throbber
Downloaded from https://academic.oup.com/jpp/article/56/12/1477/6147469 by Maine Medical Center user on 21 June 2023
`
`lrn"i1
`~ Journalof
`
`Pharmacy and Pharmacology
`
`JPP 2004, 56: 1477–1492
`ß 2004 The Authors
`Received 15 July, 2004
`Accepted 13 August, 2004
`DOI 10.1211/0022357044805
`ISSN 0022-3573
`
`Department of Physiology and
`Pharmacology, University of
`Strathclyde, Strathclyde Institute
`for Biomedical Sciences, Taylor
`Street, Glasgow G4 ONR, UK
`
`Brian Furman, Nigel Pyne
`
`School of Biomedical Sciences,
`University of Ulster, Coleraine,
`BT52 1SA, Northern Ireland
`
`Peter Flatt, Finbarr O’Harte
`
`Correspondence: B. Furman,
`Department of Physiology and
`Pharmacology, University of
`Strathclyde, Strathclyde Institute
`for Biomedical Sciences, Taylor
`Street, Glasgow G4ONR, UK.
`E-mail: b.l.furman@strath.ac.uk
`
`Funding and acknowledgements:
`The study was part funded
`by Diabetes UK, the Research
`and Development Office
`of the Department of Health
`and Personal Social Services
`for Northern Ireland, and
`University of Ulster Research
`Strategy Funding. We are
`grateful to Mrs Pat Owen
`for her assistance.
`
`Review Article
`
`Targeting b-cell cyclic 3050adenosine monophosphate
`for the development of novel drugs for treating
`type 2 diabetes mellitus. A review
`
`Brian Furman, Nigel Pyne, Peter Flatt and Finbarr O’Harte
`
`Abstract
`
`Cyclic 3’5’AMP is an important physiological amplifier of glucose-induced insulin secretion by the
`pancreatic islet
-cell, where it is formed by the activity of adenylyl cyclase, especially in response to
`the incretin hormones GLP-1 (glucagon-like peptide-1) and GIP (glucose-dependent insulinotropic
`peptide). These hormones are secreted from the small intestine during and following a meal, and are
`important in producing a full insulin secretory response to nutrient stimuli. Cyclic AMP influences
`many steps involved in glucose-induced insulin secretion and may be important in regulating
`pancreatic islet
-cell differentiation, growth and survival. Cyclic AMP (cAMP) itself is rapidly
`degraded in the pancreatic islet
-cell by cyclic nucleotide phosphodiesterase (PDE) enzymes. This
`review discusses the possibility of targeting cAMP mechanisms in the treatment of type 2 diabetes
`mellitus, in which insulin release in response to glucose is impaired. This could be achieved by the use
`of GLP-1 or GIP to elevate cAMP in the pancreatic islet
-cell. However, these peptides are normally
`rapidly degraded by dipeptidyl peptidase IV (DPP IV). Thus longer-acting analogues of GLP-1 and
`GIP, resistant to enzymic degradation, and orally active inhibitors of DPP IV have also been devel-
`oped, and these agents were found to improve metabolic control in experimentally diabetic animals
`and in patients with type 2 diabetes. The use of selective inhibitors of type 3 phosphodiesterase
`(PDE3B), which is probably the important pancreatic islet
-cell PDE isoform, would require their
`targeting to the islet
-cell, because inhibition of PDE3B in adipocytes and hepatocytes would induce
`insulin resistance.
`
`Introduction
`
`The prevalence of diabetes mellitus, especially of type 2 diabetes, is increasing and the
`World Health Organisation predicts a world total of > 300 M by 2030. The high
`prevalence of diabetes combined with the associated increased mortality and morbid-
`ity, primarily as a result of macrovascular disease and microvascular long-term com-
`plications, make it a major health problem. Overwhelming evidence, especially from the
`DCCT study (The Diabetes Control and Complications Trial Research Group 1993)
`for type 1 diabetes and the UK Prospective Diabetes Study (Turner 1998) for type 2
`diabetes suggested that good metabolic control would markedly reduce mortality and
`morbidity.
`It is now broadly accepted that type 2 diabetes results from both peripheral insulin
`insensitivity and impaired insulin secretion (Kahn 2003). There is progressive islet
-cell
`failure in patients with type 2 diabetes and a reduction in the
-cell mass (Porte & Kahn
`2001). Thus the islet
-cell remains an important target for the development of drugs for
`treating type 2 diabetes. Drugs are required that both enhance insulin secretion in response
`to normal physiological meal-related nutrient stimuli and that prevent the progressive loss
`of islet
-cell mass. Augmentation of meal-related insulin secretion by drugs should be
`glucose-dependent. This would ensure that increased insulin secretion occurs when
`required, the corollary being that insulin secretion would remain at a basal rate between
`meals, avoiding hyperinsulinaemia and thus hypoglycaemia, as seen with sulphonylureas,
`and the potential cardiovascular risks associated with chronic hyperinsulinaemia and
`insulin resistance (Juhan-Vague & Alessi 1997; Bastard et al 2000). Physiological,
`meal-related insulin secretion occurs in response to absorbed nutrients, primarily glucose.
`Glucose stimulates insulin secretion following its transport into and metabolism in the
`
`1477
`
`Novo Nordisk Exhibit 2004
`Mylan Pharms. Inc.v. Novo Nordisk A/S
`IPR2023-00722
`Page 00001
`
`

`

`Downloaded from https://academic.oup.com/jpp/article/56/12/1477/6147469 by Maine Medical Center user on 21 June 2023
`
`i ~ ~
`CY---.
`ll~ J
`
`, ,._
`
`l '
`
`I
`
`'\
`
`I v
`
`\~ _ -
`
`8p
`
`1478
`
`Brian Furman et al
`
`glucose
`
`K+
`
`K+
`
`glucose
`
`ATP
`
`nitric oxide
`
`+
`
`depolarization
`
`Ca2+
`stores
`
`+
`
`Ca2+
`
`GLP-1
`
`Ca2+
`
`GLP-1
`receptor
`
`adenylyl cyclase
`
`GIP
`
`GIP
`receptor
`
`guanylyl cyclase
`
`metabolism
`
`GDP
`
`GTP
`
`Transcription
`
`Rap1
`
`Rap1
`
`GTP
`
`Cyclic 3’5’GMP
`
`PK A
`
`PDE1
`+
`
`PDE5
`
`−
`
`Ca2+
`
`5’GMP
`
`Epac
`
`+
`
`+
`
`+
`
`Cyclic 3’5’AMP
`
`+
`
`PDE1
`
`PDE3B
`
`+
`
`+
`
`+
`
`5’AMP
`
`+
`exocytosis
`
`IGF-1
`receptor
`
`leptin
`receptor
`
`Insulin
`receptor
`
`insulin
`
`Ca2+
`
`IGF-1
`
`liver
`
`adipose
`tissue
`
`leptin
`
`
`
`Figure 1 Schematic diagram of the pancreatic islet
-cell showing the sites of action (indicated by the star symbol) of cyclic 3050AMP in
`modulating glucose-induced insulin secretion and other
-cell activities. The pathways for the formation and destruction of cAMP and the
`hormonal factors influencing these pathways are shown also.
`
`
-cell, generating ATP which closes KATP channels, resulting
`in depolarization and calcium influx (Figure 1). The effect of
`glucose on insulin secretion can be amplified by signalling
`pathways involving inositol trisphosphate and diacylglycerol
`derived from activation of phospholipase C (Howell et al
`1994; Gilon & Henquin 2001) and by cyclic 3050AMP follow-
`ing activation of adenylyl cyclase (Howell et al 1994).
`Glucose itself has long been known to elevate the pancreatic
`islet
-cell cAMP content (Grill & Cerasi 1973). Although
`glucose-induced insulin secretion does not appear to require
`cAMP or the protein kinase A (PKA)-system (Grill & Cerasi
`1973; Sharp 1979; Persaud et al 1990; Lester et al 1997),
`increases in cAMP augment the direct effect of glucose.
`This cyclic nucleotide is generally accepted as an important
`amplifier of glucose-induced insulin release (Holz & Habener
`1992), particularly when its levels are increased by glucose
`itself (Harnda¨ hl et al 2002) and, importantly, by various gut
`hormones implicated as incretins, which are secreted from
`the small intestine in response to the presence of glucose and
`other nutrients in the gut. It has also been suggested that
`cAMP is a competence factor for normal islet
-cell respon-
`siveness to glucose (Schuit 1996; Huypens et al 2000).
`Numerous hormones that stimulate insulin secretion increase
`islet
-cell cAMP, including glucagon (Huypens et al 2000),
`ACTH (Al-Majed et al 2004), and pituitary adenylate
`cyclase-activating polypeptide (PACAP; Filipsson et al
`(2001)). On the other hand, hormones inhibiting insulin
`secretion (galanin (Drews et al 1994), adrenaline (Peterhoff
`et al 2003)), reduce cAMP, although their effects on cAMP
`
`may not necessarily explain the inhibition of secretion. The
`major incretins are glucose-dependent insulinotropic poly-
`peptide (GIP) secreted by intestinal K-cells and glucagon-like
`peptide 1 secreted by intestinal L-cells. These hormones aug-
`ment glucose-induced insulin secretion through activating
`adenylyl cyclase, leading to an increase in islet
-cell cAMP.
`Their importance is illustrated clearly by the marked glucose
`intolerance and impairment of insulin secretion seen in mice
`lacking receptors for both hormones (Preitner et al 2004).
`
`Secretion and actions of glucagon-like peptide-1
`(GLP-1)
`
`GLP-1 is derived from the processing of the preprogluca-
`gon gene in intestinal L-cells (Lund et al 1982). Various
`products of the post-translational processing of progluca-
`gon exist including GLP-1(7–36)amide and GLP-1(7–37),
`which are equipotent stimulators of glucose-dependent
`insulin secretion (Holst et al 1987; Kreymann et al 1987;
`Mojsov et al 1987). For the purpose of this review, GLP-1
`refers to the active form GLP-1(7–36)amide.
`GLP-1 is secreted from intestinal L-cells in response to
`the absorption of glucose, other sugars, fatty acids and to a
`minor extent amino acids (MacIntosh et al 2001; Feinle et al
`2002; Brubaker & Anini 2003). The GLP-1 secretory
`actions of metabolizable nutrients may be linked to cellular
`ATP generation, KATP channel blockade and elevation
`of intracellular Ca2þ
`(Gribble & Reimann 2002). There is
`also a neurally-mediated arm to GLP-1 secretion and a
`
`Novo Nordisk Exhibit 2004
`Mylan Pharms. Inc.v. Novo Nordisk A/S
`IPR2023-00722
`Page 00002
`
`

`

`Downloaded from https://academic.oup.com/jpp/article/56/12/1477/6147469 by Maine Medical Center user on 21 June 2023
`
`stimulatory effect of GIP. These pathways may be espe-
`cially important for release of GLP-1 from the distal region
`of the small intestine (Holst et al 1987; Filipsson et al 2000).
`The insulinotropic actions of GLP-1 are evident in islets
`(Komatsu et al 1989; Weir et al 1989) in normal and diabetic
`animals (Hendrick et al 1993; Scrocchi et al 1998; O’Harte
`et al 2000a, 2000b, 2001) and in healthy and diabetic volun-
`teers (Gutniak et al 1992).
`Other physiological actions of GLP-1 target the pancrea-
`tic islets, liver, adipose tissue and skeletal muscle. In pan-
`creatic islets, GLP-1 stimulates insulin and somatostatin
`secretion (Holst et al 1987; Mojsov et al 1987; Eissele et al
`1990; Creutzfeldt et al 1996) and inhibits glucagon secretion
`(Creutzfeldt et al 1996). GLP-1 stimulates insulin gene tran-
`scription (Drucker et al 1987; Fehmann & Habener 1992;
`Skoglund et al 2000), pancreatic islet cell proliferation
`(Buteau et al 1999; Perfetti et al 2000), and
-cell replication
`(Edvell & Lindstrom 1999). GLP-1 leads to the differentia-
`tion of pancreatic ductal AR42J cells into glucagon- and
`insulin-producing cells (Zhou et al 1999), through a PDX-1
`dependent pathway (De La Tour et al 2001; Hui et al 2001).
`Lesser known actions of GLP-1 include promotion of glu-
`cose uptake and glycogen formation in liver and skeletal
`muscle (Valverde et al 1994; Villanueva-Penacarrillo et al
`1994; O’Harte et al 1997). GLP-1 also stimulates lactate
`production, glucose uptake and glycogen storage in skeletal
`muscle (O’Harte et al 1997).
`
`Secretion and actions of GIP
`
`The 42 amino acid peptide GIP was isolated from a crude
`porcine CCK extract and shown to inhibit histamine-
`induced gastric acid secretion (Brown et al 1969). How-
`ever, its major physiological role as a glucose-dependent
`stimulator of insulin secretion is now well recognized
`(Gault et al 2003a).
`GIP is secreted from intestinal K-cells by glucose and
`other actively transported sugars and by fatty acids.
`Amino acids however are only a weak stimulus.
`Although GIP stimulus-secretion coupling pathways are
`poorly understood, there appears to be a close link with
`K-cell metabolism (Tseng et al 1994). Indeed it appears
`likely that signal transduction of intestinal K- and L-cells
`will be found to have many parallels to other glucose-
`sensitive secretory cells, most notably the pancreatic
-
`cell (Purrello & Rabuazzo 2000). Notably, insulin forms
`an effective feedback loop by inhibiting nutrient stimu-
`lated GIP secretion (Bryer-Ash et al 1994).
`GIP is a potent glucose-dependent stimulator of insulin
`secretion in all in-vitro and in-vivo systems tested (Pederson
`et al 1998a, 1998b; Drucker 2003; Vilsboll & Holst 2004).
`Additional actions on the
-cell include stimulation of
`proinsulin gene transcription and translation (Fehmann &
`Go¨ ke 1995; Wang et al 1996), enhancement of the growth,
`differentiation and survival of pancreatic
-cells (Trumper
`et al 2001; Pospisilik et al 2003). Extra pancreatic actions of
`GIP appear to enhance its glucose-lowering ability by inhi-
`biting hepatic glucose production (Elahi et al 1986) and
`promoting glucose uptake in isolated mouse diaphragm
`muscle (O’Harte et al 1997). Functional GIP receptors
`
`Targeting cAMP in the islet
-cell
`
`1479
`
`have also been identified on adipocytes (Yip et al 1998),
`where GIP has been shown to stimulate glucose transport
`(Eckel et al 1979), fatty acid synthesis (Oben et al 1991) and
`lipoprotein lipase activity (Knapper et al 1995).
`
`Cyclic AMP in the pancreatic islet
-cell
`GLP-1 and GIP exert their insulinotropic actions through
`activation of G-protein coupled receptors
`(GPCRs)
`(Thorens 1992; Usdin et al 1993). This results in activation
`of adenylyl cyclase and an increase in intracellular cAMP.
`Their effects in stimulating insulin synthesis and in the
`enhancement of the growth, differentiation and survival
`of pancreatic
-cells are probably also mediated by
`cAMP. However, there is evidence that other pathways,
`such as the transactivation of epidermal growth factor
`receptors, may be involved in stimulation of
-cell proli-
`feration (Buteau et al 2003). Cyclic AMP has many effects
`within the
-cell, as described below (see also Figure 1).
`
`Insulin secretion Several mechanisms may mediate the
`effect of cAMP in augmenting glucose-induced insulin
`secretion. These include increased opening of voltage-sensi-
`tive Ca2þ
`channels (Kanno et al 1998), calcium-induced
`Ca2þ
`release (Kang et al 2001), activation of ryanodine
`receptors in the endoplasmic reticulum (Islam et al 1998;
`Holz et al 1999), stimulation of
-cell lipolysis (Yaney et al
`2001) and direct effects on exocytosis (Harnda¨ hl et al 2002).
`Most actions of cAMP in the
-cell appear to be mediated
`through PKA-catalysed phosphorylation events. There is
`evidence for specific targeting of PKA to particular sub-
`cellular locations within the
-cell by A-kinase anchoring
`proteins (AKAPs) and disruption of AKAPs inhibits, for
`example, the effects of cAMP in increasing intracellular
`Ca2þ
`and stimulating insulin secretion (Lester et al 1997).
`However, some effects of the cyclic nucleotide on exocytosis
`are partly PKA-independent (Renstrom et al 1997). PKA-
`independent effects on exocytosis may be mediated by the
`cAMP-binding proteins known either as cAMP-regulated
`guanine nucleotide exchange factors (GEFs) or exchange
`proteins activated by cAMP (Epacs) which target the small
`G-protein Rap1 (Kopperud et al 2003). The pancreatic islet
`
-cell expresses both Epac 1 and Epac 2 (Holz 2004).
`Treatment of pancreatic islets with antisense oligodeoxy-
`nucleotides against Epac reduced the effect of a permeant
`cAMP analogue in augmenting glucose-induced insulin
`secretion (Kashima et al 2001). A recently described, novel
`cAMP analogue, 8-(4-chloro-phenylthio)-20-O-methylade-
`nosine-30-50-cyclic monophosphate
`(8-pCPT-20-O-Me-
`cAMP) activated Epac but not PKA. This analogue was
`shown to mobilize Ca2þ
`from intracellular Ca2þ
`stores via
`Epac-mediated Ca2þ
`-induced Ca2þ
`release in human pan-
`creatic
-cells and INS-1 insulin-secreting cells (Kang et al
`2003). The cAMP-mediated insulinotropic action of the
`incretin factors glucose-dependent insulinotropic peptide
`(GIP) and glucagon-like peptide 1 (GLP-1) (Drucker et al
`1987) are mediated in part through PKA, but PKA-inde-
`pendent actions have also been demonstrated and probably
`involve Epac, as evidenced by the effects of antisense oligo-
`deoxynucleotides against Epac (Kashima et al 2001).
`
`Novo Nordisk Exhibit 2004
`Mylan Pharms. Inc.v. Novo Nordisk A/S
`IPR2023-00722
`Page 00003
`
`

`

`Downloaded from https://academic.oup.com/jpp/article/56/12/1477/6147469 by Maine Medical Center user on 21 June 2023
`
`1480
`
`Brian Furman et al
`
`Other roles in the
-cell Glucose and the insulinotropic
`hormones have actions on the
-cell which extend beyond
`the acute stimulation of insulin secretion. These include
`increased insulin synthesis and the modulation of
-cell
`growth, differentiation and apoptosis. In addition to its
`role in amplifying glucose-induced insulin secretion,
`cAMP may mediate or modulate these other effects.
`Glucose-mediated increases in insulin synthesis involve the
`phosphorylation of the transcription factor pancreatic duo-
`denal homeobox-1 (PDX-1) and its translocation to the
`nucleus (Elrick & Docherty 2001). While the effects of
`glucose itself on PDX-1 are not mediated by cAMP, there
`is strong evidence for the importance of cAMP in GLP-1-
`dependent stimulation of PDX-1 in the
-cell, as well as its
`translocation to the nucleus and its activation of the insulin
`gene promoter (Wang et al 2001). However, the role of
`cAMP in regulating insulin synthesis is unclear since the
`adenylyl cyclase activator forskolin or the cAMP analogue
`8-bromo-cAMP suppressed insulin transcription in INS-1
`cells in a PKA-independent manner (Ding et al 2003).
`Cyclic AMP may mediate effects of glucose in stimulating
`the expression of immediate early response genes such as
`c-myc (Jonas et al 2001) and c-fos (Susini et al 1998). In this
`context, cAMP can either activate or inhibit the p42/p44
`mitogen-activated protein kinase pathway depending on the
`cell type and conditions (Calleja et al 1997). Glucose itself
`activates the p42/p44 MAPK pathway (Benes et al 1998,
`1999) and this effect is amplified, although not mediated by
`cAMP. Indeed, the activation of this pathway by GIP is
`cAMP and PKA-dependent (Ehses et al 2002). Regulation
`of gene expression by cAMP may have relevance to effects
`on
-cell growth, differentiation and apoptosis. For exam-
`ple, Andersen et al (1996) showed that elevated cAMP levels
`protected rat islets against interleukin-1
-mediated induc-
`tion of nitric oxide synthase and nitric oxide production.
`Excessive nitric oxide production appears to mediate sub-
`sequent apoptosis of
-cells. On the other hand,
-cell lines
`were made more susceptible to apoptosis following expo-
`sure to dibutyryl cAMP (Loweth et al 1996) or the cAMP
`elevating agent forskolin (Ahmad et al 2000b).
`
`Formation of cAMP in the
-cell
`The pancreatic islet
-cell was shown to express several of
`the nine adenylyl cyclases (AC), including AC1, AC2,
`AC3, AC4, AC5, AC6 and AC8 (Leech et al 1999;
`Guenifi et al 2000; Delmeire et al 2003),
`including the
`calcium-calmodulin activated AC1, AC3 and AC8.
`
`Destruction of cyclic nucleotides in pancreatic
`islet b-cell
`
`The hydrolysis of cAMP and cyclic GMP (cGMP) to their
`biologically inactive 50 derivatives is catalysed by the family
`of enzymes known as the cyclic nucleotide phosphodi-
`esterases, which provide the only known system for destroy-
`ing these cyclic nucleotides. Currently there are 11 known
`gene families of CN-PDEs (PDE1–PDE11), comprising
`more than 50 enzymes with differences in their substrate
`selectivity (cAMP vs cGMP), kinetics, allosteric regulation,
`
`tissue distribution and susceptibility to pharmacological
`inhibition (reviewed by Perry & Higgs 1998; Soderling &
`Beavo 2000; Mehats et al 2002). Table 1 shows the sub-
`strate, the Km values, particular properties and inhibitors
`of some of these enzymes, particularly those which have
`been identified in pancreatic islets. In view of the clear
`importance of cyclic nucleotides, especially cAMP, in the
`pancreatic islet
-cell, it is important to understand the
`nature and the roles of the phosphodiesterase (PDE)
`enzymes expressed in pancreatic islet
-cells.
`
`PDE isoforms present in the islets
`
`Most studies to elucidate the PDE isoforms present in the
`islet
-cell have been undertaken using pancreatic islets,
`which contain four endocrine cell types; these comprise
`the insulin secreting
-cells, the glucagon secreting A cells,
`the somatostatin secreting D cells and the pancreatic poly-
`peptide (PP) cells. Non-endocrine cells, including blood
`vessels are also present. To look more selectively at the islet
`
-cell, insulin-secreting cell lines have been used extensively,
`although these have limitations as models for native
-cells.
`Evidence was obtained for the presence of PDE1 (calcium-
`calmodulin activated), PDE3 (cyclic 3050 GMP-inhibited)
`and PDE4 (cAMP-specific) in islets and
-cell lines.
`There is good evidence for the presence of PDE3 in
`insulin secreting cells for the role of this isoform in regulat-
`ing the cAMP pool relevant to insulin secretion. Thus
`membrane and cytosolic fractions of islet homogenates
`contained a low Km (1.4–2.2 M) cAMP PDE activity
`(Shafiee-Nick 1995) that was inhibited by 60–70% using
`10 M cGMP (Furman & Pyne 1990). The pellet fraction of
`homogenates of BRIN BD11 cells was also found to con-
`tain a cAMP PDE activity that was potently (IC50 0.7 M)
`inhibited (up to 30–40%) by cGMP (Ahmad et al 2000b).
`Of the two isoforms of type 3 phosphodiesterase (PDE3),
`PDE3A and PDE3B, only PDE3B appears to be expressed
`in the
-cell. Western blotting of extracts of rat pancreatic
`islets and the
-cell line HIT-T15, using a polyclonal anti-
`body against a GST-PDE3B, revealed a single protein band
`corresponding in size to the PDE3B protein found in
`extracts of rat epididymal adipose tissue (Zhao et al 1997).
`Immunostaining of rat islets showed that PDE3B was
`expressed only in cells that were co-stained with anti-insulin
`antibodies (Zhao et al 1997). Reverse transcription-poly-
`merase chain reaction (RT-PCR) using BRIN-BD11 cell
`RNA and PDE3B-specific primers demonstrated a product
`showing > 97% sequence homology with rat adipose tissue
`PDE3B (Ahmad et al 2000a, b).
`The selective PDE3 inhibitors SK&F 94836 and Org
`9935 potently inhibited rat islet PDE activity, especially in
`membrane fractions (Shafiee-Nick et al 1995) with up to
`85% of membrane-bound PDE being inhibited. Similar
`observations were made in human islets (Shafiee-Nick
`et al 1994) and in the BRIN BD11 insulin secreting cell
`line (Ahmad et al 2000a), although in homogenates of
`this cell line, unlike in islets, the drugs inhibited cAMP
`hydrolysis only in the membrane fractions. Rat and
`human islets were shown by Parker et al (1995) to contain
`a milrinone-sensitive PDE, accounting for up to 70% of
`
`Novo Nordisk Exhibit 2004
`Mylan Pharms. Inc.v. Novo Nordisk A/S
`IPR2023-00722
`Page 00004
`
`

`

`Downloaded from https://academic.oup.com/jpp/article/56/12/1477/6147469 by Maine Medical Center user on 21 June 2023
`
`Targeting cAMP in the islet
-cell
`
`1481
`
`Table 1 Properties of phosphodiesterases that have been identified in the pancreatic islet
-cell
`
`Enzyme Reference
`
`Substrate Km (mM) Properties/comment
`
`Inhibitors
`
`PDE1A Han et al (1999)
`PDE1B Clapham & Widerspin (2001)
`PDE1C Yan et al (1995)
`
`PDE3A Perry & Higgs (1998),
`Mehats et al (2002)
`PDE3B Pyne et al (1987),
`Harrison et al (1986)
`
`cGMP
`cAMP
`
`3
`1–30
`
`cAMP
`
`0.5
`
`Activated by Ca/calmodulin.
`Evidence for PDE1C in
`insulin secreting cells
`
`Zaprinast (1/5), vinpocetine (1A)
`SCH51866 (also 5,9,10A),
`8-MM-IBMX (1C)
`
`Inhibited by cGMP
`Phosphorylation by PKA and
`insulin-sensitive kinase.
`Evidence for PDE3B but not
`PDE3A in insulin secreting cells
`
`Milrinone, Org 9935, siguazodan,
`cilostamide, enoximone, OPC3911.
`No evidence that any
`of these is selective for PDE3B
`vs PDE3A
`
`Torphy (1998), Houslay (1998) cAMP
`
`0.2–4
`
`PDE4A
`PDE4B
`PDE4C
`PDE4D
`
`Phosphorylation by PKA and
`p42/p44 MAPK (PDE4D3).
`There is no indication which
`subtype(s) present in insulin
`secreting cells.
`
`PDE5
`
`Corbin & Francis (1999)
`
`cGMP
`
`2
`
`Phosphorylation by PKA/PKG
`
`PDE6
`
`Pittler et al (1990)
`
`cGMP
`
`60–100 Has been identified recently in
`pancreatic
-cells (Kaminski &
`Morgan 2004)
`
`Rolipram, RP73401, zardaverine,
`CP80633, CDP840, LAS31025,
`SB207499
`
`Zaprinast, sildenafil, dipyridamole,
`SCH51866 (also 1,9, 10A)
`
`Zaprinast, dipyridamole
`
`PDE8A,
`PDE8B
`
`Fisher et al (1998)
`
`cAMP
`
`0.7
`
`Dipyridamole
`
`IBMX-insensitive. The only
`evidence for its presence in insulin
`secreting cells is the observation
`that approximately 10% of the
`cAMP PDE activity is insensitive
`to IBMX.
`
`PDE10A Soderling & Beavo (2000)
`
`cAMP
`cGMP
`
`0.05
`3
`
`SCH51866 (also 1,5,9) dipyridamole
`
`total islet PDE activity. The PDE3 activity found in the
`soluble fraction of homogenates of rat islets might be
`PDE3A from blood vessels or other non-
-cell tissue,
`since PDE3A was shown exclusively in the cytosolic frac-
`tions of all tissues studied, whereas PDE3B was expressed
`in particulate fractions (Liu & Maurice 1998).
`Other isoforms also contribute to islet
-cell PDE
`activity. Both islets (Sugden & Ashcroft 1981; Lipson &
`Oldham 1983; Capito et al 1986) and the
-cell lines BRIN
`BD11 (Ahmad et al 2000a) and
-TC3 (Han et al
`1999) contain a Ca-calmodulin activated PDE. The
`PDE1/PDE5 inhibitor zaprinast produced a 14–30% inhi-
`bition in membrane and cytosolic fractions of rat islet
`homogenates (Shafiee-Nick et al 1995) and 25% inhibition
`of cAMP/cGMP PDE activity in BRIN BD11 cells
`(IC501–5 M) (Ahmad et al 2000a). Han et al (1999)
`showed that the presence of a PDE that was inhibited by
`zaprinast (IC50 4.5 M) and 8-MM-IBMX (IC50 7.5 M)
`but not by vinpocetine (IC50 > 100 M for PDE1C vs 1.4
`and 9.8 M for PDE1A and PDE1B, respectively). These
`findings were compatible with the presence of PDE1C but
`not PDE1A or PDE1B and this was supported by RT-
`PCR (Han et al 1999).
`A role for the cAMP-selective PDE4 in the hydrolysis
`of
-cell cAMP was supported by the inhibition of islet
`(Shafiee-Nick et al 1995; Parker et al 1995) or BRIN-
`BD11 cell (Ahmad et al 2000a) PDE by the selective
`PDE4 inhibitor rolipram. Han et al (1999) demonstrated
`
`the expression of PDE4A and PDE4D in
TC3 cells using
`RT-PCR.
`
`The importance of PD3B in regulating insulin
`secretion
`
`studies, using biochemical and pharmacological
`Our
`approaches, first suggested the role of PDE3 in modulating
`glucose-induced insulin secretion in islets of rat (Furman &
`Pyne 1990; Shafiee-Nick et al 1995) and man (Shafiee-Nick
`et al 1994). This has been generally confirmed by others in
`islets (Parker et al 1995; Zhao et al 1997; Harnda¨ hl et al
`2002), and by ourselves and others in insulin secreting cell
`lines (Ahmad et al 2000a; Harnda¨ hl et al 2002). The impor-
`tance of PDE3B in regulating
-cell cAMP in the context of
`insulin secretion was demonstrated by adenovirus-mediated
`over-expression of PDE3B in a
-cell
`line or in islets
`(Harnda¨ hl et al 2002) and by using transgenic animals
`over-expressing PDE3B in the
-cell (Harnda¨ hl et al 2004).
`Those studies clearly showed that glucose-induced, as well
`as GLP-1-induced insulin secretion was impaired by
`PDE3B over-expression in-vitro and in-vivo.
`Although there is very strong evidence supporting the
`role of PDE3B in the
-cell, some studies have suggested
`the importance of other PDEs. Rolipram, a selective PDE4
`inhibitor, augmented glucose-induced insulin secretion in
`
-cell lines (Han et al 1999; Ahmad et al 2000a), although
`not in islets (Shafiee-Nick et al 1995). This discrepancy
`
`Novo Nordisk Exhibit 2004
`Mylan Pharms. Inc.v. Novo Nordisk A/S
`IPR2023-00722
`Page 00005
`
`

`

`Downloaded from https://academic.oup.com/jpp/article/56/12/1477/6147469 by Maine Medical Center user on 21 June 2023
`
`1482
`
`Brian Furman et al
`
`between islets and cell lines may be due to differences in the
`compartmentalization of PDE4 isoforms, allowing PDE4
`to interact with relevant cAMP pools in transformed cell
`lines but not in native
-cells. A role for PDE1 in both the
`
-TC3 cell line and in islets was suggested by Han et al
`(1999) who showed that 8-MM-IBMX, purported to be a
`selective inhibitor of the Ca-calmodulin activated PDE1C
`isoform, augmented glucose-induced insulin secretion. The
`importance of PDE1 in regulating
-cell cAMP remains to
`be determined.
`
`Exploitation of cAMP mechanisms in the
`development of drugs to treat type 2 diabetes
`
`From the foregoing it is clear that increasing
-cell levels
`of cAMP will augment glucose-induced insulin secretion.
`Therapeutically, this could be achieved by activating
-
`cell adenylyl cyclase using analogues of GLP-1 or GIP,
`increasing the availability of endogenous GLP-1 and GIP
`by preventing their normal very rapid breakdown or by
`preventing the breakdown of cAMP in the
-cell using
`inhibitors of phosphodiesterase selective for the most
`important
-cell isoform i.e. PDE3B.
`
`GLP-1 analogues
`
`The metabolic stability and biological activity of many
`GLP-1 analogues have been investigated (Deacon et al
`1998; Knudsen et al 2000; Xiao et al 2001; Green et al
`2003a, b, 2004a). So far, limited clinical trials have been
`conducted using GLP-1 (Gutniak et al 1992; Meier et al
`2003; Meneilly et al 2003), GLP-1 analogues (Rolin et al
`2002; Chang et al 2003; Holz & Chepurny 2003) and
`exendin-4 (Egan et al 2002, 2003). However,
`clear
`improvements in glucose tolerance have been observed in
`experimental animals (Holst 1999).
`
`N-terminal (His7 modified) analogues of GLP-1
`Substitution of His7 with several other amino acids indi-
`cated that the imidizole ring of His7 was crucial for GLP-1
`action (Hareter et al 1997). It was shown that N-terminal
`extension of GLP-1, with insertion of an acetyl group at
`the -amino group of His7, did not significantly affect
`GLP-1 action. N-terminally extended peptides with imidi-
`zole-lactic acid, N-methyl, and -methyl groups at His7
`(Table 2) are more resistant to dipeptidyl peptidase IV
`(DPP IV) than native GLP-1, but exhibit reduced affinity
`for the GLP-1 receptor and compromised ability to sti-
`mulate cAMP production (Hareter et al 1997). His7-gluci-
`tol-GLP-1 was shown to be resistant to DPP IV whilst
`maintaining antihyperglycaemic activity in-vivo (Table 2)
`(O’Harte et al 2000a, b). Most recently, two additional
`analogues, N-acetyl-GLP-1 and N-pyroglutamyl-GLP-1,
`have been shown to be completely resistant to DPP IV and
`human plasma degradation, as well as exhibiting potent
`receptor binding, cAMP production and insulin secretory
`activity in-vitro (Table 2). In obese diabetic (ob/ob) mice,
`N-acetyl-GLP-1 and N-pyroglutamyl-GLP-1 displayed
`potent insulinotropic actions, with N-pyroglutamyl-GLP-1
`
`being particularly effective compared with the antihy-
`perglycaemic effects of the native peptide (Green et al
`2004b).
`
`Position 2 (Ala8 substituted) analogues of GLP-1
`Substitution of Ala8 for Gly conferred increased resistance
`to DPP IV and, despite a reduced affinity for the GLP-1
`receptor, corrected the fasting hyperglycaemia and glucose
`intolerance of diabetic mice (Burcelin et al 1999) (Table 2).
`Similarly, substitution with Ser significantly increased the
`plasma stability of GLP-1 without impairing its insulino-
`tropic activity in rats (Ritzel et al 1998)
`(Table 2).
`Substitution of Ala8 for Thr, Gly, Ser and -aminoiso-
`butyric acid (Aib) significantly prolonged biological half-
`lives in-vivo compared with native GLP-1, but nevertheless
`bound to the GLP-1 receptor with high affinities (Deacon
`et al 1998) (Table 2). However, only native GLP-1 and
`(Aib8)GLP-1 significantly improved insulin output over
`basal conditions (Deacon et al 1998). Similarly, analogues
`with D-Alanine (D-Ala), Ser and Gly substitutions at posi-
`tion 8 were substantially more resistant to DPP IV than
`native GLP-1, and had similar or enhanced biological half-
`life and potency (Siegel et al 1999a, b) (Table 2). Substitution
`of Ala8 for Gly and aminohexanoic acid (Aha) (Table 2)
`produced analogues that stimulated insulin secretion and
`intracellular cAMP to a similar degree to native GLP-1
`(Doyle et al 2001). In-vivo they lowered circulating blood
`glucose and increased blood insulin concentrations in
`Zucker rats. More recently, Green et al (2003b) character-
`ized two novel Ala8-substituted analogues of GLP-1,
`namely (Abu8)GLP-1 and (Val8)GLP-1. These were
`shown to be completely resistant to the actions of DPP IV
`or human plasma (Table 2). Receptor binding studies
`demonstrated that (Abu8)GLP-1 and (Val8)GLP-1 bound
`the GLP-1 receptor with high affinity, but that this was
`reduced compared with native GLP-1 (Table 2). Although
`active stimulators of
`intracellular cAMP production,
`(Abu8)GLP-1 and (Val8)GLP-1 were 1.5- and 3.5-fold less
`potent, respectively, than native GLP-1 (Green et al 2003b).
`Despite these losses in receptor affinity and cAMP produc-
`tion, this did not compromise insulinotropic activity either
`in-vitro or in-vivo (Table 2). This may be partially explained
`by the diverse mechanisms of action of GLP-1 on the
`pancreatic
-ce

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