throbber
The Journal of Clinical Investigation
`
`Discovery, characterization, and clinical development
`of the glucagon-like peptides
`
`Daniel J. Drucker,1 Joel F. Habener,2 and Jens Juul Holst3
`1Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada. 2Laboratory of Molecular Endocrinology, Massachusetts General Hospital, Harvard University,
`Boston, Massachusetts, USA. 3Novo Nordisk Foundation Center for Basic Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
`
`The discovery, characterization, and clinical development of glucagon-like-
`peptide-1 (GLP-1) spans more than 30 years and includes contributions from
`multiple investigators, science recognized by the 2017 Harrington Award
`Prize for Innovation in Medicine. Herein, we provide perspectives on the
`historical events and key experimental findings establishing the biology
`of GLP-1 as an insulin-stimulating glucoregulatory hormone. Important
`attributes of GLP-1 action and enteroendocrine science are reviewed, with
`emphasis on mechanistic advances and clinical proof-of-concept studies.
`The discovery that GLP-2 promotes mucosal growth in the intestine is
`described, and key findings from both preclinical studies and the GLP-2
`clinical development program for short bowel syndrome (SBS) are reviewed.
`Finally, we summarize recent progress in GLP biology, highlighting emerging
`concepts and scientific insights with translational relevance.
`
`The endocrine activity of the gastroin-
`
`testinal tract has been studied for more
`than a century, with gut hormones such as
`secretin emerging from the seminal stud-
`ies of Bayliss and Starling (1). The concept
`that the gut also controlled pancreatic
`islet secretions was supported by experi-
`ments demonstrating that administration
`of crude gut extracts lowered blood glu-
`cose in animals. The development of the
`insulin radioimmunoassay enabled the
`description of the incretin effect, namely
`that glucose administered into the gut
`potentiated insulin secretion to a greater
`extent than isoglycemic stimulation of
`insulin secretion achieved through i.v.
`glucose administration. The first incretin
`hormone, glucose-dependent insulinotro-
`pic polypeptide (GIP), was isolated by John
`Brown in the 1970s (2). Here, we describe
`the discovery and characterization of the
`
`second incretin hormone, GLP-1. We high-
`light 3 decades of science from multiple
`laboratories supporting the development
`of GLP-1– and GLP-2–based therapies.
`GLP-1 is now used for the treatment of
`type 2 diabetes (T2D) and obesity, whereas
`GLP-2 is used for the therapy of short bow-
`el syndrome (SBS).
`
`Discovery of GLP-1
`Although GIP was isolated through classical
`peptide purification and protein sequenc-
`ing methodology, the discovery of the
`GLP-1 sequence stemmed from the appli-
`cation of recombinant DNA approaches
`developed in the laboratories of Stanley
`Cohen, Paul Berg, and Herb Boyer in the
`early 1970s. This remarkable new techn-
`ology allowed for a rapid and accurate
`prediction of the amino acid sequences of
`proteins by the decoding of the nucleotide
`
`Conflict of interest: D.J. Drucker is supported by the Canada Research Chairs program and a BBDC-Novo Nordisk
`Chair in incretin biology, and has received speaking or consulting honorarium from Arisaph, Intarcia, Merck, Pfizer,
`and Novo Nordisk Inc. Mt. Sinai Hospital receives research support from GSK, Merck, and Novo Nordisk, for preclin-
`ical studies in the Drucker laboratory. J.J. Holst has served as a consultant or advisor to Novartis Pharmaceuticals,
`Novo Nordisk, Merck Sharp & Dohme, and Roche and has received fees for lectures from Novo Nordisk, Merck Sharp
`& Dohme, and GlaxoSmithKline.
`Reference information: J Clin Invest. 2017;127(12):4217–4227. https://doi.org/10.1172/JCI97233
`
`sequences of cloned recombinant cDNA
`copies of messenger RNAs. The Habener
`lab utilized this technology to elucidate
`proglucagon amino acid sequences from
`cDNAs and genes isolated from angler-
`fish in the early 1980s (3–5) and the rat
`proglucagon cDNA and gene sequences
`followed shortly thereafter (refs. 6, 7, Fig-
`ure 1, and Figure 2). Corresponding pro-
`glucagon sequences from hamster, bovine,
`and human were identified by Graeme
`Bell and others in the early 1980s (8–10).
`These sequences revealed that glucagon
`and re lated GLP sequences were encoded
`by larger protein precursors, termed pre-
`prohormones (Figure 2). The anglerfish
`preproglucagons (Figure 2A), isolated and
`characterized by Lund and Goodman (3–5),
`were interesting as there were two different
`cDNAs encoded by separate (nonallelic)
`genes and they each contained a glucagon-
`related sequence, in addition to glucagon.
`The two anglerfish glucagon–related pep-
`tides resembled GIP, a glucoincretin hor-
`mone released from the gut into the circu-
`lation during meals, subsequently shown
`by Dupre and Brown in 1973 to augment
`glucose-dependent insulin secretion (11).
`Unlike the two anglerfish preproglucagons,
`each of which harbored glucagon and a sin-
`gle glucagon-related peptide, the mammali-
`an preproglucagons all contained glucagon
`and two additional glucagon-related pep-
`tides, designated GLP-1 and GLP-2 (Figure
`2B). Notably, the corresponding amino acid
`sequences of the GLP-1s in the four mam-
`malian species were identical (12), with
`conservation of sequence implying as yet
`unknown but potentially important biolog-
`ical actions of GLP-1.
`Collectively, these findings further
`supported the evolving notion at the
`time that small peptide hormones are
`synthesized in the form of larger prohor-
`mones and that the final bioactive pep-
`tides are formed posttranslationally by
`selective enzymatic cleavages from the
`prohormones (Figure 2B). Earlier studies
`
`jci.org Volume 127 Number 12 December 2017
`
`4 2 1 7
`
`H A R R I N G T O N P R I Z E E S S A Y
`
`MPI EXHIBIT 1018 PAGE 1
`
`

`

`The Journal of Clinical Investigation
`
`Figure 1. Historical timeline depicting seminal events in the discovery and development of gluca-
`gon-like peptide therapeutics.
`
`had demonstrated that insulin and para-
`thyroid hormone are synthesized as pre-
`prohormones. The hydrophobic amino
`terminal sequences, termed the pre- or
`signal sequences, are cleaved from the
`nascent polypeptide chains during their
`translation on ribosomes, leaving the
`prohormones as the precursor of the
`peptide hormones. The A and B chains
`of insulin and parathyroid hormone were
`found to be cleaved from their respective
`prohormones by the actions of specific
`endopeptidases, prohormone conver-
`tases, which cleave proteins at the sites
`of two consecutive basic amino acids,
`arginine and lysine (13). These earlier
`findings of prohormone convertase spec-
`ificity were applied to deduce potential
`cleavage sites in proglucagon.
`
`Structure-activity properties
`of GLP-1
`Examination of the amino acid sequence
`of proglucagon
`initially presented a
`conundrum regarding the processes by
`which potentially bioactive GLP-1 pep-
`tides might be liberated from the pro-
`hormone (Figure 2B). In keeping with
`the rule that bioactive peptides are
`cleaved from prohormones at sites con-
`sisting of two basic amino acids (13),
`several investigators initially predicted
`that bioactive GLP-1 would be a peptide
`of 37 amino acids beginning with histi-
`dine and ending in glycine, GLP-1(1-37).
`However, further inspection of the pro-
`hormone sequences revealed a second
`single basic amino acid followed by his-
`tidine residing 6 amino acids carboxyl-
`proximal to the first histidine, predict-
`ing a GLP-1 peptide of 31 amino acids,
`
`GLP-1(7-37). Further, at the carboxyl-
`terminal region of the putative GLP-1 pep-
`tide resides a sequence RGRR predicting
`a prohormone convertase–directed cleav-
`age site followed by an amidation of the
`penultimate arginine by a peptidylglycine
`α-amidating monooxygenase (14) result-
`ing in peptides of 36 and 30 amino acids,
`GLP-1(1-36)amide and GLP-1(7-36)amide.
`The availability of the amino acid
`sequences of GLP-1 and GLP-2 obtained
`from the decoding of the nucleotide
`sequences based on the genetic code
`allowed for the chemical synthesis of the
`predicted peptides, examination of their
`biological activities, and preparation of
`peptide-specific antisera. Daniel Drucker
`joined the Habener laboratory in the sum-
`mer of 1984, with the original intent of
`studying the molecular control of thyroid
`hormone biosynthesis. However, the thy-
`roid group, led by Bill Chin, was decamp-
`ing for the Brigham and Women’s Hospi-
`tal, and Daniel was assigned to work on the
`proglucagon gene. To understand whether
`proglucagon might be processed to yield
`multiple proglucagon-derived peptides
`(PGDPs), including GLP-1 and GLP-2,
`Drucker transfected a proglucagon cDNA
`expression vector into fibroblasts, pituitary
`cells, and islet cells (15). Although minimal
`processing was observed in fibroblasts,
`immunoreactive GLP-1 and GLP-2 was
`detected by chromatography and radio-
`immunoassays in medium and extracts
`from transfected pituitary and islet cells
`(15). Application of similar chromatogra-
`phy and radioimmunoassay techniques
`in studies of rat (16), pig (17), and human
`(18) tissues revealed distinct profiles of
`PGDPs in pancreas and gut (Figure 3). Of
`
`note, in addition to glucagon, the pancreas
`contained a large peptide with immuno-
`reactive determinants for both GLP-1 and
`GLP-2 (but not glucagon), consistent with
`incompletely cleaved proglucagon (major
`proglucagon fragment [MPGF]) (Figure 3,
`A and C, and ref. 19). In contrast to find-
`ings in pancreas, immunoreactive GLP-1
`peptides detected in gut extracts consisted
`entirely of smaller peptides (refs. 15 and
`16, and Figure 3, B and C). These findings
`indicated that the cleavage of proglucagon
`into small GLP-1–immunoreactive pep-
`tides was more efficient in the gut com-
`pared with pancreas. These observations
`were also consistent with the incretin con-
`cept in which, in response to oral nutrients,
`glucoincretin hormones such as GIP (and
`subsequently GLP-1) originate from the
`gut and not the pancreas.
`Svetlana Mojsov in the Habener lab-
`oratory detected glycine-extended and
`arginine-amidated isoforms of GLP-1 — as
`well as both the amino-terminally extended
`peptides GLP-1(1-37) and GLP-1(1-36)amide
`and the amino-truncated peptides GLP-1
`(7-37) and GLP-1(7-36)amide — in extracts
`of pancreas (Figure 3A). Nevertheless, the
`abundance of these peptides was much
`greater in the gut (20).
`
`Bioactivities of the GLP-1
`peptides
`The earliest studies of the bioactivities of
`GLP-1(1-37) were indecisive. One study
`found that the 37 amino acid peptide
`activated adenylyl cyclase in membranes
`prepared from rat pituitary and hypothal-
`amus (21), whereas another study failed to
`detect any effects of the peptide on glucose
`and insulin in cortisone-treated rabbits
`
`4 2 1 8
`
`jci.org Volume 127 Number 12 December 2017
`
`H A R R I N G T O N P R I Z E E S S A Y
`
`MPI EXHIBIT 1018 PAGE 2
`
`

`

`The Journal of Clinical Investigation
`
`bioactivities for the extended forms, GLP-1
`(1-37) and GLP-1(1-36)amide, have yet been
`determined. Furthermore, no distinctive bio-
`logical activities have been attributable spe-
`cifically to the amidated forms of GLP-1.
`
`The view from Denmark
`In Copenhagen, Jens Holst and colleagues
`were interested in the incretin effect and
`were studying the condition of postpran-
`dial reactive hypoglycemia after gastric
`surgery (27). This type of hypoglycemia
`was clearly hyperinsulinemic, yet the sig-
`nal for insulin secretion was unknown.
`Looking for possible candidates, they were
`inspired by Lise Heding’s work on gluca-
`gon and her identification of the immu-
`nological differences between gut and
`pancreatic glucagon (28). Knowing that
`glucagon would stimulate insulin secre-
`tion, they were interested in the numer-
`ous cells in the gut that produce immu-
`noreactive glucagon (29). Eventually, this
`work led to the identification of glicentin
`and oxyntomodulin (Figure 2B), which
`both contain the full glucagon sequence,
`explaining the immunoreactivity in the
`gut (30–32).
`Having identified all of the molecular
`components of glicentin also in the pan-
`creas (33), they proposed that glicentin
`represents at least part of a common gut
`and pancreatic glucagon precursor, which
`undergoes differential processing in the
`two tissues, a hypothesis subsequently con-
`firmed through identification of the human
`proglucagon gene by Graeme Bell and col-
`leagues (7). However, it was also clear that
`proglucagon was larger than glicentin, and
`the interest focused on peptides contained
`within the MPGF representing the remain-
`der of proglucagon (minus glicentin) (34).
`The early work decoding the anglerfish
`proglucagon cDNA by Lund and Habener
`(3) followed by elucidation of the ham-
`ster proglucagon cDNA by Graeme Bell
`(9) supported a hypothesis that cleavage
`of MPGF might result in liberation of the
`GLPs. The Holst group quickly developed
`radioimmunoassays for GLP-1 and GLP-2
`to test this hypothesis. To their excitement,
`they found that MPGF was indeed differ-
`entially processed in the gut, but not in the
`pancreas, to yield two GLPs (ref. 17 and
`Figure 3C). However, using the perfused
`pancreas preparation, they soon realized
`that neither of the two GLPs used in these
`
`Figure 2. Structure and processing of anglerfish and human proglucagon. Representation of the
`structures of proglucagon cDNAs from Anglerfish (A) and Human (B), with tissue-specific liberation of
`individual proglucagon-derived peptides in pancreas or intestine. PC1, prohormone convertase 1; PC2,
`prohormone convertase 2; MPGF, major proglucagon fragment. Arrows in A represent sites of cleave by
`prohormone convertase enzymes.
`
`(22). To determine whether the shorter
`GLP-1 isoforms were bioactive, Drucker
`incubated truncated forms of GLP-1 with
`multiple cell lines, examining cell growth,
`gene expression, and signal transduc-
`tion. Remarkably, insulin-producing islet
`cells responded to N-terminally truncated
`forms of GLP-1, exhibiting increased lev-
`els of cyclic AMP accumulation within
`minutes of exposure to 50 pM–5 nM GLP-
`1(7-37) (Figure 4A). In contrast, neither
`glucagon, GLP-1(1-37), or GLP-1(1-36)–
`NH2 increased cAMP accumulation at
`these concentrations (23). GLP-1(7-37)
`also increased the levels of insulin mRNA
`transcripts and stimulated insulin secre-
`tion at 25 mM but not 5.5 mM glucose in
`studies with isolated islet cell lines (Figure
`4, B and C, and ref. 23). In contrast, nei-
`ther glucagon nor GLP-2 induced insulin
`gene expression. Furthermore, GLP-1
`(7-37) had no effect on levels of angioten-
`sin gene expression in the insulinoma cell
`line and did not change levels of glucagon,
`prolactin, and corticotropin mRNAs in islet
`or pituitary cell lines (23). Hence, these
`findings (Figure 4, A–C) published in 1987
`(23) established that GLP-1(7-37) directly
`augments glucose-dependent insulin bio-
`synthesis and secretion from β cells.
`
`The demonstration that GLP-1 directly
`increased cAMP levels provided condi-
`tional evidence for the existence of a Gs
`protein-coupled receptor in β cells. In
`studies of insulin secretion using the iso-
`lated perfused pancreas, Mojsov and Weir
`demonstrated that GLP-1(7-37) and not
`GLP-1(1-37) stimulated insulin secretion
`at concentrations as low as 50 pM (ref. 24
`and Figure 5, A–C). Likewise, as outlined
`below, Jens Holst and colleagues showed
`that luminal glucose stimulated GLP-1
`secretion from the perfused intestine (Fig-
`ure 5D), and doses from 500 pM to 5 nM
`GLP-1(7-36)amide stimulated insulin secre-
`tion in the perfused pig pancreas (Figure
`5E and ref. 25). Thus, it turned out that a
`cleavage in proglucagon at the single basic
`amino acid, arginine, and not the double
`basic amino acids, generated the active
`GLP-1 peptides, GLP-1(7-37) and GLP-1
`(7-36)amide (Figure 5C). First in man studies
`reported in December 1987 by Kreymann
`and Bloom (26) rapidly established the
`insulinotropic actions of GLP-1(7-36)amide
`in human subjects. Although numerous
`studies have demonstrated that the amino-
`terminally truncated forms of GLP-1, GLP-
`1(7-37), and GLP-1(7-36)amide are active
`glucoregulatory hormones, no compelling
`
`jci.org Volume 127 Number 12 December 2017
`
`4 2 1 9
`
`H A R R I N G T O N P R I Z E E S S A Y
`
`MPI EXHIBIT 1018 PAGE 3
`
`

`

`The Journal of Clinical Investigation
`
`Mentlein in Kiel, Holst and Deacon showed
`that the GLP-1 molecule was cleaved by the
`enzyme
`dipeptidyl-peptidase-4(DPP-4)
`in vivo and that inhibitors of this enzyme
`could completely protect the molecule
`(43). In fact, the circulating half-life of
`GLP-1 was only 1.5–2 minutes in human
`subjects with diabetes, and they proposed
`that inhibitors of DPP-4 could maintain
`higher levels of intact active endogenous
`GLP-1 for therapeutic purposes (44). Sub-
`sequent studies soon demonstrated that
`DPP-4–resistant GLP-1 analogues were
`longer-acting than native GLP-1 (45). Fur-
`thermore, inhibitors of DPP-4 completely
`prevented the breakdown of GLP-1 in the
`circulation and amplified the insulinotro-
`pic actions of GLP-1 (46). This exciting
`development, presented in a Perspectives
`article in Diabetes in 1998 (47), was soon
`followed by the development of clinically
`useful inhibitors, first vildagliptin and sub-
`sequently sitagliptin.
`It remained to be understood whether
`GLP-1 receptor agonists would actually
`be useful for clinical diabetes therapy or
`whether tachyphylaxis would develop
`upon chronic administration. The group
`in Copenhagen administered synthetic
`GLP-1 by constant s.c. infusion for 6
`weeks to a group of individuals with
`long-standing T2D (48). Fortunately, no
`tachyphylaxis was observed; GLP-1 ther-
`apy reduced fasting and mean plasma
`glucose by 4.3 and 5.5 mmol/l; glycated
`hemoglobin by 1.3 %; and body weight by
`2 kg. Moreover, insulin sensitivity and β
`cell function, assessed by clamp studies,
`greatly improved. Importantly, no limit-
`ing side effects were re corded (48), pro-
`viding proof of concept in 2002 for GLP-1
`therapy in subjects with T2D. It was now
`clear that GLP-1–based therapies had tre-
`mendous potential.
`
`The Toronto perspective:
`the physiology of GLPs and
`discovery of GLP-2
`Building on the availability of cloned pro-
`glucagon gene sequences in the Habener
`lab (6), Daniel Drucker and Jacques
`Philippe pursued the analysis of the molec-
`ular control of islet α cell proglucagon
`gene expression in the mid 1980s (49, 50).
`Upon returning to Toronto in 1987, Druck-
`er extended these studies to examine pro-
`glucagon gene expression in the intestine
`
`Figure 3. Processing of proglucagon and glucagon-like peptides in the pancreas and intestine.
`Detection of immunoreactive forms of GLP-1 in extracts from rat pancreas (A) and intestine (B) as
`adapted from ref. 16. Characterization of proglucagon-derived peptide immunoreactivity secreted
`from perfused pig pancreas and intestine (C) using peptide-specific antisera reveal tissue-specific
`posttranslational processing of the PGDPs, as outlined in ref. 17.
`
`studies had any effect on pancreatic hor-
`mone secretion. They therefore decided
`to isolate the naturally occurring hormone
`from porcine and human and gut extracts,
`and they found that the naturally occur-
`ring peptide was a truncated from of GLP-1
`representing proglucagon
`(aa 78-108)
`(Figure 5C) and subsequently found to be
`amidated, corresponding to proglucagon
`78-107amide (35). Importantly, this hormone
`was potently insulinotropic (Figure 5E and
`ref. 25), so they had also described a new
`incretin hormone in 1987; however, they
`wondered whether GLP-1 was more inter-
`esting than the already known incretin
`GIP, which exhibited a diminished effect
`on insulin secretion in patients with T2D
`(36). They soon found, using the perfused
`pancreas, that GLP-1 — in contrast to GIP —
`also powerfully inhibits glucagon secretion
`(37). Eventually, they demonstrated that
`during infusions of physiological amounts
`of GLP-1 into humans, insulin secretion
`would be stimulated and glucagon secre-
`tion inhibited, resulting in a decrease in
`hepatic glucose production (38). However,
`the effect was self-limiting, with the insulin-
`
`stimulating activity attenuated as plasma
`glucose levels started to fall, limiting the
`fall to 0.5–1 mmol/l.
`At that time, the Copenhagen group
`realized that GLP-1 was extremely interest-
`ing and, in further studies, demonstrated
`that it strongly inhibited gastric motility
`and gastric and pancreatic exocrine secre-
`tion (39), consistent with an important
`role for this hormone as a regulator of
`upper gastrointestinal function. They also
`demonstrated that infusions of GLP-1 in
`humans inhibited appetite and food intake,
`actions subsequently exploited in the clinic
`to treat obesity (40). In studies published
`in 1993 by Michael Nauck and colleagues
`in Göttingen, i.v. infusion of GLP-1 com-
`pletely normalized severely elevated fast-
`ing glucose concentrations in patients with
`long-standing T2D as a consequence of the
`actions of GLP-1 to stimulate insulin and
`inhibit glucagon secretion (41). Although
`GLP-1 clearly had therapeutic potential,
`s.c. injections of GLP-1 were disappoint-
`ingly ineffective (42). The explanation was
`an extremely rapid metabolism and inacti-
`vation of GLP-1. With inspiration from Rolf
`
`4 2 2 0
`
`jci.org Volume 127 Number 12 December 2017
`
`H A R R I N G T O N P R I Z E E S S A Y
`
`MPI EXHIBIT 1018 PAGE 4
`
`

`

`The Journal of Clinical Investigation
`
`Figure 4. GLP-1 directly stimulates signal transduction and glucose-dependent insulin secretion in islet cells. GLP-1(7-37) directly stimulates cyclic AMP
`accumulation (A), insulin gene expression (B), and glucose-dependent insulin secretion (C) in a rat insulinoma cell line, as adapted from ref. 23.
`
`and CNS. He isolated human neonatal
`brainstem cDNAs encoding proglucagon,
`which exhibited an identical sequence to
`that described for islet proglucagon (51).
`Molecular cloning of the rat intestinal pro-
`glucagon cDNA similarly revealed an open
`reading frame identical to that elucidated
`for the rat pancreatic islet proglucagon
`cDNA (52). Moreover, in studies carried
`out in collaboration with Patricia Brubak-
`er, forskolin, cholera toxin, and dibutyryl
`cyclic AMP increased the synthesis and
`secretion of intestinal PGDPs from primary
`cultures of rat intestinal cells (52). At the
`time, there were no differentiated GLP-1–
`secreting enteroendocrine cell lines suit-
`able for studies of intestinal proglucagon
`gene expression. Accordingly, Ying Lee, a
`fellow in the Drucker laboratory, generated
`a transgenic mouse expressing the SV40 T
`antigen cDNA under the control of the pro-
`glucagon gene promoter. This transgenic
`mouse reproducibly developed GLP-1–
`secreting enteroendocrine tumors of the
`colon (53), enabling isolation of the first
`differentiated GLP-1–producing entero-
`endocrine L cell line in 1992, designated
`GLUTag cells. GLUTag cells were easily
`propagated ex vivo; secreted immunoreac-
`tive GLP-1, glicentin, oxyntomodulin, and
`GLP-2 in response to cyclic AMP analogues
`(54, 55); and resembled primary cultures
`of nonimmortalized gut endocrine cells
`in regard to their response to a battery of
`secretagogues (56).
`
`Two unexpected observations were
`made during isolation of GLUTag cells.
`First, mice harboring s.c. GLUTag cell
`tumors exhibited a marked reduction
`of pancreatic islet α cell mass (57). Sec-
`ond, mice with s.c. GLUTag, InR1-G9, or
`RIN1056A glucagon-producing
`tumors
`all exhibited marked enlargement of the
`small bowel. These findings led Drucker
`to reinvestigate the link between glucagon-
`producing tumors and gut growth, first
`reported in a human subject studied at the
`Hammersmith hospital in 1970 by Dowl-
`ing and colleagues in London (58). A series
`of simple experiments from the Drucker
`lab published in 1996 identified GLP-2 as
`the PGDP with the most potent intestino-
`trophic activity in mice (58). Remarkably,
`although immunoreactive GLP-2 had been
`detected in intestinal extracts of various
`species (refs. 16, 59, and Figure 3), no pre-
`vious biological activity had yet been iden-
`tified for GLP-2 in vivo.
`The actions of GLP-2 to stimulate
`small bowel growth were rapid, detectable
`within days, and associated with increased
`crypt cell proliferation (58). Surprising-
`ly, when similar doses of GLP-2 were
`administered to rats, intestinal growth
`was not significantly increased, although
`an increase in crypt plus villus height was
`observed (60). With hindsight, these find-
`ings reflected the importance of DPP-4 for
`the degradation of GLP-2, more evident
`in rats than in mice. Subsequent studies in
`
`the Drucker lab demonstrated that native
`GLP-2 robustly increased small bowel
`growth in Fischer 344 rats with an inac-
`tivating mutation in the Dpp4 gene (60).
`Furthermore, a GLP-2 analogue with a sin-
`gle amino acid substitution [Gly2]-GLP-2
`exhibited substantial resistance to DPP-4
`cleavage and robust intestinotrophic activ-
`ity in normal rats in vivo (60). Hence, the
`importance of DPP4 for cleavage of both
`GLP-1 and GLP-2 became evident very
`early in the study of the GLPs.
`The identification of GLP-2 as an
`intestinal growth factor spurred a series
`of experiments examining the actions of
`GLP-2 in the context of experimental gut
`injury. GLP-2 administration was gener-
`ally associated with preservation of gut
`mucosal structure and function in the
`setting of chemical, radiation, or surgical-
`ly induced intestinal injury in preclinical
`studies (61–63). Notably, GLP-2 also rapid-
`ly increased nutrient absorption in normal
`rodents (64) and in animals with surgical
`gut resection mimicking SBS (65). Excit-
`ingly, the findings in animals were soon
`extended to humans with SBS. In a pilot
`study carried out around 2000–2001,
`Jeppesen and colleagues demonstrated
`that native GLP-2 administered twice dai-
`ly for 35 days increased nutrient absorp-
`tion, energy absorption, and weight gain
`in human subjects with SBS (66). These
`findings, namely expansion of intesti-
`nal mucosal surface area coupled with
`
`jci.org Volume 127 Number 12 December 2017
`
`4 2 2 1
`
`H A R R I N G T O N P R I Z E E S S A Y
`
`MPI EXHIBIT 1018 PAGE 5
`
`

`

`The Journal of Clinical Investigation
`
`generation of Glp1r–/– mice. These ani-
`mals generated by Louise Scrocchi exhib-
`ited impaired oral glucose tolerance and
`reduced insulin levels after glucose stim-
`ulation, demonstrating the critical role of
`endogenous GLP-1 as an incretin hormone
`in 1996 (69). Unexpectedly, Glp1r–/– mice
`also exhibited fasting hyperglycemia, and
`impaired i.p. glucose tolerance, establish-
`ing the importance of the GLP-1 for β cell
`function beyond its original description as
`an incretin. Moreover, GLP-1R–deficient β
`cells exhibit enhanced sensitivity to apop-
`totic injury, whereas pharmacological acti-
`vation of GLP-1R signaling attenuated β
`cell death in mice in vivo, as well as in cul-
`tures of purified rat β cells studied ex vivo
`(70). The importance of GLP-1R signaling
`for the response to cellular stress was high-
`lighted by findings that activation of GLP-
`1R signaling attenuated the development
`of ER stress in β cells by enhancing ATF-4
`induction and accelerating recovery from
`translational repression via augmentation
`of ER stress–stimulated ATF-4 translation
`(71). Collectively, these findings explain
`how GLP-1, despite acting to simultane-
`ously enhance insulin biosynthesis and
`secretion, maintains functional β cell mass
`through attenuation of ER stress.
`The actions of GLP-1 to stimulate insu-
`lin and inhibit glucagon secretion, together
`with its inhibitory effects on food intake
`and gastric emptying, spurred considerable
`effort in development of GLP-1–based ther-
`apeutics. Remarkably, the first GLP-1R ago-
`nist approved for clinical use was exenatide
`(synthetic exendin-4), a peptide originally
`isolated from Heloderma suspectum lizard
`venom by John Eng in 1992 (72). Subse-
`quent molecular cloning studies published
`by the Drucker laboratory in 1997 demon-
`strated that the lizard genome contains
`two distinct proglucagon gene sequences
`encoding GLP-1 and a separate proexendin
`gene, largely restricted in its expression to
`the salivary gland (73). The original clinical
`development program for exendin-4 uti-
`lized twice-daily injections of the unmodi-
`fied peptide. Pivotal studies in subjects with
`T2D ultimately resulted in the approval
`of twice-daily exenatide, the first GLP-1R
`agonist in 2005 (Figure 1). These efforts
`spurred the development of a once-weekly
`form of exenatide in a microsphere suspen-
`sion, ultimately the first once-weekly ther-
`apy approved for diabetes in 2012 (Figure
`
`Figure 5. Truncated forms of GLP-1 stimulate glucose-
`dependent insulin secretion. GLP-1(7-37) (A) but
`not GLP-1(1-37) (B) stimulates insulin secretion from
`the perfused rat pancreas as adapted from ref. 24.
`Sequences of GLP-1(1-37), GLP-1(7-37), and GLP-1
`(7-36amide). (C) Intraluminal glucose stimulates entero-
`glucagon, GLP-1, and GLP-2 secretion from the perfused
`pig ileum (D), as adapted from ref. 17. Demonstration
`that GLP-1(7-36)amide stimulates insulin secretion from
`the perfused pig pancreas (E), as adapted from ref. 25.
`
`enhanced nutrient absorption (67), togeth-
`er with substantial preclinical data, sup-
`ported the initiation of a drug development
`program to test the efficacy of GLP-2 in
`human subjects with parenteral nutrition–
`dependent (PN-dependent) SBS. Clinical
`studies were initiated with h[Gly2]-GLP-2,
`a degradation-resistant GLP-2 analogue
`discovered in the Drucker lab (60) and sub-
`sequently designated teduglutide. Once
`daily teduglutide administration in human
`subjects with PN-dependent SBS resulted
`
`in increased fluid and energy absorption,
`while reducing the requirements for PN, in
`two separate placebo-controlled Phase 3
`studies (67, 68). Teduglutide was approved
`for chronic therapy of subjects with
`PN-dependent SBS in the US in Decem-
`ber 2012 (67).
`
`The physiology of GLP-1 action
`Complementary studies in the Drucker lab
`were focused on defining the physiological
`importance of endogenous GLP-1 through
`
`4 2 2 2
`
`jci.org Volume 127 Number 12 December 2017
`
`H A R R I N G T O N P R I Z E E S S A Y
`
`MPI EXHIBIT 1018 PAGE 6
`
`

`

`The Journal of Clinical Investigation
`
`1). Exenatide once weekly was more effec-
`tive than the twice-daily preparation, with
`greater reduction of glycemia and compa-
`rable control of body weight (74). Today,
`multiple GLP-1R agonists (small peptides
`and high molecular weight proteins) are
`approved for the treatment of T2D, and a
`single drug, liraglutide, was approved for
`the treatment of obesity in 2014 (Figure 1).
`The widespread distribution of GLP-1
`receptors in extrapancreatic tissues stimu-
`lated considerable research into the nong-
`lycemic actions of GLP-1. Indeed, we now
`understand that GLP-1 controls inflam-
`mation (75), reduces experimental kidney
`injury (76), and like GLP-2, acts as a potent
`intestinal growth factor through mech-
`anisms including stimulation of crypt
`fission (ref. 77 and Figure 6). Among the
`actions of GLP-1 that have engendered the
`most interest are its effects in the cardio-
`vascular system. Native GLP-1 increases
`flow-mediated vasodilation, enhances
`heart rate (HR) and cardiac output, and
`is cardioprotective in preclinical studies,
`most notably in animals with ischemic
`cardiac injury (78). Although degradation-
`resistant GLP-1R agonists have minimal
`effects on blood vessels (79), they increase
`HR and reduce cardiac
`inflammation
`and infarct size in animals with experi-
`mental myocardial infarction (75, 80).
`Understanding the mechanisms through
`which GLP-1 exerts cardioprotection is
`challenging, since the GLP-1R is largely
`expressed in atrial and not ventricular car-
`diomyocytes (81). Furthermore, the GLP-
`1R–dependent control of inflammation in
`heart and blood vessels is complex, as the
`predominant site of GLP-1R expression in
`the (murine) immune system is within the
`intestinal intraepithelial lymphocyte (82).
`Several cardiovascular outcome stud-
`ies have documented the safety of GLP-1R
`agonists in human subjects with T2D and
`preexisting cardiovascular disease (83, 84).
`Excitingly, reduction of major adverse car-
`diovascular events was reported in human
`subjects treated with long-acting liraglutide
`and semaglutide (85, 86). These findings,
`reve

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