throbber
OPEN ACCESS
`
`EDITED BY
`Christoph Thiemermann,
`Queen Mary University of London,
`United Kingdom
`
`REVIEWED BY
`Marcin Filip Osuchowski,
`Ludwig Boltzmann Institute for
`Experimental and Clinical Traumatology,
`Austria
`Basilia Zingarelli,
`Cincinnati Children’s Hospital Medical
`Center, United States
`
`*CORRESPONDENCE
`Jesse Roth
`jroth2@northwell.edu
`
`†These authors have contributed
`equally to this work and share
`first authorship
`
`RECEIVED 19 January 2023
`ACCEPTED 26 April 2023
`PUBLISHED 17 May 2023
`
`CITATION
`Mehdi SF, Pusapati S, Anwar MS,
`Lohana D, Kumar P, Nandula SA,
`Nawaz FK, Tracey K, Yang H, LeRoith D,
`Brownstein MJ and Roth J (2023)
`Glucagon-like peptide-1: a multi-faceted
`anti-inflammatory agent.
`Front. Immunol. 14:1148209.
`doi: 10.3389/fimmu.2023.1148209
`
`COPYRIGHT
`© 2023 Mehdi, Pusapati, Anwar, Lohana,
`Kumar, Nandula, Nawaz, Tracey, Yang,
`LeRoith, Brownstein and Roth. This is an
`open-access article distributed under the
`terms of the Creative Commons Attribution
`License (CC BY). The use, distribution or
`reproduction in other forums is permitted,
`provided the original author(s) and the
`copyright owner(s) are credited and that
`the original publication in this journal is
`cited, in accordance with accepted
`academic practice. No use, distribution or
`reproduction is permitted which does not
`comply with these terms.
`
`TYPE Review
`PUBLISHED 17 May 2023
`DOI 10.3389/fimmu.2023.1148209
`
`Glucagon-like peptide-1:
`a multi-faceted
`anti-inflammatory agent
`
`Syed Faizan Mehdi 1†, Suma Pusapati 1†,
`Muhammad Saad Anwar 1, Durga Lohana 1, Parkash Kumar 1,
`Savitri Aninditha Nandula 1, Fatima Kausar Nawaz 1,
`Kevin Tracey 1, Huan Yang 1, Derek LeRoith 2,
`Michael J. Brownstein 3 and Jesse Roth 1*
`
`1The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States,
`2Division of Endocrinology, Diabetes & Bone Disease, Icahn School of Medicine at Mt. Sinai,
`New York, NY, United States, 3Azevan Pharmaceuticals, Bethlehem, PA, United States
`
`Inflammation contributes to many chronic conditions. It is often associated with
`circulating pro-inflammatory cytokines and immune cells. GLP-1 levels correlate
`with disease severity. They are often elevated and can serve as markers of
`inflammation. Previous studies have shown that oxytocin, hCG, ghrelin, alpha-
`MSH and ACTH have receptor-mediated anti-inflammatory properties that can
`rescue cells from damage and death. These peptides have been studied well in
`the past century. In contrast, GLP-1 and its anti-inflammatory properties have
`been recognized only recently. GLP-1 has been proven to be a useful adjuvant
`therapy in type-2 diabetes mellitus, metabolic syndrome, and hyperglycemia. It
`also lowers HbA1C and protects cells of the cardiovascular and nervous systems
`by reducing inflammation and apoptosis. In this review we have explored the link
`between GLP-1, inflammation, and sepsis.
`
`KEYWORDS
`
`GLP-1 - glucagon-like peptide-1, incretin, GLP-1 agonists, hormone, inflammation,
`anti-inflammation
`
`1 Introduction to GLP-1
`
`Glucagon-like peptide-1 (GLP-1) is a peptide hormone that is produced in the intestine
`and in multiple other sites that are known for their role in regulating glucose metabolism.
`GLP-1 is also involved in multiple other physiological processes including appetite,
`cardiovascular function, and inflammation (1).
`Acute Inflammation is central to in-vivo responses to a wide range of challenges
`including viral and bacteriological infections, and to host repair processes. Chronic
`inflammation, on the other hand, is associated with conditions like type 2 diabetes,
`metabolic syndrome, obesity, cancer, arthritis, and bowel diseases like Crohn’s disease and
`
`Frontiers in Immunology
`
`01
`
`frontiersin.org
`
`Novo Nordisk Exhibit 2016
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00001
`
`

`

`Mehdi et al.
`
`10.3389/fimmu.2023.1148209
`
`ulcerative colitis (2). Our recent studies have revealed the anti-
`inflammatory properties of several peptide hormones such as hCG,
`oxytocin, ghrelin, and vasopressin.(3-6) In this review article, we
`focus on the anti-inflammatory properties of the incretin hormone
`‘Glucagon-like Peptide-1 (GLP-1). Known for promoting glucose
`homeostasis and weight loss, the anti-inflammatory properties of
`GLP-1 suggest that it may also blunt inflammation and protect
`against organ damage (3–6).
`
`2 Functions of GLP-1 and its receptors
`
`Glucagon-like peptide-1 (GLP-1) is a 30-31 amino acid long
`incretin that is produced when proglucagon undergoes post-
`translational processing. This glucose-lowering agent is secreted
`by intestinal enteroendocrine L-cells in response to nutritional and
`inflammatory stimuli and by neurons in the nucleus of the solitary
`tract in the brainstem. GLP-1 activates a seven transmembrane G
`protein coupled receptor, GLP-1R. GLP-1 receptors are expressed
`in pancreatic islet b-cells, pulmonary epithelial cells, atrial cardiac
`myocytes, vagal afferent neurons, neurons in a number of brain
`regions, as well as cells lining gastric pits and small intestinal
`mucosa. The GLP-1R can couple to the Gs or Gq proteins,
`leading to increases in intracellular cAMP and/or Ca2+ levels and
`activation of PKA, Epac-2, phospholipase C and ERK1/2 signal
`transduction pathways. Activation of GLP-1R by GLP-1 or other
`exogenous agonists, including exendin-4 and liraglutide, decreases
`inflammatory responses in several animal models like rat heart and
`whole animal model. The hypoglycemic activity of GLP-1 is
`associated with the stimulation of glucose-dependent insulin
`secretion, inhibition of glucagon production and regulation of
`islet cell proliferation, differentiation, and survival. Under
`physiological conditions, GLP- 1 is rapidly degraded by dipeptidyl
`peptidase-4 (DPP- 4) after it is released (7).
`
`3 Discovery of GLP-1
`
`In 1923, Charles Kimball and John Murlin, in an attempt to
`purify commercial insulin, precipitated a pancreatic fraction that had
`a hyperglycemic effect (8). Identifying it as a secreted factor, they
`named it ‘Glucagon’ or ‘Glucose Agonist’. In 1959, Roger Unger et al.,
`developed the first antibody that could be used in a
`radioimmunoassay to detect glucagon in tissue samples and blood
`(9, 10). In 1966, Ellis Samols, Vincent Marks and others confirmed
`the presence of glucagon-like immunoreactivity in extra-pancreatic
`tissue, especially the intestine. Subsequently in 1967, Samols and
`Marks reported glucagon-like material
`in pancreatectomized
`humans, indicating its extra-pancreatic origin (11). In 1968, Roger
`Unger demonstrated that intraduodenal administration of glucose
`increased the levels of a circulating glucagon-like substances (9, 10).
`In contrast to glucagon, the intestinal glucagon-like material
`stimulated the release of insulin. It was clear that glucagon and the
`g l u c a g o n - l i k e m a t e r i a l w e r e d i s t i n c t e n t i t i e s , a n d
`immunohistochemical studies revealed that intestinal cells that
`were positive for the glucagon-like material had a different
`
`morphology from glucagon secreting a-cells. The cells that made
`glucagon-like material were called L-cells. In 1970 the glucagon
`precursor, proglucagon, was identified. In the pancreas,
`proglucagon undergoes post-translational cleavage yielding two
`fragments. One was a mature glucagon and the other was called
`the proglucagon fragment. In 1980, the intestinal glucagon-like
`material, glicentin, was identified along with a smaller species
`named oxyntomodulin in 1982. Collectively, these studies suggested
`that proglucagon undergoes tissue-specific processing resulting in
`formation of glicentin and oxyntomodulin in the intestine, and
`glucagon plus the N-terminal fragment of glicentin in the pancreas.
`In the 1980s Joel Habener described a new glucagon-related peptide
`encoded in the anglerfish preproglucagon cDNA. Subsequently two
`glucagon-related peptides were identified in rat, bovine, hamster, and
`human proglucagon. These two peptides are now called glucagon-like
`peptides 1 and 2 (GLP-1 and GLP-2) as shown in Figure 1 “The
`proglucagon precursor (12–16).
`
`4 GLP-1 receptor
`
`The GLP-1 receptor is a member of the secretin subfamily (B1)
`of G-protein coupled receptors (GPCRs). It consists of 463 amino
`acids (17). These amino acids are arranged in seven transmembrane
`(7TM) alpha-helices with an N-terminal domain that is located
`extracellularly and a C-terminal domain that is intracellular. The
`transmembrane helices are connected by three extracellular and
`three intracellular loops (17, 18). Ligand binding to its receptor
`occurs in two stages. The first step involves the binding of the
`extracellular domain to the C-terminus of the ligand. This causes a
`conformational shift that leads to attachment of the N-terminus of
`the ligand to the 7TM domain (18–20). (Figure 2) for a more
`detailed figure, refer to reference 18.
`GLP-1 receptor signaling occurs primarily through the Gas
`stimulatory G protein (21). Coupling of Gas and Gaq in beta cells
`of pancreas lead to an increase in cAMP by activation of adenylyl
`cyclase and phosphoinositol 3 kinase (PI3K) pathway. cAMP
`activates PKA and Epac-2 signal transduction pathways (22).
`PKA and Epac-2 inhibit the K-channel, altering Kv currents
`leading to calcium influx as well as calcium release from the
`endoplasmic reticulum. This results in calcium-induced release of
`insulin granules (23). PKA and Epac-2 also activate cyclin D and
`CREB,
`leading to beta-cell proliferation, differentiation, and a
`decrease in endoplasmic reticulum stress response (24). Exenatide
`decreases ER stress in response to synthetic stressors (25). In mouse
`models, exendin-4 increases beta cell proliferation by activation of
`epidermal growth factor receptors (26). Human beta cells exposed
`to GLP-1 show increased beta cell proliferation (24, 25) (Figure 3).
`Activation of the GLP-1 pathways decreases the inflammatory
`response in multiple models. GLP-1 Analog liraglutide improves
`vascular function in polymicrobial sepsis by reduction of oxidative
`stress and inflammation (24, 27–31). Exendin in diabetic mice
`diminishes inflammatory responses by increasing the expression
`of regulatory T cells (32). Liraglutide has anti-inflammatory effects
`on endothelial cells by decreasing activation of NF-kB, inhibiting
`TNF-alpha, and increasing nitric oxide production (28). Like GLP-1
`
`Frontiers in Immunology
`
`02
`
`frontiersin.org
`
`Novo Nordisk Exhibit 2016
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00002
`
`

`

`Mehdi et al.
`
`10.3389/fimmu.2023.1148209
`
`FIGURE 1
`GLP-1 synthesis: In the intestine — The proglucagon precursor gives rise to oxyntomodulin, GLP1 (and its two equipotent, truncated derivatives) and
`GLP-2. Like the GLPs, the intervening peptides (IP-1 and IP-2) may also have physiological functions (12, 13). In the pancreas, the proglucagon
`precursor yields glucagon and the glicentin-related pancreatic peptide (GRPP) (12–16) Figure modified from article 12. Figure from Open access
`(Molecular Metabolism) permissible to re-use under a CC-BY 4.0 license.
`
`agonists, dipeptidyl peptidase-4 (DPP-4) inhibitors, which block the
`degradation of GLP-1, also cause attenuation of the inflammatory
`responses. Sitagliptin decreases the LPS-inflammatory response by
`inhibiting the NF-kB pathway. This leads to decreased production
`of proinflammatory cytokines including TNF-a, IL-6, IL-1b and
`decreased expression of COX-2 in cardiomyocytes (33).
`
`5 GLP-1 and various organs
`
`GLP-1 has been shown to carry out numerous protective and
`regulatory functions in different organ systems. The functions are
`illustrated in the Figure (Figure 4).
`
`6 GLP-1: anti-inflammatory effects
`
`6.1 Cardiovascular system
`
`The antioxidant and anti-inflammatory effects of GLP-1 protect
`the cardiovascular system. GLP-1 levels are elevated in patients post
`myocardial infarction. Administration of GLP-1 analogs (or DPP-4
`inhibitors, which inhibit the degradation of GLP-1), decreased
`cardiovascular and thrombotic complications in animal models of
`LPS-induced sepsis. They also suppressed inflammation and
`formation of reactive oxygen species (ROS) in vasculature,
`resulting in vasorelaxation and amelioration of hypotension.
`Moreover, reduced organ damage by thrombotic occlusion in the
`
`FIGURE 2
`(Original by authors of the manuscript): The seven transmembrane alpha-helices are bound to G-protein subunits (19). These consist of alpha
`subunit and beta-gamma subunit complexes bound to GDP. In the inactive state, the a-subunit is bound to GDP. Upon binding of GLP-1, GDP is
`replaced by GTP, which then activates the a-subunit. The a-subunit and GTP complex activate signaling cascades through adenylyl cyclase and
`phospholipase C (20).The third intracellular loop is most important in receptor signaling.
`
`Frontiers in Immunology
`
`03
`
`frontiersin.org
`
`Novo Nordisk Exhibit 2016
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00003
`
`

`

`Mehdi et al.
`
`10.3389/fimmu.2023.1148209
`
`FIGURE 3
`GLP-1 receptor signaling (21-26). Figure modified from article 23. Figure from Open access (Gastroenterology) permissible to re-use under a CC-BY
`4.0 license.
`
`lung has been reported in LPS-induced sepsis due to improvement
`in microvascular circulation by GLP-1 analogs. In a polymicrobial
`model of sepsis induced by cecal ligation and puncture, a GLP-1
`analog ameliorated vascular inflammation and oxidative stress by
`improving endothelial function (28)
`In a cardiac fibrosis model, the GLP-1 analogue liraglutide
`reduced vascular reactivity, cardiac hypertrophy, fibroblast
`accumulation, collagen deposition and MCP-1 production (40).
`
`Another GLP-1 analogue, exendin-4, also prevented cardiac
`remodeling and diastolic dysfunction in an experimental diabetes
`model. This was associated with a reduction in macrophage
`infiltration, lower expression of IL-1b and IL-6, and an increase
`in IL-10 in the heart (41).
`GLP-1 improved left ventricular function in patients with
`chronic heart failure and in dogs with dilated cardiomyopathy.
`Survival rates after myocardial infarction also improved after GLP-1
`
`FIGURE 4
`(Original by authors of the manuscript): Effects of GLP-1 on various organ systems (34–39).
`
`Frontiers in Immunology
`
`04
`
`frontiersin.org
`
`Novo Nordisk Exhibit 2016
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00004
`
`

`

`Mehdi et al.
`
`10.3389/fimmu.2023.1148209
`
`administration. Sitagliptin, a DPP-4 inhibitor, improves myocardial
`response in coronary artery disease patients. LPS-induced cardiac
`dysfunction recovered in DPP-4 deficient rats after treatment with
`sitagliptin. Exendin-4 and DPP-4 de ficiency prevented
`vasoconstriction and multiple organ injury after LPS treatment,
`and improved survival in endotoxemic rats (31).
`In animal studies, GLP-1 and its analogs reduced macrophage
`infiltration in blood vessels, and production of pro-inflammatory
`cytokines such as IL-6, IL-1b, TNF-a, and CRP. It has been
`speculated that liraglutide, a GLP-1 analog, suppresses cytokine
`release in bacterial septic shock and in SARS-CoV-2 viral sepsis.
`GLP-1 analogs and DPP-4 inhibitors have shown promise in animal
`models of cardiovascular disease. Studies in humans should be done
`(42, 43) (Table 1).
`
`6.2 Gastrointestinal system
`
`GLP-1 is secreted into the distal intestine by enteroendocrine L
`cells in response to nutrient ingestion (42). GLP-1 receptors are
`widely distributed in the gastrointestinal tract, pancreas, heart, lungs,
`kidneys, and nervous system. These receptors contribute to the wide
`range of physiological functions (45). Besides metabolic effects, GLP-
`
`1 improves mucosal integrity and diminishes inflammation (42, 46).
`Exendin-4, a GLP-1 mimetic peptide, decreases the production of
`pro-inflammatory cytokines, and diminishes the enteric immune
`response. GLP-1 decreases production of pro-inflammatory
`cytokines, mainly by downregulating NF-kB phosphorylation and
`nuclear translocation (45).
`Several recent studies have suggested that GLP-1 should be
`considered as a treatment for a wide range of intestinal diseases,
`including Inflammatory bowel diseases, intestinal mucositis, coeliac
`disease and short bowel syndrome (45). GLPs, (including GLP-1,
`GLP-2 and DPP-4) have recently gained increased attention from
`researchers studying Inflammatory bowel diseases (IBDs).
`IBDs including Crohn’s disease and ulcerative colitis are
`chronic relapsing-remitting diseases with multifactorial etiologies
`and complex pathogenesis. The Incidence and prevalence of IBDs
`are rising globally. GLPs including GLP-1 regulate weight and
`glycemia. GLP-1 also inhibits gastric emptying, decreases food
`ingestion, and increases crypt cell proliferation. It also improves
`intestinal growth and nutrient absorption. GLPs have been
`proposed to improve tissue healing of injured epithelium, regulate
`T-cell growth and function, control innate immune cells such as
`macrophages and dendritic cells, and lower pro-inflammatory
`cytokines in IBD (47) (Table 2).
`
`Nephropathy¶
`
`RetinopathyD
`
`Cardiovascular
`Overall
`mortality
`
`Benefit
`
`Benefit
`
`?
`
`Benefit
`
`Benefit
`
`Neutral
`
`?
`
`Neutral
`
`Neutral
`
`Unexpected increase
`in retinopathy
`outcomes◊
`
`Benefit
`
`?
`
`Benefit
`
`Benefit
`
`Benefit
`
`?
`
`Benefit
`
`?
`
`TABLE 1 Efficacy of GLP-1 agonists efficacy (adapted from UpToDate) (44).
`
`Elimination
`half-life
`
`Glycemic
`efficacy (reduction
`in
`A1C in
`% points)*
`
`Cardiovascular out-
`comes
`ASCVD/HF
`
`Long-acting GLP-1 receptor agonists (more pronounced effect on fasting glucose)
`
`Dulaglutide
`
`5 days
`
`Efpeglenatide
`
`6 to 7 days
`
`Exenatide
`
`8 to 14 days
`
`Liraglutide
`
`11 to 15 hrs
`
`–1 to –1.5
`
`–1 to –1.11
`
`–1.5 to –1.6
`
`–0.8 to –1.5
`
`Semaglutide
`
`6 to 7 days
`
`–1.5 to –2
`
`Benefit
`
`Benefit
`
`Neutral
`
`Benefit
`
`Benefit
`
`Short-acting GLP-1 receptor agonists (more pronounced effect on postprandial glucose)
`
`Exenatide
`
`2 to 3 hrs
`
`–1
`
`Lixisenatide
`
`3 to 5 hrs
`
`–0.8 to –1
`
`Dual-acting GLP-1 and GIP receptor agonists
`
`?
`
`Neutral
`
`Tirzepatide
`
`5 days
`
`–2 to –2.5
`
`?§
`
`?
`
`Neutral
`
`?
`
`?
`
`?
`
`?
`
`GLP-1, glucagon-like peptide 1; A1C, glycated hemoglobin; ASCVD, atherosclerotic cardiovascular disease; HF, heart failure; SubQ, subcutaneously; ?, inadequate data; GIP, glucose-dependent
`insulinotropic polypeptide; eGFR, estimated glomerular filtration rate.
`*Reduction in A1C is dependent upon a number of factors, such as baseline A1C and background therapy. In trials directly comparing shorter- versus longer-acting GLP-1 receptor agonists,
`longer-acting had better glycemic efficacy.
`¶Nephropathy is defined as elevated albuminuria, reduced eGFR (usually <60 mL/min/1.73 m2), or both.
`DRetinopathy outcomes were not systematically evaluated or adjudicated.
`◊The higher rate of retinopathy complications with subcutaneous semaglutide was unexpected and may be a consequence of rapid glycemic control similar to that seen in other settings. If
`subcutaneous semaglutide is prescribed to a patient with diabetic retinopathy, titrate slowly to avoid rapid declines in A1C and perform retinal screening within 6 months of drug initiation to
`detect progression of retinopathy.
`§In preliminary trials, tirzepatide did not increase the risk of major cardiovascular events.
`
`Frontiers in Immunology
`
`05
`
`frontiersin.org
`
`Novo Nordisk Exhibit 2016
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00005
`
`

`

`Mehdi et al.
`
`10.3389/fimmu.2023.1148209
`
`6.3 Hepatobiliary system
`
`GLP-1 based therapies have shown promise in liver diseases e.g.
`non-alcoholic fatty liver disease (NAFLD) and non-alcoholic
`steatohepatitis (NASH). In recent years, the prevalence of non-
`alcoholic fatty liver disease (NAFLD) has continued to rise, and
`10%-25% of NAFLD cases progress to non-alcoholic steatohepatitis
`
`(NASH). 10%-15% of NASH cases will develop into hepatocellular
`carcinoma, approximately 700,000 people die from the disease each
`year (53).
`Nonalcoholic steatohepatitis is associated with inflammation
`of the liver, driven by an aberrant accumulation of fat. In rats fed
`with a high-fat diet, treatment with liraglutide, a GLP-1R analog,
`reduced steatosis and lobular inflammation compared to the
`
`TABLE 2 GLP-1 analogues under investigation in vitro and in vivo (animal and human studies).
`
`Models
`
`Treatments
`
`In-vitro study
`Macrophage RAW
`264.7
`cell culture
`
`Pre-treated with
`exendin-4 for 6hrs
`followed by LPS for
`24hrs
`
`Exendin-4 inhibits production of many LPS-induced inflammatory factors, thereby decreasing production
`of ROS reactive oxygen species.
`
`(Lu et al.,
`2019) (27)
`
`Results
`
`References
`
`GLP-1 self-
`associated with
`PEGylated
`phospholipid
`micelles i.p
`
`—
`
`GLP-1-SSM (sterically stabilized phospholipid micelles) improve architecture of the intestine, partially
`preserve goblet cell number, decrease IL1-b secretion and improve diarrhea induced by DSS.
`
`(Anbazhagan
`et al., 2017)
`(48)
`
`I.P or I.V administration of LPS caused a significant rise in plasma levels of GLP-1 through the TLR-4
`mechanism.
`
`(Lebrun et al.,
`2017) (46)
`
`Exendin-4 (GLP-1
`agonist) s.c.
`
`DSS-induced colitis in GLP-1 R knockout mice showed dysregulation of intestinal gene expression, as
`well as abnormal representation of microbes in feces and increased sensitivity to intestinal injury. Also,
`Exendin-4 administration caused significantly increased expression of genes encoding cytokines and
`chemokines in gut injury.
`
`(Yusta et al.,
`2015) (49)
`
`LPS+/-Exendin-4
`
`Exendin-4 Inhibited production of pro-inflammatory cytokines including TNF-a and IL-1 a in LPS-
`induced inflammation in mouse model.
`
`(Al-Dwairi
`et al., 2018)
`(50)
`
`(Işbil
`Büyükcoşkun
`et al., 2007)
`(51)
`
`In-vivo animal
`studies
`DSS-induced colitis
`in
`mouse model
`
`DSS-induced colitis
`in
`mouse model
`— Ischemia/
`reperfusion
`
`DSS-induced colitis
`in
`mouse with GLP-
`1R
`knockout
`
`Colonic smooth
`muscle
`cells of male
`BALB/c
`mice cultured in
`DMEM
`
`Wistar rat model
`
`MPTP-treated
`Parkinson
`Disease mouse
`model
`Human A53T
`a-synuclein
`transgenic
`PD mouse
`model
`(MPTP=1-methyl-
`4-phenyl-1,2,3,6-
`tetrahydropyridine)
`
`Human study
`a) Healthy
`volunteers
`b)Ischemia/
`reperfusion injury
`model of human
`gut
`
`a) GLP-1 injected
`i.c.v
`b) GLP-1 receptor
`antagonist, Exendin
`9-39
`I.c.v and i.p
`
`CCK analogues
`or
`Liraglutide and
`GLP-1 analogues
`i.p
`
`—
`
`Centrally injected Exendin 9-39 inhibited the gastroprotective effects of GLP-1 agonists,
`suggesting that this effect is managed by central mechanisms.
`
`CCK analogues or GLP-1 analogues restored the disruption of intestinal tight junction, reduced colonic
`inflammation, inhibited colonic dopaminergic neuron reduction and the accumulation of a-synuclein
`oligomers in the colon of both PD mouse models.
`
`(Su et al.,
`2022) (52)
`
`3hrs after LPS injection
`a)
`plasma GLP-1 levels
`rose significantly.
`b)
`45 min after ischemia
`in the human intestine,
`GLP-1 levels rose
`significantly and
`returned to baseline
`after reperfusion.
`
`(Lebrun et al.,
`2017) (46)
`
`Frontiers in Immunology
`
`06
`
`frontiersin.org
`
`Novo Nordisk Exhibit 2016
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00006
`
`

`

`Mehdi et al.
`
`10.3389/fimmu.2023.1148209
`
`saline-injected group. Exendin-4, a GLP-1R agonist, was shown in
`another study to lower hepatic production of the inflammatory
`markers TNF-, IL-1, and IL-6, as well as macrophage markers
`cluster of differentiation 68 (CD68), and F4/80 in mice fed a
`western-type (high fat) diet (54).
`C-reactive-protein (CRP) is produced by the liver and is a
`marker of inflammation. Liraglutide produced a significant decrease
`in the mean concentration of CRP in a retrospective investigation of
`110 obese patients with type 2 diabetes mellitus, indicating its
`potential as an anti-inflammatory drug. Exenatide plus metformin
`caused a significant reduction in baseline CRP and TNF-a. These
`findings show that GLP-1-based treatments improve fatty liver
`disease in rats and humans via reducing inflammation (42).
`NAFLD is associated with cell death and fibrosis that ultimately
`progress to cirrhosis. In obese patients with NAFLD, Fibroblast
`growth factor-21 protein (FGF21) and RNA levels are higher in the
`liver. Treatment with GLP-1R agonists reduced the level of FGF21.
`This supports its use in cirrhosis. Note that 80% of patients who
`develop hepatocellular carcinoma had cirrhosis beforehand (55, 56).
`GLP-1RA significantly reduced cell necrosis and apoptosis, the
`two major forms of liver cell death. Hepatic cell death mainly
`includes two forms: apoptosis and cell necrosis. Gupta et al. showed
`that a GLP-1RA significantly reduced cell necrosis and
`apoptosis.The reduction of abdominal visceral adiposity by GLP-
`1RAs results in a reduction in liver fat content that can alleviate
`NAFLD. The ability of GLP-1 to reduce fat is due to its binding to a
`specific GLP-1R present in adipose tissue (57). Vendrell et al.
`confirmed the expression of GLP-1R in mature adipose cells by
`the detection of the mRNA and protein (58). A 6-month-long
`treatment with GLP-1RAs in obese patients with T2DM resulted in
`significant reductions in intrahepatic lipids (IHL). In addition, the
`median relative reduction in IHL was 42% (53) (Figure 5).
`
`6.4 Central nervous system
`
`Glucagon-like peptide-1 is produced in the brainstem and has
`numerous functions, including neuroprotection (59–61) GLP-1 and
`GLP-1 analogs can cross the blood-brain barrier (62–68) GLP-1
`receptors have been observed in the neurons of the nucleus tractus
`solitarius that project to GLP-1R–expressing regions in the
`hindbrain, hypothalamus, including the paraventricular nucleus
`(PVN), dorsal medial nucleus of the hypothalamus, and arcuate
`nucleus (ARC) (42, 64, 66, 67, 69, 70). GLP-1-based therapies have
`anti-inflammatory effects on multiple tissues (42, 53, 71–73).
`Chronic inflammation is a significant risk factor for many
`neurodegenerative disorders, e.g., Alzheimer’s disease and
`Parkinson’s disease (42, 74–78).
`
`6.4.1 Parkinson’s disease
`The prevalence of Parkinson’s disease has been rising in recent
`years (79, 80). It
`is the second most common chronic
`neurodegenerative disease and affects between 1% - 2% of people
`above age 60 and 4% of those above age 80 (81–87). Parkinson’s
`disease occurs when dopaminergic neurons in the substantia nigra
`pars compacta form Lewy bodies and gradually die (88–90). The
`Lewy body is an abnormal aggregate containing alpha-synuclein.
`Most Parkinson’s disease treatments focus on managing symptoms
`by replacing dopamine and improving dopaminergic signaling, but
`these treatments fail to address the underlying cellular degeneration
`(64, 91). Since dopamine breaks down to form reactive oxygen
`species, it may contribute to disease progression (92, 93). Activation
`of microglia plays a crucial role in spontaneous Parkinson’s Disease
`in humans (64, 94–96). MPTP (1-methyl-4-phenyl-1,2,3,6-
`tetrahydropyridine) induces Parkinson’s disease in rodents.
`MPTP is a pro-drug for the neurotoxin MPP+ (1-methyl-4-
`
`FIGURE 5
`Effects of glucagon-like peptide-1 receptor agonist on non-alcoholic fatty liver disease. PPAR-a, Peroxisome proliferator-activated receptor; IHL,
`intrahepatic lipids; AMPK, AMP-activated protein kinase; CRP, C reactive protein; AGEs, Advanced glycation and end products; JNK, c-Jun NH2-
`terminal kinase; GLP-1RA, Glucagon-like peptide-1 receptor agonist; NAFLD, Non-alcoholic fatty liver disease (53). Modified figure from reference
`53. Figure from Open access (World Journal of Gastroenterology) permissible to re-use under a CC-BY 4.0 license).
`
`Frontiers in Immunology
`
`07
`
`frontiersin.org
`
`Novo Nordisk Exhibit 2016
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00007
`
`

`

`Mehdi et al.
`
`10.3389/fimmu.2023.1148209
`
`phenylpyridinium). This agent destroys dopaminergic neurons in
`the substantia nigra (81, 97–104).Exendin- 4, a GLP-1 R agonist,
`has inhibitory effects on microglial activation and greatly reduces
`the expression of TNF-a and IL-1b caused by MPTP (67, 105–107).
`Exendin-4 inhibits 6-hydroxydopamine (6-OHDA)-induced
`dopaminergic cell death in neuronal culture. The intraventricular
`administration of GLP-1 protects mice from MPTP-induced
`dopaminergic cell loss (64, 86, 108, 109).
`
`6.4.2 Alzheimer’s disease
`Alzheimer’s disease is the most common form of dementia; it is
`responsible for 60–70% of cases (110, 111). About 1 person in 9
`(10.8%) in the US population age 65 and older has AD (112). People
`65+ years of age in Europe had a pooled incidence rate of AD of 19.4
`per 1000 person-years (113–115). The Alzheimer’s disease
`population increased by 5% from age 65 to 73, 13.1% from age 75
`to 84, and 32% from age 85 and older (112, 116). Alzheimer’s
`disease was the seventh-leading cause of death in 2020 and
`2021 (112).
`In AD, IL-1 beta is significantly increased in the frontal cortex
`and hippocampus and may contribute to cognitive dysfunction by
`promoting the synthesis of amyloid precursor protein (117, 118).
`GLP-1 therapies may have preventive and restorative effects on
`Alzheimer’s disease (42, 119). Exogenous GLP-1 (7–36) amide
`administration inhibited IL-1 beta transcription and prevented
`beta-induced amnesia and cell death (36, 59, 113, 114). Also, it
`restores learning and memory by stimulating LTP (long-term
`( 6 0, 1 2 0– 1 2 2 ) .
`p o t e n t i a t i o n )
`I n a r o d e n t m o d e l ,
`neuroinflammation was reduced due to suppression of TNF-alpha
`when GLP-1 exenatide (20 ug/kg/day) was given intraperitoneally.
`The peptide improved memory and prevented the loss of
`hippocampal neurons (111, 123). Treatment with liraglutide in a
`
`mouse model of Alzheimer’s disease reduced the inflammatory
`response in the cortex by decreasing the number of activated
`microglia (60, 65, 107, 124, 125). Mice that express two human
`mutant genes linked to early-onset Alzheimer’s disease develop a
`chronic inflammatory response (126). In these animals, D-Ala2-
`GIP reduces the activation of microglia and astrocytes in the brain,
`decreasing the release of pro-inflammatory cytokines and oxidative
`stress (127).Microglia and astroglia express GIP receptors (128,
`129). Activating them reduces central inflammatory responses. GIP
`receptor activation increases microglia expression of key growth
`factors such as brain-derived neurotrophic factor(BDNF), glial cell-
`line derived neurotrophic factor (GDNF), and nerve growth factor
`(NGF) in a phosphoinositide 3-kinase (PI3K) and protein kinase A
`(PKA) dependent manner (130) (Figure 6).
`Brain irradiation has been demonstrated to increase the
`expression of IL-6, IL-1b, and IL-12p70 cytokines. Liraglutide
`reduces the proinflammatory cytokine gene expression caused by
`X-ray irradiation (42, 131).
`In a study on rats, when cultured astrocytes were stimulated by
`LPS, IL-1b mRNA expression increased temporally. GLP-1 therapy
`decreased IL-1b mRNA production compared to the LPS alone-
`treated cultures (67, 72). The GLP-1 suppresses TNF-alpha and
`associated cytokines in microglia (Figure 7).
`
`6.5 Stroke models
`
`Strokes in the elderly can cause permanent neurological damage
`and are among the leading causes of death. Patients who have
`hyperglycemia and diabetes mellitus type 2 (T2DM) have a higher
`stroke frequency than those who do not have these conditions (132,
`133). Stimulating GLP-1Rs with exendin-4 reduces brain damage
`
`FIGURE 6
`Overview of the main pathways induced by GLP-1 in neurons. Activation of the GLP-1R activates adenylyl cyclase and increases cAMP levels. This
`activates PKA and other downstream kinases related to growth factor signaling. GLP-1 supports neurogenesis, reduces inflammation, and inhibits
`apoptosis while improving learning and memory in the hippocampus.(modify from reference 130) (AATP, adenosine triphosphate; cAMP, Cyclic
`adenosine monophosphate; CREB, cAMP response element binding protein; PKA, protein kinase A; PI3K, phosphatidylinositol-3 kinase; PKC, protein
`kinase c; mTOR, Mammalian target of rapamycin; ERK, extracellular signal-regulated kinase; BRAF, v-raf murine sarcoma viral oncogene homolog
`B1.) Figure modified from reference 130, Open access (Peptides journal) permissible to re-use under a CC-BY 4.0 license).
`
`Frontiers in Immunology
`
`08
`
`frontiersin.org
`
`Novo Nordisk Exhibit 2016
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00008
`
`

`

`Mehdi et al.
`
`10.3389/fimmu.2023.1148209
`
`muscle tissues (142). Covid victims who took GLP-1R agonists had
`fewer hospital admissions (143).
`
`7 GLP-1 in obstructive lung disease
`and asthma
`
`Asthma affects about 25 million people in the US and more than
`330 million people world-wide (144). GLP-1 receptor agonists
`decreased allergic responses in asthma by preventing the activation
`of NF-kB leading to decreased release of proinflammatory cytokines
`(IL-5, IL-13, IL-33) and neutrophils, eosinophils, basophils and CD4
`+ T cell numbers (142, 145). Exendin-4 also relaxes bronchial smooth
`muscles by acting on the cAMP-PKA pathway (144).
`A recent study demonstrated that GLP-1 agonists improve survival
`and lung function in mouse models of asthma and COPD. The results
`showed that GLP-1R agonists have therapeutic potential in the
`treatment of ch

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