of
`NN2211 to adult male Wistar rats dose-dependently de-
`creased body weight over the entire 10-day treatment
`period with significant differences obtained with the 200
`␮g/kg b.i.d. dosing regimen between treatment days 7 and
`14 (Fig. 4). Loss in body weight was preceded by de-
`creased food and water intake and increased diuresis (Fig.
`5). Food intake was significantly lower during the initial 3
`days of treatment for normal animals that were treated
`with 100 ␮g/kg b.i.d. (data not shown). Thereafter, the
`anorectic effect of this low dose was no longer statistically
`significant. In animals that were treated with 200 ␮g/kg
`b.i.d., food intake was significantly lower throughout the
`10-day treatment period. After cessation of the treatment,
`food intake normalized within a few days and body weight
`
`2533
`
`FIG. 3. Single subcutaneous injections of NN2211 dose-dependently
`inhibit food and water in both normal and MSG-treated rats. Dose-
`response curves for 720 min of food and water intake as well as diuresis
`in normal and MSG-treated rats receiving a single subcutaneous dose
`of NN2211. *P < 0.05 versus vehicle (ANOVA followed by Scheffe’s post
`hoc analysis); aP < 0.05 versus 10 ␮g/kg NN2211 (ANOVA followed by
`Scheffe’s post hoc analysis); bP < 0.05 versus 50 mg/kg NN2211
`followed by Scheffe’s post hoc analysis).
`
`session, 200 ␮g/kg NN2211 significantly inhibited food
`intake (3.8 ⫾ 0.3 vs. 5.2 ⫾ 0.3 g of food). The following
`morning (t720), both 50 and 200 ␮g/kg significantly inhib-
`DIABETES, VOL. 50, NOVEMBER 2001
`
`MPI EXHIBIT 1086 PAGE 4
`
`

`

`WEIGHT LOSS THERAPY WITH GLP-1 DERIVATIVE
`
`FIG. 5. The effect of subchronic administration of 200 ␮g/kg NN2211 to normal male Wistar rats (circles) or MSG-treated rats (boxes) on food
`and water intake as well as diuresis and feces excretion. Open symbols represent vehicle-treated animals; closed symbols represent
`NN2211-treated animals. Shaded areas reflect baseline values obtained from all animals in the group throughout the week before onset of
`treatment.
`
`2534
`
`DIABETES, VOL. 50, NOVEMBER 2001
`
`MPI EXHIBIT 1086 PAGE 5
`
`

`

`TABLE 1
`NN2211 treatment (200 ␮g/kg BID) exerts no adverse effects on plasma glucose, potassium, sodium, and plasma variables that reflect
`renal function (creatinine, carbamide, and protein)
`
`Normal vehicle
`(n ⫽ 8)
`
`Normal NN2211
`(n ⫽ 8)
`
`MSG vehicle
`(n ⫽ 4)
`
`MSG NN2211
`(n ⫽ 8)
`
`P.J. LARSON AND ASSOCIATES
`
`Treatment values (day 7)
`Glucose (mmol/l)
`Potassium (K⫹) (mmol/l)
`Sodium (Na⫹) (mmol/l)
`Creatinine (mmol/l)
`Carbamide (mmol/l)
`Protein (g/l)
`Recovery-phase values (day 14)
`Glucose (mmol/l)
`Potassium (K⫹) (mmol/l)
`Sodium (Na⫹) (mmol/l)
`Creatinine (mmol/l)
`Carbamide (mmol/l)
`Protein (g/l)
`
`Data are means ⫾ SD.
`
`6.7 ⫾ 0.5
`4.6 ⫾ 0.3
`136.5 ⫾ 0.4
`87.6 ⫾ 1.7
`8.4 ⫾ 0.2
`64.0 ⫾ 1.2
`
`6.8 ⫾ 0.1
`5.2 ⫾ 0.5
`139.5 ⫾ 0.6
`85.3 ⫾ 1.2
`7.9 ⫾ 0.2
`67.2 ⫾ 1.2
`
`6.9 ⫾ 0.2
`4.2 ⫾ 0.1
`136.9 ⫾ 0.6
`75.6 ⫾ 2.4
`8.9 ⫾ 0.5
`63.4 ⫾ 1.2
`
`6.4 ⫾ 0.2
`4.6 ⫾ 0.3
`138.7 ⫾ 0.4
`81.9 ⫾ 1.4
`8.0 ⫾ 0.1
`65.0 ⫾ 0.6
`
`6.1 ⫾ 1.0
`4.3 ⫾ 0.2
`138.2 ⫾ 0.5
`76.3 ⫾ 3
`7.3 ⫾ 0.3
`65.4 ⫾ 1.0
`
`5.7 ⫾ 1.0
`4.3 ⫾ 0.4
`141.4 ⫾ 0.7
`85.0 ⫾ 1.1
`7.9 ⫾ 0.3
`66.3 ⫾ 0.7
`
`6.7 ⫾ 0.3
`4.1 ⫾ 0.1
`139.0 ⫾ 0.3
`77.5 ⫾ 1.9
`8.5 ⫾ 0.3
`64.1 ⫾ 0.6
`
`6.2 ⫾ 0.2
`5.0 ⫾ 0.1
`140.2 ⫾ 1.2
`90.4 ⫾ 3.9
`7.3 ⫾ 0.2
`64.3 ⫾ 0.7
`
`gradually increased toward that of vehicle-treated animals
`(Fig. 5).
`Similar effects of NN2211 treatment on food intake were
`seen in obese MSG-treated rats. Thus, two daily injections
`of 200 ␮g/kg NN2211 significantly decreased body weight
`throughout the 10-day treatment period, whereas the 100
`␮g/kg b.i.d. dosing regimen displayed a trend toward
`weight reduction without reaching statistical significance.
`Also, food intake was significantly lower in MSG-treated
`animals that were treated with 200 ␮g/kg b.i.d. NN2211.
`Effect of subchronic administration of NN2211 on
`water intake, diuresis, and feces excretion. In con-
`trast to food intake, water intake was lower in NN2211-
`treated animals only at the initial days of treatment,
`because from day 4 onward, NN2211-treated groups dis-
`played considerably higher water intake than correspond-
`ing vehicle-treated groups (Fig. 5). However, these effects
`were not statistically significant and had no impact on
`long-term body fluid homeostasis (see below).
`In normal rats that were receiving 100 ␮g/kg b.i.d.
`NN2211,
`increased diuresis was accompanied by in-
`creased water intake from treatment day 2 onward to
`cessation of dosing (data not shown). In animals that were
`receiving 200 ␮g/kg b.i.d., however, fluid homeostasis was
`severely affected during the first 2 days of dosing, because
`a state of very low water intake coexisted with markedly
`increased diuresis (Fig. 5). For normal rats that were
`treated with 200 ␮g/kg b.i.d., apparent fluid balance with
`water consumption and diuresis at levels similar to vehi-
`cle-treated animals occurred from day 4 onward. Despite
`marked increasing effects on urinary water excretion,
`plasma sodium and potassium remained unaffected in
`animals that were treated with 200 ␮g/kg b.i.d. NN2211
`(Table 1). Also, plasma variables reflecting renal function
`(creatinine, carbamide, and total protein) were unaffected
`by NN2211 treatment (Table 1). Feces excretion was
`followed in normal animals that were receiving 200 ␮g/kg
`b.i.d. and was observed to decrease during administration
`of NN2211 coincident with the decrease in food intake
`(Fig. 5).
`In MSG-treated rats, NN2211 treatment (both 100 and
`200 ␮g/kg b.i.d.) induced a statistically significant reduc-
`
`tion of water intake and increased diuresis, leading to an
`initial negative water balance (Fig. 5). The compensatory
`water homeostatic mechanisms were clearly more undu-
`lating in MSG-treated animals that were treated with 200
`␮g/kg b.i.d. because they went through a phase character-
`ized by polyuria and hyperdipsia from day 3 to day 6
`before full compensation was obtained (Fig. 5). In the
`recovery phase after cessation of
`the treatment,
`the
`slightly lower water intake that accompanied lower food
`intake at the end of the NN2211 treatment period rapidly
`returned to control levels. As with normal animals, MSG-
`treated animals’ plasma electrolyte levels were unaffected
`by NN2211 treatment both during and after cessation of
`drug administration (Table 1).
`Experiment 4
`Effect of subchronic administration of NN2211 on
`EE, body composition,
`food intake, and body
`weight. As seen in experiment 3, 7 days of NN2211
`treatment significantly lowered body weight (373.3 ⫾ 14.7
`vs. 417.3 ⫾ 15.3 g), and the body weights in the pair-fed
`group were reduced similarly (378.5 ⫾ 10.5). After 7 days
`of treatment, EE was considerably lower in both NN2211
`and pair-fed animals (control 1,775 ⫾ 39; pair fed 1,634 ⫾
`49; NN2211 1,641 ⫾ 27 kcal/h; n ⫽ 6 –7, average of 3-h
`measurements). However, when EE was expressed as
`oxygen consumption per kilogram of body mass, no such
`differences were seen throughout the observation period
`(Fig. 6). Also, the RER was unaffected by 7 days of NN2211
`treatment (Fig. 7). In contrast, pair-fed animals displayed a
`switch toward lipid metabolism, probably reflecting that
`these animals had been starved for a longer period of time
`before the onset of experiment.
`Body composition analysis was carried out using a
`DEXA scanner specialized for small animals. Treatment
`with NN2211 reduced body weight by affecting both lean
`and adipose tissue mass, although the loss of fat mass did
`not quite reach statistical significance (Table 2). However,
`the percentages of lean and adipose tissue mass of total
`body weight changed in neither NN2211-treated nor pair-
`fed animals. The hydration status after 7 days of treatment
`was further assessed by measuring the hematocrit, and
`pair-fed animals displayed significant hemoconcentration
`
`DIABETES, VOL. 50, NOVEMBER 2001
`
`2535
`
`MPI EXHIBIT 1086 PAGE 6
`
`

`

`WEIGHT LOSS THERAPY WITH GLP-1 DERIVATIVE
`
`FIG. 6. Oxygen consumption was determined by indirect calorimetry by
`the end of the study (day 7). The rats received treatment at time point
`0 (10:00 A.M.). The rats had no access to food or water while being
`subjected to indirect calorimetry. On each day of experimentation, an
`animal treated with the chemical uncoupler 2,4-dinitrophenol (DNP)
`was included as a “positive” instrument. The results are expressed as
`mean ⴞ SE
`
`in comparison to both ad libitum–fed controls and
`NN2211-treated animals (ad libitum 45.6 ⫾ 0.8; NN2211
`47.0 ⫾ 0.9; pair-fed 49.1 ⫾ 0.9%; n ⫽ 7).
`Effect of subchronic administration of NN2211 on
`plasma glucose and lipids. A number of biochemical
`plasma variables were assessed in animals that were
`subjected to either 7 or 10 days of NN2211 treatment
`(experiments 3 and 4). Data obtained from experiment 3
`showed no effect of NN2211 on

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