throbber
A Recombinant Human Glucagon-Like Peptide
`(GLP)-1–Albumin Protein (Albugon) Mimics
`Peptidergic Activation of GLP-1 Receptor–Dependent
`Pathways Coupled With Satiety, Gastrointestinal
`Motility, and Glucose Homeostasis
`
`Laurie L. Baggio,1 Qingling Huang,1 Theodore J. Brown,2 and Daniel J. Drucker1
`
`Peptide hormones exert unique actions via specific G
`protein– coupled receptors; however, the therapeutic
`potential of regulatory peptides is frequently compro-
`mised by rapid enzymatic inactivation and clearance
`from the circulation. In contrast, recombinant or cova-
`lent coupling of smaller peptides to serum albumin
`represents an emerging strategy for extending the cir-
`culating t1/2 of the target peptide. However, whether
`larger peptide-albumin derivatives will exhibit the full
`spectrum of biological activities encompassed by the
`native peptide remains to be demonstrated. We report
`that Albugon, a human glucagon-like peptide (GLP)-1–
`albumin recombinant protein, activates GLP-1 receptor
`(GLP-1R)-dependent cAMP formation in BHK-GLP-1R
`cells, albeit with a reduced half-maximal concentration
`(EC50) (0.2 vs. 20 nmol/l) relative to the GLP-1R agonist
`exendin-4. Albugon decreased glycemic excursion and
`stimulated insulin secretion in wild-type but not GLP-
`1Rⴚ/ⴚ mice and reduced food intake after both intrace-
`rebroventricular and intraperitoneal administration.
`Moreover, intraperitoneal injection of Albugon inhib-
`ited gastric emptying and activated c-FOS expression in
`the area postrema, the nucleus of the solitary tract, the
`central nucleus of the amygdala, the parabrachial, and
`the paraventricular nuclei. These findings illustrate
`that peripheral administration of a larger peptide-albu-
`min recombinant protein mimics GLP-1R– dependent ac-
`tivation of central and peripheral pathways regulating
`energy intake and glucose homeostasis in vivo. Diabetes
`53:2492–2500, 2004
`
`From the 1Department of Medicine, Banting and Best Diabetes Centre,
`Toronto General Hospital, Toronto, Ontario, Canada; and the 2Division of
`Reproductive Science, Samuel Lunenfeld Research Institute, Mount Sinai
`Hospital, and the University of Toronto, Toronto, Ontario, Canada.
`Address correspondence and reprint requests to Dr. Daniel J. Drucker,
`Toronto General Hospital, Banting and Best Diabetes Centre, 200 Elizabeth
`St., MBRW4R-402, Toronto, Canada M5G 2C4. E-mail: d.drucker@utoronto.ca.
`Received for publication 4 December 2003 and accepted in revised form
`28 May 2004.
`CCK, cholecystokinin; CNS, central nervous system; DPP-IV, dipeptidyl
`peptidase-IV; Ex-4, exendin-4; GLP, glucagon-like peptide; GLP-1R, GLP-1
`receptor; HSA, human serum albumin; ICV, intracerebroventricular; IP, intra-
`peritoneal; NTS, nucleus of the solitary tract.
`© 2004 by the American Diabetes Association.
`
`2492
`
`Glucagon-like peptide (GLP)-1 is a 30 –amino
`
`acid peptide hormone secreted from gut endo-
`crine cells in response to nutrient ingestion that
`promotes nutrient assimilation through regula-
`tion of gastrointestinal motility and islet hormone secre-
`tion (1). Infusion of GLP-1 into normal or diabetic human
`subjects stimulates insulin and inhibits glucagon secre-
`tion, thereby indirectly modulating peripheral glucose
`uptake and control of hepatic glucose production (2).
`GLP-1 also produces anorectic effects, and short-term
`infusion of GLP-1 is associated with diminished appetite
`and reduced energy consumption in normal, obese, and
`diabetic human subjects (3). Taken together, the actions of
`GLP-1 to reduce glycemia while preventing concomitant
`weight gain have attracted considerable interest in phar-
`maceutical approaches to enhancing GLP-1 action for the
`treatment of type 2 diabetes.
`A major challenge for the therapeutic use of regulatory
`peptides, including native GLP-1, is a short circulating t1/2,
`due principally to rapid enzymatic inactivation and/or re-
`nal clearance. Although infusion of native GLP-1 is highly
`effective in lowering blood glucose in subjects with type 2
`diabetes, a single subcutaneous injection of the native pep-
`tide is quickly degraded and disappears from the circulation
`within minutes (4). Hence, the majority of pharmaceutical
`approaches to the development of GLP-1 mimetic agents
`have focused on the development of long-acting degrada-
`tion-resistant peptides (5). The naturally occurring lizard
`salivary gland peptide exendin-4 (Ex-4) is a potent GLP-1
`receptor (GLP-1R) agonist and exhibits therapeutic efficacy
`in studies of patients with type 2 diabetes (6). Similarly, a
`fatty acylated human GLP-1 analog, liraglutide, exhibits a
`more sustained duration of action and potently reduces
`glycemic excursion in diabetic subjects (7).
`Although degradation-resistant GLP-1R agonists appear
`to be promising agents for the treatment of diabetes, the
`need for once- or twice-daily injection of these peptides
`has fostered complementary efforts directed at identifica-
`tion of more potent longer-acting agents with sustained
`efficacy in vivo. Given the long circulating t1/2 of albumin-
`linked drugs (8), a GLP-1–albumin protein should exhibit a
`much more prolonged circulating t1/2 and hence requires a
`
`DIABETES, VOL. 53, SEPTEMBER 2004
`
`MPI EXHIBIT 1007 PAGE 1
`
`

`

`reduced frequency of parenteral administration, relative to
`native GLP-1. Nevertheless, the molecular interaction with
`the GLP-1R, volume of distribution, and access to the
`central nervous system (CNS) would be predicted to be
`markedly different for a much larger albumin-based mole-
`cule (8).
`Whether all of the desirable actions of native GLP-1,
`including the activation of the CNS centers regulating food
`intake and gastrointestinal motility, would be mimicked by
`a much larger GLP-1–albumin protein is currently unclear.
`Accordingly, we have examined the biological actions of
`Albugon, a recombinant GLP-1– human serum albumin
`(HSA) fusion protein, using a combination of cell line stud-
`ies in vitro and both wild-type and GLP-1R⫺/⫺ mice in vivo.
`
`RESEARCH DESIGN AND METHODS
`Reagents. Tissue culture medium, serum, and G418 were purchased from
`Invitrogen (San Diego, CA). Forskolin and 3-isobutyl-1-methylxanthine were
`obtained from Sigma (St. Louis, MO). HSA and Albugon were provided by
`Human Genome Sciences (Rockville, MD). Ex-4 and exendin (9-39) were
`purchased from California Peptide Research (Napa, CA).
`Measurement of cAMP. Baby hamster kidney (BHK) cells stably transfected
`with rat GLP-1R were generated and propagated in medium containing 0.05
`mg/ml G418 as described previously (9). Before analysis, BHK-GLP-1R cells
`were grown to 70 – 80% confluence in 24-well plates in the absence of G418 at
`37°C. Ex-4 and exendin (9-39) were dissolved in PBS, and cells were treated
`with control or test reagents in Dulbecco’s modified Eagle’s medium contain-
`ing serum and 100 ␮mol/l 3-isobutyl-1-methylxanthine. Cells were incubated
`with 1 ␮mol/l exendin (9-39) or medium alone for 5 min at 37°C, followed by
`an additional 10-min incubation in the presence of increasing concentrations
`of Ex-4, HSA, or Albugon. All reactions were carried out in triplicate and
`terminated by the addition of ice-cold absolute ethanol. Cell extracts were
`collected and stored at ⫺80°C until assayed. For cAMP determinations,
`aliquots of ethanol extracts were lyophilized, and cAMP levels were measured
`using a cAMP radioimmunoassay kit (Biomedical Technologies, Stoughton, MA).
`Mice. All animal experiments were carried out in accordance with protocols
`and guidelines approved by the Toronto General Hospital Animal Care
`Committee. GLP-1R⫺/⫺ mice on the C57BL/6 genetic background, and age-
`matched (8- to 15-week-old male) wild-type C57BL/6 mice (Charles River
`Laboratories, Montreal, PQ) were maintained on a 12-h light/dark cycle and
`allowed free access to standard rodent food and water, except where noted.
`Wild-type mice were acclimated to the animal facility for a minimum of 1 week
`before analysis.
`Glucose tolerance tests and measurement of plasma insulin. Oral or
`intraperitoneal (IP) glucose tolerance tests were carried out after an overnight
`fast (16 –18 h). Glucose (1.5 mg/g body wt) was administered orally through a
`gavage tube or by injection into the peritoneal cavity. A blood sample was
`drawn from the tail vein at 0, 10, 20, 30, 60, 90, and 120 min after glucose
`administration, and blood glucose levels were measured using a Glucometer
`Elite blood glucose meter (Bayer, Toronto, Ontario, Canada). For plasma
`insulin determination, a blood sample (100 ␮l) was removed from the tail vein
`during the 10- to 20-min time period after glucose administration and
`immediately mixed with a 10% volume of a chilled solution containing 5,000
`KIU/ml Trasylol (Bayer), 32 mmol/l EDTA, and 0.1 nmol/l Diprotin A (Sigma).
`Plasma was separated by centrifugation at 4°C and stored at ⫺80°C until
`assayed. Plasma insulin levels were measured using a rat insulin enzyme-
`linked immunosorbent assay kit (Crystal Chem, Chicago, IL) with mouse
`insulin as standard.
`Measurement of plasma glucagon. After an overnight fast (16 h), mice were
`given an IP injection of 1 mg/kg HSA or Albugon. At 20 min after HSA or
`Albugon administration, mice were killed and cardiac blood was obtained and
`mixed with a 10% volume of a chilled solution containing 5,000 KIU/ml
`Trasylol, 32 mmol/l EDTA, and 0.1 nmol/l Diprotin A. Plasma was separated by
`centrifugation at 4°C and stored at ⫺80°C until assayed. Plasma glucagon
`levels were measured using a glucagon radioimmunoassay kit (Linco Re-
`search, St. Charles, MO).
`Feeding studies. For analysis of food intake, mice were fasted overnight
`(16 –18 h), and control (PBS or HSA) or test (Ex-4 or Albugon) reagents were
`administered by intracerebroventricular (ICV) or IP injection. For ICV injec-
`tions, mice were lightly anesthetized using isoflurane inhalation (Abbott
`Laboratories, Saint-Laurent, Quebec, Canada), and reagents were adminis-
`tered in a total volume of 5 ␮l by injection into the lateral ventricles using a
`2.5 mm ⫻ 30-gauge needle attached to a Hamilton syringe as described (10).
`
`L.L. BAGGIO AND ASSOCIATES
`
`Mice were allowed to recover from the anesthetic (⬃10 min) before assess-
`ment of food intake. For IP injections, 100 ␮l control or test reagent was
`injected into the peritoneal cavity. Immediately after ICV or IP injection, mice
`were weighed and then placed into individual cages containing preweighed
`rodent food, with free access to water. At 2, 4, 7, and 24 h after reagent
`administration, the food was reweighed and total food intake (g/g of body wt)
`was calculated.
`Gastric emptying. The gastric emptying rate was determined as described
`(11). Briefly, mice were fasted overnight (18 h) and then allowed free access
`to preweighed rodent food for 1 h. After the 1-h refeeding, the remaining food
`was weighed and food intake was determined. Mice were then given IP
`injections of PBS, Ex-4 (0.17 mg/kg), HSA (2.7 mg/kg), Albugon (3 mg/kg), or
`cholecystokinin (CCK)-8 (4 ␮g) and deprived of food for an additional 4 h.
`Mice were anesthetized with Somnotol (sodium pentobarbital solution; MTC
`Pharmaceuticals, Cambridge, Ontario, Canada), their stomachs were re-
`moved, and stomach content wet weight was determined. Gastric emptying
`rate was calculated using the following: gastric emptying rate (%) ⫽ [1 ⫺
`(stomach content wet weight/food intake)] ⫻ 100.
`Measurement of c-FOS activation in the murine CNS. The number of
`c-FOS immunoreactive neurons in specific brain regions was assessed quan-
`titatively in both wild-type C57BL/6 and GLP-1R⫺/⫺ mice as described (12,13).
`Briefly, animals were given IP injections of PBS, Ex-4, HSA, or Albugon in a
`100 ␮l volume. At 10 and 60 min after injection, mice were anesthetized with
`Somnotol. All mice were perfused intracardially with ice-cold normal saline
`followed by 4% paraformaldehyde solution. Brains were removed immediately
`at the end of perfusion, kept in ice-cold 4% paraformaldehyde solution for 3
`days, and then transferred to a solution containing paraformaldehyde and 10%
`sucrose for 12 h. Brains were cut into 25-␮m sections using a Leica SM2000R
`sliding microtome (Leica Microsystems, Richmond Hill, Ontario, Canada) and
`stored at ⫺20°C in a cold cryoprotecting solution. Sections were processed
`for immunocytochemical detection of FOS using a conventional avidin-biotin-
`immunoperoxidase method (Vectastain ABC Elite Kit; Vector Laboratories,
`Burlingame, CA) as described (12). The FOS antibody (Sigma-Aldrich,
`Oakville, Ontario, Canada) was used at a 1:50,000 dilution. Brain sections
`corresponding to the level of the area postrema, the nucleus of the solitary
`tract (NTS), the central nucleus of the amygdala, and the parabrachial and
`paraventricular nuclei were defined according to the mouse brain atlas of
`Franklin and Paxinos (14) and selected for analyses.
`Statistical analysis. All data are presented as means ⫾ SE. Statistical
`significance was determined by ANOVA and Bonferroni post-test using Prism
`version 3.03 software (GraphPad Software, San Diego, CA). A P value ⬍0.05
`was considered to be statistically significant.
`
`RESULTS
`Albugon is a recombinant human protein that contains a
`dipeptidyl peptidase-IV (DPP-IV)-resistant human GLP-1
`analog encoded in the same open reading frame as the
`HSA amino acid sequence. To evaluate whether the bioac-
`tive domain(s) of a much smaller peptide hormone could
`still recognize and functionally interact with its cognate
`receptor when constrained within a much larger heterol-
`ogous protein, we examined whether Albugon activated
`GLP-1R in vitro. Albugon produced a dose-dependent
`stimulation of cAMP accumulation in BHK-GLP-1R cells
`that was significantly diminished by coincubation with the
`GLP-1R antagonist exendin (9-39) (Fig. 1). Nevertheless,
`Albugon was not as effective in activating the GLP-1R
`when compared with the much smaller more potent
`lizard-derived GLP-1R agonist Ex-4 (Fig. 1; EC50 ⫽ 0.2 vs.
`20 nmol/l for Ex-4 vs. Albugon, respectively).
`These experiments demonstrate that the considerably
`altered peptide conformation that arises after insertion of
`GLP-1 sequences into a much larger albumin open reading
`frame does not eliminate the ability of essential GLP-1
`motif(s) to recognize and activate GLP-1Rs. To ascertain
`whether circulating Albugon is capable of reaching key
`sites and activating GLP-1R– dependent actions in vivo, we
`carried out oral glucose tolerance testing in mice. Glyce-
`mic excursion was significantly reduced after parenteral
`Albugon administration to wild-type mice (Fig. 2A). Further-
`
`DIABETES, VOL. 53, SEPTEMBER 2004
`
`2493
`
`MPI EXHIBIT 1007 PAGE 2
`
`

`

`ACTIONS OF A RECOMBINANT GLP-1 ALBUMIN PROTEIN
`
`FIG. 1. Albugon exhibits similar efficacy but lower potency than Ex-4 at
`the rat GLP-1R in vitro. A stable BHK cell line expressing the rat
`GLP-1R was pretreated for 5 min with 1 ␮mol/l exendin (9-39) [Ex(9-
`39)] or medium alone before a 10-min treatment with increasing
`concentrations of Ex-4, Albugon, or HSA. cAMP levels in lyophilized
`aliquots of cell extracts were measured by radioimmunoassay and used
`to calculate total cAMP content per well. Values are expressed as
`means ⴞ SE and are representative of data from two independent
`experiments, each performed in triplicate. *P < 0.05, **P < 0.01,
`***P < 0.001 for Ex-4 – vs. Albugon-treated cells.
`
`more, the glucose-lowering properties of Albugon depend-
`ed on a functional GLP-1 receptor because Albugon had no
`effect on blood glucose in GLP-1R⫺/⫺ mice (Fig. 2B). Con-
`sistent with the known actions of smaller GLP-1R peptide
`agonists, Albugon, at doses ranging from 0.1 to 10 mg/kg,
`markedly reduced glycemic excursion after not only oral but
`also IP glucose loading, in association with a significant
`increase in the insulin-to-glucose ratios (Fig. 3A and B).
`Albugon increased the levels of plasma insulin after an IP
`glucose challenge (Fig. 3C) but did not significantly reduce
`the levels of plasma glucagon in mice after an overnight
`fast (Fig. 3D). Taken together, these results demonstrate
`that Albugon lowers blood glucose and enhances insulin
`secretion through GLP-1R–dependent mechanisms in vivo.
`Smaller 30 – to 40 –amino acid GLP-1R agonists, includ-
`ing native GLP-1, Ex-4, and liraglutide, have been shown to
`inhibit food intake, presumably via activation of central
`hypothalamic centers regulating satiety (15). To assess
`whether a much larger recombinant GLP-1–albumin pro-
`tein would also exert anorectic effects, we studied food
`intake in normal mice after ICV or IP peptide administra-
`tion. ICV Albugon significantly lowered food intake in
`mice (relative to the HSA control) in a dose-dependent
`manner, and this effect was detectable for up to 7 h (Fig.
`4A) but was not sustained over the entire 24-h observation
`period. In contrast, Ex-4 exerted a more potent and
`sustained anorectic effect after ICV administration, with a
`highly significant reduction of food intake observed even
`at the end of the 24-h study period (Fig. 3).
`The blood-brain barrier is relatively impermeable to
`larger proteins such as albumin under both normal phys-
`iological conditions and in the setting of diabetes (16,17).
`Although GLP-1Rs are expressed on hypothalamic neurons
`regulating satiety (18,19), whether GLP-1R agonists re-
`quire direct access to the brain for activation of the central
`anorexic pathway remains uncertain (20). Accordingly, we
`examined whether Albugon would also exhibit anorectic
`effects after peripheral administration. Although lower
`doses of IP Albugon, 0.16 –11 nmol/kg, did not affect food
`
`intake, the highest dose tested (110 nmol/kg) reduced food
`intake at all time points studied, from 2 to 24 h after
`Albugon administration (Fig. 4C). In contrast, much small-
`er doses of Ex-4 significantly reduced food ingestion af-
`ter IP administration in the same experiments (Fig. 4D).
`Hence, although Albugon is capable of exerting anorectic
`actions after both central and peripheral administration, it
`is less potent compared with the smaller GLP-1R agonist,
`Ex-4. To ascertain whether the anorectic actions of periph-
`erally administered Albugon were due to the nonspecific
`effects of the larger amount of injected protein, we re-
`peated the experiments using age- and sex-matched wild-
`type control and GLP-1R⫺/⫺ mice. Albugon reduced food
`intake at all time points in wild-type mice but had no effect
`on food intake in GLP-1R⫺/⫺ mice. These findings demon-
`strate that IP Albugon inhibits food intake in a GLP-1R–
`dependent manner.
`GLP-1R agonists markedly inhibit gastric emptying (21),
`and gastric distension induces c-FOS in GLP-1– expressing
`neurons in the rat medulla (22). However, whether direct
`CNS access is required for either GLP-1R– dependent
`inhibition of food intake or gastric emptying has not been
`determined. The basal rate of gastric emptying was com-
`parable in wild-type and GLP-1R⫺/⫺ mice (Fig. 5). Both
`Ex-4 and CCK-8 significantly inhibited gastric emptying in
`wild-type mice; however, CCK-8, but not Ex-4, also inhib-
`ited gastric emptying in GLP-1R⫺/⫺ mice. Similarly, Al-
`bugon potently inhibited gastric emptying in wild-type but
`not in GLP-1R⫺/⫺ mice (Fig. 5).
`The anorectic actions of small peptide GLP-1R agonists
`are associated with c-FOS activation in CNS centers
`
`FIG. 2. Albugon lowers blood glucose in wild-type but not in GLP-1Rⴚ/ⴚ
`mice. Oral glucose tolerance in wild-type GLP-1Rⴙ/ⴙ (A) and GLP-1Rⴚ/ⴚ
`(B) mice given IP injections of 5 mg/kg of HSA (E) or Albugon (ALB; f)
`60 min before an oral glucose load. Values are expressed as means ⴞ
`SE; n ⴝ 5– 6 mice/group. **P < 0.01, ***P < 0.001 for Albugon- vs.
`HSA-treated mice.
`
`2494
`
`DIABETES, VOL. 53, SEPTEMBER 2004
`
`MPI EXHIBIT 1007 PAGE 3
`
`

`

`L.L. BAGGIO AND ASSOCIATES
`
`FIG. 3. Albugon dose-dependently lowers the
`glucose excursion and increases plasma insulin
`and insulin-to-glucose ratios after an oral or
`peripheral glucose challenge. Oral glucose toler-
`ance test (A) and IP glucose tolerance test (B) in
`wild-type mice treated with different doses of IP
`HSA or Albugon (ALB) 60 min before a glucose
`load. The bottom panel in A and B indicates the
`plasma insulin-to-glucose ratios (ng/ml:mmol/l)
`at the 20-min time point after glucose adminis-
`tration in HSA- or ALB-treated mice. C: Plasma
`insulin levels at the 20-min time point after IP
`glucose administration in mice treated with 1
`mg/kg HSA (䡺) or ALB (f). D: Plasma glucagon
`levels in fasted mice at 20 min after IP adminis-
`tration of 1 mg/kg HSA (䡺) or ALB (f). Values
`are expressed as means ⴞ SE; n ⴝ 5–12 mice/
`group. *P < 0.05, **P < 0.01, ***P < 0.001 for
`Albugon- vs. HSA-treated mice.
`
`coupled with the control of energy intake (18,20,23). To
`determine whether peripherally administered Albugon
`was capable of activating neuronal FOS expression, we
`examined the pattern and extent of CNS FOS expression
`after IP Albugon administration. Ex-4 markedly increased
`c-FOS expression in the area postrema, the NTS, the
`central nucleus of the amygdala, the parabrachial nucleus,
`and the hypothalamic paraventricular nuclei (Fig. 6A–E).
`Similarly, Albugon significantly activated c-FOS expres-
`sion in the identical brain regions, although much less
`robustly in the NTS and paraventricular nucleus than in
`
`Ex-4 (Fig. 6A–E). Analysis of the murine CNS after periph-
`eral (IP) administration of HSA did not detect immuno-
`reactive HSA in brain parenchyma (data not shown),
`consistent with the inability of HSA to rapidly cross the
`normal blood-brain barrier (8).
`
`DISCUSSION
`GLP-1– based therapies for type 2 diabetes are attracting
`increasing attention in part because of the preliminary
`efficacy demonstrated in clinical studies (6) and because
`
`DIABETES, VOL. 53, SEPTEMBER 2004
`
`2495
`
`MPI EXHIBIT 1007 PAGE 4
`
`

`

`ACTIONS OF A RECOMBINANT GLP-1 ALBUMIN PROTEIN
`
`FIG. 4. Albugon (ALB) reduces food intake in
`fasted mice but is less anorectic than Ex-4. After
`an overnight fast, wild-type mice were given ICV
`(A and B) or IP (C–E) injections of PBS or
`increasing doses (nmol/kg) of Ex-4, HSA, or ALB.
`E: Wild-type and GLP-1Rⴚ/ⴚ mice were fasted
`overnight and then given IP injections of 110
`nmol/kg of HSA or ALB. Food intake was mea-
`sured at 2, 4, 7, and 24 h after recovery from
`injection. Values are expressed as means ⴞ SE;
`n ⴝ 4 –5 mice/group. *P < 0.05, **P < 0.01, ***P <
`0.001 vs. control-treated (PBS or HSA) mice.
`
`of unique yet complementary mechanisms of action (1).
`Unlike some antidiabetic agents that reduce blood glucose
`while promoting weight retention, GLP-1R activation is
`coupled with short-term inhibition of food intake in both
`rodent and human studies (15). Moreover, prolonged
`GLP-1 administration for 6 weeks to diabetic human sub-
`jects was associated with a significant reduction in body
`
`weight over the study period (24). The anorectic proper-
`ties of GLP-1 and its peptide analogs are thought to be due
`in part to both inhibition of gastric emptying and activa-
`tion of central satiety centers coupled with reduction of
`energy intake (20,25). Although development of GLP-1–
`based small peptide analogs resistant to enzymatic inacti-
`vation is a major focus of current pharmaceutical activity
`
`2496
`
`FIG. 5. Albugon (ALB) reduces the
`gastric emptying rate in wild-type but
`not GLP-1Rⴚ/ⴚ mice. Gastric emptying
`rate in wild-type (WT) (A) and GLP-
`1Rⴚ/ⴚ (B) mice at 4 h after IP admin-
`istration of PBS, Ex-4 (0.17 mg/kg),
`HSA (2.7 mg/kg), ALB (3 mg/kg), or
`CCK octapeptide (CCK-8; 4 ␮g/
`mouse) is shown. Values are ex-
`pressed as means ⴞ SE; n ⴝ 3– 4 mice/
`group. *P < 0.05, ***P < 0.001 vs.
`PBS; ##P < 0.01 vs. HSA.
`
`DIABETES, VOL. 53, SEPTEMBER 2004
`
`MPI EXHIBIT 1007 PAGE 5
`
`

`

`L.L. BAGGIO AND ASSOCIATES
`
`FIG. 6. IP Albugon and Ex-4 increase c-FOS levels in the mouse CNS. Representative photomicrographs are shown of c-FOS–stained coronal brain
`sections of area postrema (AP) (A), NTS (B), hypothalamic parabrachial nucleus (PB) (C), central nucleus of the amygdala (CeA) (D), and
`paraventricular nucleus of the hypothalamus (PVH) (E) from wild-type mice at 60 min after IP injection of PBS, Ex-4 (11 nmol/kg), HSA (110
`nmol/kg), or Albugon (110 nmol/kg). No hypoglycemia was detected after administration of either Albugon or Ex-4 in these experiments. Original
`magnification, ⴛ200. CC, central canal; 3V, third ventricle. The number of c-FOS immunopositive (Fosⴙ) cells are depicted below the
`corresponding CNS section. Data are presented as means ⴞ SE; n ⴝ 3 mice/treatment. ***P < 0.001 for Ex-4 – vs. PBS-treated mice at 60 min; #P <
`0.05, ###P < 0.001 for Albugon- vs. HSA-treated mice at 60 min; ^^P < 0.01, ^^^P < 0.001 for Ex-4 – vs. Albugon-treated mice at 60 min.
`
`(1), the need for once- or twice-daily injection of these
`peptides has stimulated efforts toward development of
`even longer-acting molecules that retain the ability to
`activate GLP-1Rs.
`The relatively prolonged circulating t1/2 of endogenous
`and exogenously administered albumin has fostered at-
`tempts directed at coupling peptide moieties to albumin,
`thereby increasing the circulating t1/2 of the albumin-
`peptide complex (26). Albuferon is an interferon-␣–albu-
`min fusion protein that retains the biological activity of
`interferon yet exhibits a markedly extended pharmacoki-
`netic profile relative to native interferon in Cynomolgus
`monkeys (27). Similarly, fusion of the amino acid sequence
`of human growth hormone to albumin prolonged the
`circulating t1/2 of Albutropin relative to the native growth
`hormone, yet Albutropin retained the ability to stimulate
`IGF-I and body weight gain in vivo (28). Moreover, peptide
`binding to albumin extended the pharmacokinetic proper-
`ties of several smaller proteins including insulin (29), Fab
`antibody fragments (26), and coagulation factor VIIa
`inhibitor 1a (30). In contrast to Albugon, however, these
`albumin-peptide derivatives exert their predominant ac-
`tions outside the CNS, and activation of the CNS is not
`critical for their therapeutic actions.
`
`More recent efforts have been directed at extending the
`t1/2 of the much smaller GLP-1 molecule using albumin-
`based approaches. Liraglutide is a fatty acylated human
`DPP-IV–resistant GLP-1 analog that binds to albumin and
`exhibits a t1/2 of ⬃11–15 h after parenteral administration
`in humans (31). Liraglutide inhibits food ingestion in rats
`(32) and decreases gastric emptying in human subjects
`(33); hence, the transient GLP-1–albumin interaction does
`not preclude communication with CNS centers important
`for control of satiety and gastrointestinal motility. How-
`ever, liraglutide binds to albumin in a noncovalent disso-
`ciable manner, with the actions of liraglutide attributed to
`a free peptide unconstrained by its intermittent associa-
`tion with albumin in the circulation.
`We recently studied the properties of CJC-1131, a DPP-
`IV–resistant human GLP-1 analog that covalently couples
`with HSA after parenteral administration (34). CJC-1131
`binds to the GLP-1R and activates a broad spectrum of
`GLP-1R– dependent actions associated with glucose reduc-
`tion in db/db mice, including stimulation of insulin secre-
`tion and insulin gene expression and expansion of islet
`mass (34). Intriguingly, CJC-1131 also activates FOS ex-
`pression in hypothalamic neurons and reduces food intake
`and weight gain in normal and diabetic mice (23,34).
`
`DIABETES, VOL. 53, SEPTEMBER 2004
`
`2497
`
`MPI EXHIBIT 1007 PAGE 6
`
`

`

`ACTIONS OF A RECOMBINANT GLP-1 ALBUMIN PROTEIN
`
`FIG. 6. Continued.
`
`Nevertheless, because CJC-1131 is administered parenter-
`ally as the free GLP-1 analog, which subsequently forms a
`covalent linkage with albumin in vivo (34), it remains
`likely that some or all of the acute effects of CJC-1131 on
`the brain reflect the rapid initial actions of free CJC-1131
`before covalent coupling with albumin.
`In contrast, Albugon contains the sequences of human
`GLP-1 linked in the same open reading frame with recom-
`binant HSA; hence, no “free” GLP-1 is associated with
`Albugon administration in vivo. Consistent with studies of
`CJC-1131 covalently conjugated to albumin (34), Albugon
`activates the cloned GLP-1R, but with a reduced affinity
`relative to the potent GLP-1R agonist Ex-4. Because the
`blood-brain barrier is relatively impermeable to albumin,
`peripheral administration of the much larger Albugon
`protein provides an opportunity to determine the relative
`importance of peripheral versus central GLP-1R networks
`for control of satiety and gut motility. Remarkably, Alb-
`ugon inhibited food intake after not only ICV but also after
`IP administration in mice. Similarly, Albugon significantly
`inhibited gastric emptying after IP administration. Further-
`more, the distribution of neuronal c-FOS activation in
`different CNS nuclei after peripheral Albugon administra-
`tion was highly similar, albeit less robust, compared with
`the pattern of FOS activation observed after Ex-4. These
`findings support a model whereby peripheral activation of
`GLP-1R– dependent vagal afferents is capable of activating
`
`CNS centers, transducing the effects of GLP-1 in the brain
`(35,36)
`Given the clinical importance of the anorectic actions of
`GLP-1R agonists for prevention of weight gain in the
`treatment of diabetes, the mechanisms and pathways
`activated by GLP-1R agonists that converge on inhibition
`of feeding centers are of considerable interest. Both native
`GLP-1 and Ex-4 cross the blood-brain barrier through a
`GLP-1R–independent pathway (37–39); hence, these pep-
`tides are capable of directly penetrating and activating
`CNS centers after exogenous administration. In contrast,
`peripheral administration of much larger albumin-based
`GLP-1R agonists, such as CJC-1131 and Albugon, at doses
`that do not induce hypoglycemia, rapidly activates neuro-
`nal c-FOS in distinct brain regions. Furthermore, intrave-
`nous but not ICV CJC-1131 induced tyrosine hydroxylase
`gene transcription in the area postrema in vivo (23).
`Hence, our data suggest that peripheral activation of
`central GLP-1R systems coupled with regulation of FOS
`expression, gastric emptying, and food intake does not
`require direct exposure to GLP-1R agonists in the CNS, but
`may be achieved through activation of ascending path-
`ways coupled with central GLP-1R– dependent networks.
`The current data demonstrate that as little as 0.1 mg/kg
`(1.4 nmol/kg) of Albugon was sufficient for lowering of
`blood glucose; however, much larger doses (110 nmol/kg),
`were required for inhibition of food intake after IP admin-
`
`2498
`
`DIABETES, VOL. 53, SEPTEMBER 2004
`
`MPI EXHIBIT 1007 PAGE 7
`
`

`

`istration in mice. These findings imply that acute periph-
`eral Albugon administration is significantly more effective
`at reduction of glycemic excursion relative to inhibition of
`appetite. Nevertheless, the experimental design used here
`did not examine the anorectic effects of more prolonged
`sustained administration, a paradigm more representative
`of the potential therapeutic use of Albugon in humans. Fur-
`thermore, the t1/2 of circulating HSA, and by inference
`Albugon, is much longer in humans than in mice (8). Given
`the intense interest in the development of long-acting GLP-1R
`agonists for the treatment of type 2 diabetes, the biological
`properties and mechanisms of action of GLP-1–albumin
`derivatives merit further investigation.
`
`ACKNOWLEDGMENTS
`D.J.D. was supported in part by operating grants from the
`Juvenile Diabetes Research Foundation and the Canadian
`Diabetes Association and is a Senior Scientist of the
`Canadian Institutes for Health Research. Q.H. was sup-
`ported by a research fellowship from the Canadian Diabe-
`tes Association.
`We thank Human Genome Science scientists for the
`generous provision of Albugon and HSA used in these
`studies.
`
`REFERENCES
`1. Drucker DJ: Enhancing incretin action for the treatment of type 2 diabetes.
`Diabetes Care 26:2929 –2940, 2003
`2. Vella A, Shah P, Basu R, Basu A, Holst JJ, Rizza RA: Effect of glucagon-like
`peptide 1(7-36) amide on glucose effectiveness and insulin action in people
`with type 2 diabetes. Diabetes 49:611– 617, 2000
`3. Flint A, Raben A, Astrup A, Holst JJ: Glucagon-like peptide 1 promotes
`satiety and suppresses energy intake in humans. J Clin Invest 101:515–520,
`1998
`4. Deacon CF, Johnsen AH, Holst JJ: Degradation of glucagon-like peptide-1
`by human plasma in vitro yields an N-terminally truncated peptide that is
`a major endogenous metabolite in vivo. J Clin Endocrinol Metab 80:952–
`957, 1995
`5. Drucker DJ: Development of glucagon-like peptide-1-based pharmaceuti-
`cals as therapeutic agents for the treatment of diabetes. Curr Pharm Des
`7:1399 –1412, 2001
`6. Fineman MS, Bicsak TA, Shen LZ, Taylor K, Gaines E, Varns A, Kim DW,
`Baron AD: Effect on glycemic control of synthetic exendin-4 (AC2993)
`additive to existing metformin and/or sulfonylurea treatment in patients
`with type 2 diabetes. Diabetes Care 27:2370 –2377, 2003
`7. Chang AM, Jakobsen G, Sturis J, Smith MJ, Bloem CJ, An B,

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