throbber
See discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/10882242
`
`Development and Characterization of a Glucagon-Like Peptide 1-Albumin
`Conjugate The Ability to Activate the Glucagon-Like Peptide 1 Receptor In
`Vivo
`
`Article  in  Diabetes · March 2003
`
`DOI: 10.2337/diabetes.52.3.751 · Source: PubMed
`
`CITATIONS
`227
`
`8 authors, including:
`
`Jung Guk Kim
`Kyungpook National University
`
`90 PUBLICATIONS   2,168 CITATIONS   
`
`SEE PROFILE
`
`Dominique Bridon
`Independent Researcher
`
`47 PUBLICATIONS   1,178 CITATIONS   
`
`SEE PROFILE
`
`READS
`349
`
`Laurie Baggio
`Mount Sinai Hospital, Toronto
`
`81 PUBLICATIONS   10,632 CITATIONS   
`
`SEE PROFILE
`
`Corinne Benquet
`
`12 PUBLICATIONS   448 CITATIONS   
`
`SEE PROFILE
`
`Some of the authors of this publication are also working on these related projects:
`
`Study of mechanisms of action and the therapeutic potential of DPP-4 inhibition or invalidation in obesity and type 2 diabetes: View project
`
`Albumin conjugates as HIV fusion inhibitors View project
`
`All content following this page was uploaded by Jung Guk Kim on 13 October 2014.
`
`The user has requested enhancement of the downloaded file.
`
`MPI EXHIBIT 1033 PAGE 1
`
`

`

`Development and Characterization of a Glucagon-Like
`Peptide 1-Albumin Conjugate
`The Ability to Activate the Glucagon-Like Peptide 1
`Receptor In Vivo
`
`Jung-Guk Kim,1 Laurie L. Baggio,1 Dominique P. Bridon,2 Jean-Paul Castaigne,2 Martin F. Robitaille,2
`Lucie Jette´ ,2 Corinne Benquet,2 and Daniel J. Drucker1
`
`The rapid degradation of native glucagon-like peptide 1
`(GLP-1) by dipeptidyl peptidase-IV (DPP-IV) has fos-
`tered new approaches for generation of degradation-
`resistant GLP-1 analogues. We examined the biological
`activity of CJC-1131, a DPP-IV–resistant drug affinity
`complex (DAC) GLP-1 compound that conjugates to
`albumin in vivo. The CJC-1131 albumin conjugate bound
`to the GLP-1 receptor (GLP-1R) and activated cAMP
`formation in heterologous fibroblasts expressing a GLP-
`1R. CJC-1131 lowered glucose in wild-type mice, but not
`in GLP-1Rⴚ/ⴚ mice. Basal glucose and glycemic excur-
`sion following glucose challenge remained significantly
`reduced 10 –12 h following a single injection of CJC-
`1131. Twice daily administration of CJC-1131 to db/db
`mice significantly reduced glycemic excursion following
`oral and IP glucose challenge (P < 0.01 to 0.05) but did
`not significantly lower body weight during the 4-week
`study period. Levels of random fed glucose were signif-
`icantly lower in CJC-1131–treated ⴙ/ⴙ and db/db mice
`and remained significantly lower even 1 week following
`discontinuation of CJC-1131 administration. CJC-1131
`increased levels of pancreatic proinsulin mRNA tran-
`scripts, percent islet area, and the number of bromode-
`oxyuridine-positive islet cells. These findings demon-
`strate that an albumin-conjugated DAC:GLP-1 mimics
`the action of native GLP-1 and represents a new ap-
`proach for prolonged activation of GLP-1R signaling.
`Diabetes 52:751–759, 2003
`
`Glucagon-like peptide 1 (GLP-1) is a progluca-
`
`gon-derived peptide secreted from intestinal
`L-cells in response to nutrient ingestion (1,2).
`GLP-1 acts as an incretin to lower postprandial
`glycemic excursion via stimulation of insulin secretion and
`inhibition of glucagon secretion. GLP-1 also exerts actions
`
`From the 1Banting and Best Diabetes Centre, Department of Medicine,
`University of Toronto, Toronto General Hospital, Toronto, Ontario; and
`2Conjuchem, Montreal, Quebec.
`Address correspondence and reprint requests to Dr. Daniel J Drucker, 200
`Elizabeth St., MBRW-4R-402, Toronto, Ontario, Canada M5G 2C4. E-mail:
`d.drucker@utoronto.ca.
`Received for publication 16 July 2002 and accepted in revised form 6
`November 2002.
`AUC, area under the curve; CHO, Chinese hamster ovary; DAC, drug affinity
`complex; DPP-IV, dipeptidyl peptidase-IV; GIP, glucose-dependent insulino-
`tropic peptide; GLP-1, glucagon-like peptide 1; GLP-1R, GLP-1 receptor; HSA,
`human serum albumin.
`
`independent of islet hormone secretion, including inhibi-
`tion of both gastric emptying and food intake (3,4), and
`stimulation of ␤-cell proliferation (5,6).
`Although the structurally related gut hormone glucose-
`dependent insulinotropic peptide (GIP) also potentiates
`glucose-dependent insulin secretion (7), unlike GLP-1, the
`insulinotropic actions of GIP are diminished in diabetic
`rodents or in human subjects with type 2 diabetes (8,9). In
`contrast, GLP-1 administration rapidly lowers glucose in
`both normal and diabetic subjects (9 –11), and 6 weeks of
`continuous subcutaneous infusion of native GLP-1 signif-
`icantly decreased blood glucose and HbA1c in human
`patients with type 2 diabetes (12). Hence there is consid-
`erable enthusiasm for the development of GLP-1– based
`pharmaceutical agents for the treatment of type 2 diabetes
`(2,13).
`Although native GLP-1 effectively lowers blood glucose
`following acute peptide administration (14,15), both en-
`dogenous and exogenously administered GLP-1 exhibit a
`short t1/2 in vivo due primarily to NH2-terminal cleavage
`and inactivation by the enzyme dipeptidyl peptidase (DPP-
`IV) (16,17). The physiological importance of DPP-IV for
`GLP-1 degradation and glucose homeostasis is exemplified
`by studies of mice or rats with inactivating mutations in
`the DPP-IV gene. These rodents exhibit enhanced glucose
`clearance following glucose challenge and increased cir-
`culating levels of intact GLP-1 in vivo (18,19). Similarly,
`administration of DPP-IV enzyme inhibitors is associated
`with reduced glycemic excursion, enhanced insulin secre-
`tion, and reduced degradation of GLP-1 in normal and
`diabetic rodents (20 –22) and in human subjects (23).
`Accordingly, there is considerable interest in comple-
`mentary strategies for circumventing the rapid cleavage of
`GLP-1, including the development of GLP-1– based analogs
`with enhanced resistance to degradation and increased
`biological potency in vivo (13,24). The majority of these
`analogs exhibit one or more amino acid substitutions that
`reduce the affinity of the peptide for DPP-IV and subse-
`quent cleavage both in vitro and in vivo. Similarly, the
`naturally occurring lizard peptide exendin-4 is a potent
`GLP-1R agonist that exhibits reduced DPP-IV mediated
`cleavage and a longer duration of action in both rodents
`and human subjects (25,26). We have recently initiated
`studies of GLP-1 derivatives that are resistant to DPP-IV
`and are synthesized with a short covalent reactive chem-
`
`DIABETES, VOL. 52, MARCH 2003
`
`751
`
`MPI EXHIBIT 1033 PAGE 2
`
`

`

`CJC-1131, A GLP-1 ALBUMIN GLP-1R AGONIST
`
`ical linker that interacts with a specific cysteine residue in
`the albumin molecule following parenteral administration
`of the modified GLP-1 peptide. The resultant GLP-1–
`albumin drug affinity complex (DAC) is predicted to retain
`the actions of GLP-1, yet exhibit a more prolonged dura-
`tion of action due to a combination of DPP-IV resistance
`and the longer t1/2 conferred by serum albumin in vivo. To
`test the hypothesis that albumin-bound DAC–GLP-1 deriv-
`atives retain the biological properties of native GLP-1, we
`studied the activity of a DAC–GLP-1 compound, CJC-1131,
`using cells and in normal GLP-1R⫺/⫺ and diabetic ro-
`dents.
`
`RESEARCH DESIGN AND METHOD
`Animals. All animal experiments were carried out in accordance with
`protocols approved by the Toronto General Hospital Animal Care Committee
`or the Comite´ Institutionnel de Protection des Animaux de l’UQAM. C57BL/6
`db/db mice (The Jackson Laboratory, Bar Harbor, ME) and age- and sex-
`matched C57BL/6 wild-type mice from the same genetic background were
`used for chronic administration studies following a minimum 1-week accli-
`matization period in the animal facility. Wild-type CD-1 mice and GLP-1R⫺/⫺
`mice (27) in the CD-1 background were used for acute peptide administration
`experiments. Mice were allowed ad libitum access to food and water, except
`where noted. Animals were on a 12-h light, 12-h dark cycle (lights on 0700 h).
`For dose-response experiments shown in Fig. 2B, female CD-1 mice (Charles
`River Canada, St-Constant, QC), 7- to 10-weeks old, were studied, whereas for
`glycemic measurements shown in Figs. 2C and D, experiments were carried
`out with female db/db mice aged 7–10 weeks. CJC-1131 is a synthetic
`modification of GLP-1 (Fig. 1A) consisting of a single amino acid substitution
`of L-Ala8 to D-Ala8 at position 2, enabling some additional protection from
`DPP-IV and the addition of a lysine (Lys37) to the COOH-terminal of the
`peptide. The other modification involves the selective attachment of a
`[2-[2-[2-maleimidopropionamido(ethoxy)ethoxy]acetamide to the epsilon
`amino group of Lys37.
`In vitro experiments: binding affinity and cyclic AMP generation.
`Binding affinity was determined by incubating either native GLP-1 or CJC-1131
`and 0.03 nmol/l 125I-labeled GLP-1(7–36) amide with Chinese hamster ovary
`(CHO) cells stably transfected with the human GLP-1R cDNA using an
`incubation buffer of 20 mmol/l Tris-HCl, pH 7.4, 5 mmol/l MgCl2, 20 mmol/l
`NaCl, 2% BSA for 90 min at 37°C. For analysis of cAMP generation, CHO–
`GLP-1R cells were incubated for 20 min with various peptide ligands in the
`presence of 100 mmol/l IBMX followed by analysis of cAMP as described (28).
`Experimental protocol for db/db mouse studies. Baseline plasma glucose,
`oral and intraperitoneal glucose tolerance testing, and food intake were
`assessed before beginning CJC-1131 treatment. During the 4-week treatment
`period, wild-type and db/db mice were given an intraperitoneal injection of
`either 25 ␮g of CJC-1131 or an equal volume of saline twice daily at 800 and
`1600 h. Following the 4-week treatment period, a subset of mice from each
`group was sacrificed for analyses, whereas an additional set of mice continued
`to be monitored for another 3 weeks.
`Glucose tolerance tests and measurement of plasma insulin. Oral and
`intraperitoneal glucose tolerance tests were carried out following an over-
`night fast (16 –18 h) as described (27,29). Glucose (1.5 mg/g body wt) was
`administered orally through a gavage tube or via injection into the peritoneal
`cavity, and blood was drawn from a tail vein at 0, 10, 20, 30, 60, 90, and 120 min
`after glucose administration. Blood glucose levels were measured by the
`glucose oxidase method using a glucose meter (Glucometer Elite; Bayer,
`Toronto, Canada). For plasma insulin determinations, a blood sample was
`removed from the tail vein during the 10- to 20-min period following glucose
`administration, and plasma insulin was determined using a rat insulin enzyme-
`linked immunosorbent assay kit (Crystal Chem, Chicago, IL) with mouse
`insulin as a standard.
`RNA isolation and Northern blot analysis. Mice were anesthetized with
`CO2, and pancreata were removed immediately for RNA extraction by the
`acid-guanidinium isothiocyanate method. Total RNA (10 ␮g) was electropho-
`resed in a 1% (wt/vol) formaldehyde-agarose gel and transferred to a nylon
`membrane (Nytran Plus; Schleicher & Schuell, Keene, NH). For Northern blot
`analysis, the blot was hybridized to 32P-labeled random-primed complemen-
`tary DNA probes corresponding to rat proglucagon, rat insulin, or 18S RNA.
`Analysis of islet size and ␤-cell proliferation. Mice were injected with 100
`mg/kg 5-bromo-2⬘-deoxyuridine (BrdU, Sigma-Aldrich, St. Louis, MO) intra-
`peritoneally 6 h before being sacrificed. The pancreas was removed, fixed
`overnight in either 10% buffered formalin or 4% paraformaldehyde, and
`
`FIG. 1. A: Structural representation of CJC-1131 consisting of a single
`amino acid substitution of L-Ala8 to D-Ala8 at position 2 and a Lys37
`addition to the COOH-terminus with selective attachment of a [2-
`[2-[2-maleimidopropionamido(ethoxy)ethoxy]acetamide to the epsilon
`amino group of Lys37. B: Binding affinity of native GLP-1 (f) or
`CJC-1131:HSA (E) in CHO cells transfected with the human recombi-
`nant GLP-1R. C: cAMP response of native GLP-1 (f) or CJC-1131:HSA
`(E) in CHO cells transfected with the human recombinant GLP-1R.
`
`embedded in paraffin. Sections were obtained and stained with hematoxylin
`and eosin using standard protocols. Histological sections were immuno-
`stained for insulin using guinea-pig anti-insulin (Dako Diagnostics Canada,
`Mississauga, ON, Canada) as primary antibody (1:100 dilution) and rabbit
`anti– guinea-pig immunoglobulin (Dako Diagnostics Canada) as secondary
`antibody (1:50 dilution). Antibody binding was visualized by 3,3-diaminoben-
`zidine, and sections were counterstained by Meyers hematoxylin. Islet histo-
`morphometry was carried out according to the principles of Delesse (30). The
`sections were examined using a Leica (Leitz Labor Lux S; Leica Microsystem,
`Heerbrugg, Switzerland) microscope equipped with a video camera and
`connected to a computer with imaging software (Q500MC; Leica Microsys-
`tem). Estimates of islet area, islet number, number of BrdU immunopositive
`cells, and total pancreatic area were determined in a blinded manner as
`described (29,31).
`Statistics. Results are expressed as mean ⫾ SD or mean ⫾ SE. Statistical
`significance was calculated by Student’s t test using SPSS windows version 10
`(SPSS, Chicago, IL). P ⬍ 0.05 was considered to be statistically significant.
`
`RESULTS
`To circumvent the short biological t1/2 of native GLP-1 due
`principally to DPP-IV–mediated degradation, we designed
`a new DPP-IV–resistant GLP-1 derivative that would form
`
`752
`
`DIABETES, VOL. 52, MARCH 2003
`
`MPI EXHIBIT 1033 PAGE 3
`
`

`

`KIM AND ASSOCIATES
`
`FIG. 2. A: Blood glucose levels in 9-week-old wild-type C57ⴙ/ⴙ male
`mice during an oral glucose tolerance test performed 30 min after
`either intraperitoneal or subcutaneous administration of saline or
`CJC-1131 (25 ␮g); n ⴝ 5 mice/group. Values are expressed as means ⴞ
`SE. B: Blood glucose levels in 7- to 10-week-old female CD-1 mice
`during an oral glucose tolerance test 5 min after an intravenous
`administration of saline (f) or 1 (䡺), 10 (F), or 100 (E) nmol/kg of
`CJC-1131; n ⴝ 10 mice/group. Values are expressed as mean ⴞ SE; *P <
`0.05, **P < 0.02, ***P < 0.001 for differences in area under the curve
`(AUC) glucose for mice treated with saline vs. 1, 10, and 100 nmol/kg
`CJC-1131, respectively. C: Basal glycemia in 6- to 7-week-old female
`diabetic db/db mice after a single subcutaneous injection of either
`vehicle (f) or 100 nmol/kg CJC-1131 (E); n ⴝ 5 mice/group. Values are
`expressed as means ⴞ SE; *P < 0.02 for AUC glucose in saline versus
`CJC-1131–treated mice. D: Blood glucose levels in 7- to 10-week-old
`female db/db mice during an oral glucose tolerance test performed 12 h
`after intravenous administration of saline (f), 100 nmol/kg of native
`GLP-1 (䡺), or 100 nmol/kg of CJC-1131 (E); n ⴝ 5 mice/group. Values
`are expressed as mean ⴞ SE; *P < 0.05 for AUC glucose in saline versus
`CJC-1131–treated mice. E: Blood glucose levels in wild-type CD-1ⴙ/ⴙ
`(E) and GLP-1R ⴚ/ⴚ (F) mice during an oral glucose tolerance test
`performed 60 min after intraperitoneal administration of CJC-1131
`(25 ␮g); n ⴝ 9 –10 mice/group. Values are expressed as mean ⴞ SE; *P <
`0.05; **P < 0.01. #P < 0.001 for AUC glucose in GLP-1Rⴙ/ⴙ vs.
`GLP-1Rⴚ/ⴚ mice.
`
`a covalent bond with albumin in vivo, thereby conferring
`to covalently linked GLP-1 the longer circulating t1/2 of
`albumin (32). A D-alanine residue at position 2 was substi-
`tuted for the native alanine, and the COOH-terminal end of
`the GLP-1 molecule was coupled to a reactive chemical
`linker capable of forming a 1:1 covalent bond to the Cys
`residue in serum albumin (Fig. 1A). The resultant GLP-1
`derivative, designated CJC-1131, should retain the biolog-
`ical properties of native GLP-1 yet exhibit DPP-IV resis-
`tance and a prolonged t1/2 consistent with the clearance of
`native serum albumin in vivo (33).
`To assess the properties of a CJC-1131– human serum
`albumin (HSA) conjugate, we incubated HSA with CJC-
`1131 in vitro and utilized the purified CJC-1131:HSA con-
`
`jugate for studies of GLP-1R binding using CHO cells
`transfected with human recombinant GLP-1R (Fig. 1B).
`The displacement of 125I-labeled GLP-1 by native GLP-1
`versus the CJC1131:HSA complex was highly similar over
`a range of CJC-1131 concentrations (Ki ⫽ 5.16 nmol/l for
`native GLP-1 vs. 12 nmol/l for CJC-1131:HSA). To assess
`the bioactivity of CJC-1131, we measured cAMP accumu-
`lation using CHO cells transfected with human recombi-
`nant GLP-1R. These experiments demonstrated virtually
`identical dose-response relations for cAMP accumulation,
`with native GLP-1 exhibiting an EC50 of 13 nmol/l, whereas
`the EC50 for CJC-1131–HSA was 11–13 nmol/l (Fig. 1C).
`We next assessed whether the route of CJC-1131 admin-
`istration was an important determinant of bioactivity in
`
`DIABETES, VOL. 52, MARCH 2003
`
`753
`
`MPI EXHIBIT 1033 PAGE 4
`
`

`

`CJC-1131, A GLP-1 ALBUMIN GLP-1R AGONIST
`
`C57BL/6 mice following glucose loading. CJC-1131 mark-
`edly reduced glycemic excursion following intraperitoneal
`or subcutaneous administration (Fig. 2A). Similarly, intra-
`venous CJC-1131 administration produced a dose-depen-
`dent reduction in glycemic excursion following glucose
`loading (Fig. 2B). To ascertain whether a single injection
`of CJC-1131 would lower blood glucose in diabetic mice,
`we administered CJC-1131 by subcutaneous injection to
`db/db mice. Remarkably, basal random glycemia de-
`creased rapidly and remained lower in db/db mice for up to
`10 h following a single subcutaneous injection of CJC-1131
`(Fig. 2C). Furthermore, the glucose-lowering effect of
`CJC-1131 was still evident during an oral glucose tolerance
`test performed 12 h after a single intravenous CJC-1131
`injection (Fig. 2D). Although CJC-1131 consistently low-
`ered glucose in wild-type mice, no effect was observed in
`GLP-1R⫺/⫺ mice (Fig. 2E), demonstrating the critical
`importance of an intact GLP-1R for the biological activity
`of CJC-1131 in vivo.
`These findings demonstrated that single injections of
`CJC-1131 exert glucose-lowering effects in normal and
`diabetic mice. We next assessed whether more prolonged
`repeated administration of CJC-1131 would lower glucose
`in mice with severe diabetes. Wild-type control C57BL/6
`(C57⫹/⫹) ordb/db mice were treated with saline or
`CJC-1131 twice daily for 4 weeks. Before initiation of
`CJC-1131, mean fasting glucose was 4.3 ⫾ 0.9 mmol/l in
`C57⫹/⫹ vs. 17.9 ⫾ 6.7 mmol/l in db/db mice (Fig. 3A). After
`2 weeks of twice daily saline or CJC-1131 administration,
`fasting blood glucose was significantly lower in both
`control C57⫹/⫹ and db/db mice treated with CJC-1131
`(4.5 ⫾ 0.7 vs. 3.3 ⫾ 0.5 mmol/l and 19.2 ⫾ 5.8 vs. 12.5 ⫾ 3.7
`mmol/l, saline vs. CJC-1131 in C57⫹/⫹ vs. db/db mice,
`respectively; P ⬍ 0.01, Fig. 3A). Fasting glucose remained
`significantly lower in C57⫹/⫹ mice but not in db/db mice
`treated with CJC-1131 at the end of the 4-week treatment
`period (Fig. 3A). In contrast, fed blood glucose was
`significantly lower in CJC-1131–treated C57⫹/⫹ and db/db
`mice throughout the 4-week experiment (Fig. 3B; P ⬍ 0.01,
`saline versus CJC-1131). Furthermore, fed blood glucose
`remained significantly lower in db/db mice 1 week follow-
`ing the last injection of CJC-1131 (Fig. 3B, P ⬍ 0.05).
`Although CJC-1131 produced a small but significant de-
`crease in body weight in C57⫹/⫹ mice, no differences in
`body weight were detected in db/db mice treated with
`saline versus CJC-1131 over the 4-week experiment (Fig.
`3C).
`To determine whether repeated administration of CJC-
`1131 was associated with improvement in glucose toler-
`ance, we performed oral and intraperitoneal glucose
`tolerance testing in C57⫹/⫹ and db/db mice treated with
`saline or CJC-1131 for 2 weeks. Blood glucose excursion
`was significantly lower
`in CJC-1131–treated control
`C57⫹/⫹ mice following either oral or intraperitoneal
`glucose loading (Fig. 4A and B). Similarly, both fasting
`glucose and the glycemic excursion following glucose
`loading were modestly but significantly reduced in CJC-
`1131–treated db/db mice at multiple time points (Fig. 4A
`and B). Although plasma insulin levels were significantly
`greater in db/db mice compared with C57⫹/⫹ mice both in
`the fasting state and after glucose loading, plasma insulin
`
`FIG. 3. A: Weekly fasting blood glucose in C57ⴙ/ⴙ (E, F) and db/db (‚,
`Œ) mice before and during i.p treatment with saline (E, ‚) or 25␮g
`CJC-1131 (F, Œ) twice daily for 4 weeks and for several weeks
`following discontinuation of active treatment. The arrow denotes the
`end of the 4-week treatment period. B: Weekly fed blood glucose in
`C57ⴙ/ⴙ (E, F) and db/db (‚, Œ) mice treated with saline (E, ‚) or 25
`␮g CJC-1131 (F, Œ) twice daily observation for 4 weeks and observation
`for 3 weeks. C: Weekly body weight in C57ⴙ/ⴙ (E, F) and db/db (‚, Œ)
`mice treated with saline or 25 ␮g CJC-1131 twice daily by intraperitoneal
`injection for 4 weeks. Values are expressed as means ⴞ SE; *P < 0.05,
`**P < 0.01; n ⴝ 10–20 mice per group for experiments depicted in A–C.
`
`levels were not further increased in db/db mice treated
`with CJC-1131 (data not shown).
`GLP-1R agonists have been shown to increase proinsu-
`lin gene expression and promote islet neogenesis and
`␤-cell proliferation (5,6,34). To determine whether a larger
`CJC-1131:albumin conjugate exhibits comparable actions
`on the diabetic pancreas in vivo, we carried out Northern
`blot analysis using pancreatic RNA from C57⫹/⫹ and
`db/db mice. Levels of pancreatic insulin mRNA transcripts
`were comparable in normoglycemic wild-type C57⫹/⫹
`
`754
`
`DIABETES, VOL. 52, MARCH 2003
`
`MPI EXHIBIT 1033 PAGE 5
`
`

`

`KIM AND ASSOCIATES
`
`compared with control C57⫹/⫹ mice, but was not signif-
`icantly different in saline– vs. CJC-1131–treated C57⫹/⫹ or
`db/db mice (Fig. 5B).
`To determine whether CJC-1131 administration was
`associated with changes in the number or size of pancre-
`atic islets, we carried out morphometric analysis of pan-
`creata from C57⫹/⫹ and db/db mice following 4 weeks of
`treatment with either saline or CJC-1131 (Fig. 6). Total
`percent islet area was increased in C57⫹/⫹ mice and
`significantly increased in db/db mice following CJC-1131
`administration (P ⬍ 0.01; Fig. 6B). We next assessed the
`proportion of small, medium, and large islets, as previ-
`ously described (35). CJC-1131 treatment significantly
`increased the percent large islet area in db/db mice (Fig.
`6C; P ⬍ 0.01). The percentage of large islets was also
`increased in control mice treated with CJC-1131, but this
`difference did not achieve statistical significance (Fig. 6C).
`In contrast, the number of small islets was significantly
`increased in both C57⫹/⫹ (212.2%, P ⬍ 0.05) and db/db
`mice (94.6%, P ⬍ 0.05) treated with CJC-1131 (Fig. 6D).
`Furthermore, CJC-1131 significantly increased the total
`number of islets in both C57⫹/⫹ and db/db mice (Fig. 6E).
`To determine whether the observed differences in per-
`cent islet area and number of islets were attributable in
`part to increased rates of islet cell replication, we assessed
`the numbers of BrdU⫹ islet cells in saline– or CJC-1131–
`treated mice (Fig. 6F and G). CJC-1131 administration
`significantly increased the number of BrdU⫹ islet cells in
`db/db mice (413.1%, P ⬍ 0.01; Fig. 6G).
`
`FIG. 4. Blood glucose levels following oral (A) or intraperitoneal (B)
`glucose challenge in C57ⴙ/ⴙ (E, F) and db/db mice (‚, Œ), n ⴝ 20 mice
`per group, after 2 weeks of twice daily administration of either saline
`(E, ‚) or 25␮g CJC-1131 (F, Œ). AUC for oral glucose tolerance
`testing in C57ⴙ/ⴙ mice (saline treated 825.4 ⴞ 36.3 and CJC-1131–
`treated 550.2 ⴞ 29.0; P < 0.001) and db/db mice (saline treated
`3,620.6 ⴞ 55.0 and CJC-1131–treated 3,081.4 ⴞ 161.0; P < 0.01). AUC
`for intraperitoneal glucose tolerance testing in C57ⴙ/ⴙ mice (saline-
`treated 985.1 ⴞ 65.6 and CJC-1131–treated 434.2 ⴞ 28.0; P < 0.001)
`and db/db mice (saline-treated 3,298.1 ⴞ 204.8 and CJC-1131–treated
`2,824.6 ⴞ 218.6; P < 0.05). Values are expressed as mean ⴞ SE; *P <
`0.05, **P < 0.01.
`
`mice treated with either saline or CJC-1131. In contrast, 4
`weeks of CJC-1131 administration significantly increased
`the levels of proinsulin mRNA transcripts in db/db mice
`(Fig. 5A). Pancreatic insulin content was reduced in db/db
`
`DISCUSSION
`The observation that GLP-1 administration effectively re-
`duces blood glucose in human subjects with type 2 diabe-
`tes has
`stimulated considerable effort
`toward the
`development of GLP-1– based agonists for pharmaceutical
`administration. Although infusion of native GLP-1 is highly
`effective in lowering blood glucose in diabetic patients
`(12,36), the native peptide is an excellent substrate for
`DPP-IV (16,17), and exhibits a very short t1/2 in vivo.
`Hence, the requirement for constant GLP-1 infusion places
`potential constraints on the widespread use of native
`GLP-1 in the diabetes clinic. Accordingly, there remains
`ongoing interest in the development and characterization
`of peptidase-resistant GLP-1 analogs with longer durations
`
`FIG. 5. A: Pancreatic insulin mRNA; the lower panel depicts relative ratios of insulin/18s RNA from multiple analyses. **P < 0.01. B: Insulin
`content in C57ⴙ/ⴙ (n ⴝ 6) and db/db (n ⴝ 6) mice after 4 weeks of saline or CJC-1131 (25 ␮g twice daily) administration.
`
`DIABETES, VOL. 52, MARCH 2003
`
`755
`
`MPI EXHIBIT 1033 PAGE 6
`
`

`

`CJC-1131, A GLP-1 ALBUMIN GLP-1R AGONIST
`
`FIG. 6. Histological and morphometric analysis of pancreatic islets
`from C57ⴙ/ⴙ and db/db mice after 4 weeks of saline (䡺) or twice daily
`CJC-1131 (25 ␮g) treatment (f). A: Immunohistochemical staining for
`insulin. B: Percent total islet area/total pancreatic area. C: Percent
`large islet area/total pancreatic area. D: Number of small islets/
`pancreatic area (1 ⴛ 106 ␮m2). E: Total islet number/pancreatic area.
`F: Histological representation of BrdU immunopositive islet cells. G:
`quantitation of BrdU positive cells per pancreatic area (1 ⴛ 107 ␮m2)
`in C57ⴙ/ⴙ (n ⴝ 6) and db/db (n ⴝ 6) mice. Values are expressed as the
`means ⴞ SE; *P < 0.05, **P < 0.01.
`
`756
`
`DIABETES, VOL. 52, MARCH 2003
`
`MPI EXHIBIT 1033 PAGE 7
`
`

`

`of action suitable for subcutaneous administration in vivo
`(13).
`Several approaches have been undertaken to develop
`stable long-acting GLP-1 analogs, with DPP-IV–resistant
`molecules such as NN2211 currently being tested in clini-
`cal trials for the treatment of type 2 diabetes (37). Ex-
`endin-4, a naturally occurring lizard salivary gland peptide
`(38) that exhibits potent GLP-1–like activity and exerts its
`actions through the GLP-1R, is also undergoing clinical
`evaluation for the treatment of type 2 diabetes (26,39). The
`data presented here describe the results of introducing
`two specific modifications of the native GLP-1 molecule,
`one to engineer DPP-IV resistance, the second to achieve
`chemical coupling of the GLP-1 analog (CJC-1131) to
`serum albumin. Albumin is known to exhibit a longer t1/2
`in vivo, ⬃19 days in humans (40), which is much greater
`than the t1/2 of short-lived regulatory peptides such as
`GLP-1. Accordingly, peptide binding to albumin has been
`used to improve the pharmacokinetic properties of several
`smaller proteins, including Fab antibody fragments (33),
`coagulation factor VIIa inhibitor 1a (41), and insulin (42).
`The present results indicate that a much larger albumin–
`GLP-1 covalently linked conjugate retains the biological
`properties of native GLP-1, including binding to and acti-
`vation of the GLP-1R in vitro. Furthermore, the glucose-
`lowering effects of CJC-1131 were abolished in GLP-
`1R⫺/⫺ mice,
`providing
`important
`complementary
`evidence that the actions of CJC-1131 in vivo are mediated
`by the known GLP-1R.
`Several experimental results provide indirect evidence
`for a sustained duration of CJC-1131 action on glucose
`lowering in vivo. First, oral glucose tolerance remained
`significantly improved in mice even 12 h after a single
`CJC-1131 injection. Furthermore, fed glucose remained
`significantly lower in db/db mice treated with CJC-1131,
`even 1 week after the last CJC-1131 injection. These
`findings are consistent with data demonstrating sustained
`improvements in insulin secretion in Zucker diabetic fatty
`rats detectable even 1 week following a transient GLP-1
`infusion (43). Similarly, blood glucose remained lower in
`exendin-4 –treated partially pancreatectomized rats sev-
`eral weeks following discontinuation of exendin-4 admin-
`istration (5). Although fed glucose and glucose tolerance
`were clearly improved in db/db mice treated with CJC-
`1131, significant hyperglycemia persisted even during re-
`peated CJC-1131
`administration.
`In contrast, daily
`administration of exendin-4 to younger, less severely dia-
`betic db/db mice (starting fasting hyperglycemia of 5.1
`mmol/l) prevented the progression to more severe hyper-
`glycemia over the 2-week treatment period (44). However,
`a 2-week treatment of older, more severely diabetic db/db
`mice with twice daily administration of 200 ␮g/kg NN221
`or exendin-4 (100 ␮g/kg) did not prevent ongoing deterio-
`ration in glucose tolerance, with mice remaining very
`hyperglycemic even after the 2-week treatment period
`(45). Hence the effects of chronic GLP-1 agonist adminis-
`tration in the db/db mouse appear dependent, in part, on
`the clinical severity of the diabetes and age of the mice at
`the onset of treatment.
`Although treatment of rodents and human subjects with
`GLP-1R agonists has been associated with prevention of
`weight gain or modest weight loss (12,25,46), we did not
`
`DIABETES, VOL. 52, MARCH 2003
`
`KIM AND ASSOCIATES
`
`observe a significant reduction in body weight in db/db
`mice treated with CJC-1131 over the 4-week treatment
`period. Similarly, body weight was not significantly re-
`duced in db/db mice treated with NN2211 or exendin-4
`over comparable 2- to 4-week treatment periods (45,47). At
`present, it remains unclear whether restricted transport of
`an albumin–GLP-1 conjugate across the blood-brain bar-
`rier might compromise the ability of molecules such as
`albumin–CJC-1131 to reduce food intake and weight gain
`in vivo. Nevertheless, significant weight loss was observed
`in control C57⫹/⫹ mice treated with CJC-1131 over the
`same time period. Furthermore, intravenous infusion of
`CJC-1131 rapidly induced c-fos immunoreactivity in sev-
`eral regions of
`the rat central nervous system (H.
`Yamamoto, J. Elmquist, and D.J.D., unpublished observa-
`tions), providing complementary evidence suggesting that
`an albumin–GLP-1 conjugate is capable of activating cen-
`tral nervous system functions in vivo.
`Despite the failure to observe normalization of glycemia
`in the older db/db mouse, CJC-1131 administration in-
`creased islet area, with a significant increase in both the
`number and the relative percentage of small and large
`islets detected following 4 weeks of CJC-1131 treatment.
`Furthermore, the number of BrdU⫹ islet cells was signif-
`icantly increased in CJC-1131–treated mice, consistent
`with previous studies demonstrating that activation of the
`GLP-1R is coupled to expansion of islet mass in the db/db
`mouse (6,44,45). Hence, although fenestration of islet
`capillaries does restrict access and reduce permeability
`toward larger molecules in the islet microcirculation (48),
`the comparatively larger CJC-1131–albumin conjugate ap-
`parently retains the ability to promote islet growth and
`enhance insulin gene expression in the mouse in vivo.
`Despite the increase in islet area and proinsulin mRNA
`transcripts in CJC-1131–treated mice, we did not observe a
`significant increase in insulin content in the db/db mice
`treated with CJC-1131. Indeed, pancreatic insulin content
`was modestly decreased in control and CJC-1131–treated
`db/db mice relative to C57⫹/⫹ mice consistent with the
`functional exhaustion of ␤-cell insulin stores in db/db mice
`with blood glucose levels approaching 20 mmol/l.
`Given the increasing interest in strategies based on
`enhancement of incretin action for the treatment of dia-
`betes, there remains an unmet need for developing ap-
`proaches that circumvent the requirement for daily or
`continuous administration of GLP-1 analogs. Accordingly,
`long-acting or slow-release forms of GLP-1 analogs that
`exhibit desirable therapeutic properties are awaited with
`interest. The administration of albumin-based GLP-1 deriv-
`atives should confer a longer circulating t1/2 to the newly
`derived albumin-peptide conjugate, compared with the
`native GLP-1 peptide alone (32,33). The data presented
`here demonstrate that a prototype DPP-IV–resistant GLP-1
`analog that covalently couples to albumin, CJC-1131, re-
`tains the biological properties generally ascribed to
`GLP-1R agonists. The findings that CJC-1131 binds to and
`activates the GLP-1R,
`lowers blood glucose,
`increases
`proinsulin gene expression, and stimulates islet cell pro-
`liferation suggest that further assessment of the efficacy
`and mechanisms of action of albumin-based GLP-1 deriv-
`atives appears warranted.
`
`757
`
`MPI EXHIBIT 1033 PAGE 8
`
`

`

`CJC-1131, A GLP-1 ALBUMIN GLP-1R AGONIST
`
`ACKNOWLEDGMENTS
`The w

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