throbber
Review Article
`
`Glucagon-like peptide-1 receptor
`agonists as neuroprotective agents for
`ischemic stroke: a systematic scoping
`review
`
`Journal of Cerebral Blood Flow &
`Metabolism
`2021, Vol. 41(1) 14–30
`! The Author(s) 2020
`Article reuse guidelines:
`sagepub.com/journals-permissions
`DOI: 10.1177/0271678X20952011
`journals.sagepub.com/home/jcbfm
`
`, Christian Holscher3
`Mark P Maskery1,2
`,
`Stephanie P Jones4, Christopher I Price5, W David Strain6,
`Caroline L Watkins4, David J Werring7 and
`Hedley CA Emsley1,2
`
`Abstract
`Stroke mortality and morbidity is expected to rise. Despite considerable recent advances within acute ischemic stroke
`treatment, scope remains for development of widely applicable neuroprotective agents. Glucagon-like peptide-1 recep-
`tor agonists (GLP-1RAs), originally licensed for the management of Type 2 Diabetes Mellitus, have demonstrated pre-
`clinical neuroprotective efficacy in a range of neurodegenerative conditions. This systematic scoping review reports the
`pre-clinical basis of GLP-1RAs as neuroprotective agents in acute ischemic stroke and their translation into clinical trials.
`We included 35 pre-clinical studies, 11 retrospective database studies, 7 cardiovascular outcome trials and 4 prospective
`clinical studies. Pre-clinical neuroprotection was demonstrated in normoglycemic models when administration was
`delayed by up to 24 h following stroke induction. Outcomes included reduced infarct volume, apoptosis, oxidative
`stress and inflammation alongside increased neurogenesis, angiogenesis and cerebral blood flow. Improved neurological
`function and a trend towards increased survival were also reported. Cardiovascular outcomes trials reported a signif-
`icant reduction in stroke incidence with semaglutide and dulaglutide. Retrospective database studies show a trend
`towards neuroprotection. Prospective interventional clinical trials are on-going, but initial
`indicators of safety and
`tolerability are favourable. Ultimately, we propose that repurposing GLP-1RAs is potentially advantageous but appro-
`priately designed trials are needed to determine clinical efficacy and cost-effectiveness.
`
`Keywords
`Acute stroke, neuroprotection, reperfusion, clinical trials, animal models
`
`Received 2 January 2020; Accepted 18 July 2020
`
`Introduction
`
`Stroke accounts for 6.5 million deaths per year globally
`and by 2030 will result in an annual loss of over 200
`million disability-adjusted life years.1,2 With an increas-
`ing number of strokes occurring in younger patients,
`alongside an increased number of stroke survivors, the
`cost of post-stroke care is rising. There is, therefore,
`significant scope to improve upon the current position.
`Considerable advances have been made in acute
`ischemic stroke (AIS) treatment, notably reperfusion
`therapies, but these are limited to 10–20% of total
`stroke patients following careful clinical and radiolog-
`ical selection.3 Even when intravenous thrombolysis
`and/or endovascular thrombectomy are administered,
`
`1Lancaster Medical School, Lancaster University, Lancaster, UK
`2Department of Neurology, Royal Preston Hospital, Preston, UK
`3Research and Experimental Center, Henan University of Chinese
`Medicine, Zhengzhou, Henan Province, China
`4Faculty of Health and Wellbeing, University of Central Lancashire,
`Preston, UK
`5Institute of Neuroscience, Stroke Research Group, Newcastle
`University, Newcastle, UK
`6NIHR Exeter Clinical Research Facility and Institute of Biomedical and
`Clinical Science, University of Exeter Medical School, Royal Devon &
`Exeter NHS Foundation Trust, Exeter, UK
`7Stroke Research Centre, Department of Brain Repair and Rehabilitation,
`UCL Institute of Neurology and The National Hospital for Neurology
`and Neurosurgery, London, UK
`
`Corresponding author:
`Mark Maskery, Department of Neurology, Royal Preston Hospital,
`Lancashire Teaching Hospitals NHS Foundation Trust, Sharoe Green
`Lane, Preston PR2 9HT, UK.
`Email: Mark.Maskery@doctors.org.uk
`
`Novo Nordisk Exhibit 2104
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00001
`
`

`

`Maskery et al.
`
`15
`
`reduction in disability is highly time dependent.4,5
`Scope remains for further improvement, especially for
`patients who are unsuitable for reperfusion therapies or
`those within remote environments.
`Using simpler clinical selection processes, neuropro-
`tective therapies could bring benefits to a wider patient
`group. Neuroprotectants could also enhance the bene-
`fits of reperfusion therapies by preservation of the
`ischemic penumbra and reduction in ischemic reperfu-
`sion injury. Despite many demonstrating pre-clinical
`potential, a suitable agent has not yet been identified
`by translational studies.6 There remains a multitude of
`factors affecting the translation from bench-to-bedside.
`Namely, animal models are not perfect in their repre-
`sentation of the heterogeneity of clinical stroke.7 Stroke
`in humans occurs
`in the
`context of
`ageing,
`co-morbidity (hypertension, diabetes mellitus, atrial
`fibrillation, pre-existing cerebrovascular disease) and
`concomitant medication use.8 Furthermore,
`factors
`such as gender, cerebral blood flow, body temperature
`and glycemic status may influence stroke mechanism
`and outcomes associated with therapy.6,9–11
`Glucagon-Like Peptide-1 (GLP-1) receptor agonists
`are gaining increasing momentum as possible neuro-
`protective agents in AIS. GLP-1 is an incretin hor-
`mone. Alongside its role in insulin secretion from the
`pancreas and glucagon suppression, it also crosses the
`blood-brain barrier (BBB) and promotes synaptic func-
`tion, enhances neurogenesis, reduces apoptosis and
`protects neurons from oxidative stress.12 GLP-1 is pro-
`duced in the brain and receptors are distributed
`system.13 GLP-1
`throughout
`the central nervous
`Receptor Agonists (GLP-1RAs), licensed for Type 2
`Diabetes Mellitus (T2DM) have already demonstrated
`pre-clinical neuroprotective efficacy in Alzheimer’s
`Disease and clinical trials in neurodegenerative condi-
`tions are ongoing.12,14
`The aim of this systematic scoping review is to
`report the pre-clinical basis of GLP-1RAs as neuropro-
`tective agents in AIS and their translation into clinical
`trials. In addition to describing the characteristics and
`quality of studies, the objectives are to specifically con-
`sider timing of administration, association with glyce-
`mic status, neuroprotective outcomes and application
`to clinical care.
`
`Materials and methods
`
`Eligibility criteria
`
`in vivo studies
`Pre-clinical: We included pre-clinical
`which administered naturally occurring GLP-1, a
`mimetic or analogue, before, during or after stroke
`induction. Normoglycemic, hyperglycemic and induced
`T2DM models were included.
`
`Studies were excluded if their only focus was hem-
`orrhagic stroke as this does not reflect the proposed
`mechanism for how GLP-1 is involved in ischemic
`tissue injury. Those studies which reported incidence
`of hemorrhagic transformation as a complication of
`AIS were
`included as
`these
`reflect post-stroke
`complications.
`Clinical: We included all prospective clinical trials
`which administered GLP-1RAs before, during or
`after stroke onset with outcome measures defined to
`identify neuroprotective efficacy by way of stroke
`volume reduction or improvement in post-stroke func-
`tion or mortality. We also included any potential fea-
`sibility or safety-based studies in this area.
`Our scoping searches identified that very few pro-
`spective clinical trials measuring stroke outcomes were
`available. Pragmatically, we therefore also included all
`retrospective database analyses of stroke incidence or
`composite cardiovascular outcomes in patients treated
`with GLP-1RAs. Furthermore, we included cardiovas-
`cular outcome trials (CVOTs) of GLP-1RAs to evalu-
`ate the incidence of stroke in this relatively higher risk
`cohort.
`Studies were excluded if their full-text was not avail-
`able or not published in English. Efforts were made to
`contact authors directly to obtain any missing articles
`or data.
`
`Database search strategy
`initial scoping searches,15 we accessed
`After several
`Web of Science on 19 March 2020 to search
`MEDLINE, Web of Science core collection, BIOSIS
`and SciELO from 1 January 2000. Keywords were
`EITHER ‘GLP(-)1, glucagon like peptide(-)1, exena-
`tide, liraglutide, lixisenatide, albiglutide, dulaglutide,
`semaglutide’ AND EITHER ‘stroke, CVA, cerebrovas-
`cular, h(a)emorrhage, small vessel disease’. Articles
`were cross-referenced and references were searched to
`identify further studies of interest.
`All articles/studies were screened independently in
`an unblinded, standardised manner by MM and HE
`by way of title and abstract to identify those suitable
`for full-text review. Queries and disagreements were
`resolved by discussion. Preferred Reporting Items for
`Systematic Reviews and Meta-Analyses (PRISMA)
`guidelines were applied. Pre-clinical
`studies were
`appraised according to Animal Research: Reporting
`of In Vivo Experiments (ARRIVE) guidelines,16 and
`the updated Stroke Therapy Academic
`Industry
`Roundtable Preclinical Recommendations (STAIR)
`guidelines.7 Data supporting the findings of
`this
`review are available from the corresponding author
`upon reasonable request.
`
`Novo Nordisk Exhibit 2104
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00002
`
`

`

`16
`
`Results
`
`Study selection
`
`The literature search identified 797 results (see Figure 1)
`alongside 10 from other sources. After removal of dupli-
`cates, this left 794 for screening. We excluded 593
`articles based upon title and review of abstract leaving
`201 full text articles to review. In total, 35 preclinical
`studies, 11 retrospective database studies, 7 cardiovascu-
`lar outcome trials and 4 prospective clinical studies met
`the inclusion criteria.
`
`Pre-clinical studies
`included studies. As shown in
`Characteristics of
`Table 1, 35 pre-clinical studies were included within
`this review. Studies were completed between 2009 and
`2020. Studies were predominantly based upon mouse
`and rat models of stroke; however, one study utilised a
`gerbil model.17 Stroke induction was either via tran-
`sient (range 30–120 min) or permanent common carot-
`id (CCAO) or middle
`cerebral artery occlusion
`(MCAO). Most studies induced unilateral occlusion
`in keeping with spontaneously occurring stroke onset
`in humans, but six studies utilised a bilateral occlusion
`model. Cerebral ischemia was induced by either liga-
`tion, filament occlusion or ablation of the relevant
`artery.
`
`Journal of Cerebral Blood Flow & Metabolism 41(1)
`
`Twelve studies administered exendin-4,17–28 nine
`used liraglutide,29–37 three used rhGLP-1 (recombinant
`human GLP-1),38–40 three used lixisenatide41–43 and
`one study each reported the utility of semaglutide,43
`poly-microspheres),44
`PEx-4
`(exendin-4
`loaded
`proGLP-1 (long acting GLP-1RA),45 DMB (GLP-1R
`agonist/modulator),46 dual GLP-1/Glucose-dependent
`Insulinotropic Peptide (GIP) agonist
`(GLP-1/GIP
`DA),47 oxyntomodulin (co-activates GLP-1R and glu-
`cagon receptor),48 P7C3 (aminopropyl carbazole com-
`pound)49 and one study directly compared exendin-4
`with liraglutide.50 In eight studies, GLP-1R antago-
`nists, such as Ex-9-39, were administered to study the
`role of
`the GLP-1R in neuroprotective mecha-
`nisms.19,21–23,41,45,46,49
`Two studies investigated multiple doses of GLP-
`1RAs to compare neuroprotective efficacy and con-
`cluded that neuroprotection was dose-dependent.20,51
`Most studies administered GLP-1RAs via intraper-
`itoneal, subcutaneous or transvenous routes. However,
`Zhang et al. reported neuroprotection with both oral
`DMB46 and intranasal exendin-4.22
`Some 14 studies administered GLP-1RAs chroni-
`cally prior to the onset of stroke. Clinically, this
`would represent those patients who receive GLP-
`1RAs as part of
`routine T2DM management
`and then go on to experience AIS.18,19,22,23,29,31,38,
`39,42,44–46,48,51 Chronic
`pre-treatment
`occurred
`
`Figure 1. PRISMA flow chart demonstrating the selection of studies.
`
`Novo Nordisk Exhibit 2104
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00003
`
`

`

`Maskery et al.
`
`17
`
`(continued)
`
`byshRNAsuggestingNPisGLP-1Rmediated.
`insulinandglucoselevelsandNPwasblocked
`tionthanintraperitonealNoimpactonplasma
`
`3),"EAAT2,
`"SOD),#apoptosis("Bcl-2,#Bax,#caspase-
`#oxidativestressparameters(#MDA,"GSH,
`NP–#infarctvolume,#neurologicaldeficit,
`a).LixisenatidemoreNPthanglimepiride.
`#inflammation/apoptosis(#caspase-3,#TNF-
`#oxidativestress(#MDA,"GSH,"catalase),
`
`NP–#infarctvolume,#neurologicaldeficit,
`NP–#infarctvolume,"locomotiveactivity
`inflammation(#COX-2,#PE2)
`#neurologicaldeficit,#oxidativedamage/
`
`NP–#infarctvolume(byupto75%),
`
`byEx-9-39.
`ferenttoEx-4.NPblockedbyshRNAbutnot
`diabeticmice.DMBactivationofGLP-1Rdif-
`onplasmainsulinandglucoselevelsinnon-
`
`Intranasalrouteproduced"CNSconcentra-
`#neurologicaldeficit,#apoptosis(#caspase-3)
`
`NPgivenintranasally–#infarctvolume,
`
`deficit,#apoptosis("Bcl-2,#Bax).Noimpact
`NPgivenorally–#infarctvolume,#neurological
`markers(#S100B,#NSE,#MBP)
`group),#neurologicaldeficit,#braininjury
`NPinDM–#infarctvolume(morethaninsulin
`
`GLP-1RmediatedNP)
`dependent.NPblockedbyshRNA(suggesting
`
`#apoptosis("Bcl-2,#Bax).NPnotglucose
`
`NP–#infarctvolume,#neurologicaldeficit,
`
`moreNPthanEx-4inDM.
`
`#oxidativestress,"cerebralbloodflow.PEx-4
`
`NP–#brainoedema,#cognitivedeficit,
`
`reduceinfarctvolume.
`volumereduction,insulintreatmentdidnot
`diabetic&DMmodelswithsimilarinfarct
`
`#apoptosis("Bcl-2,#Bax).NPinbothnon-
`stressparameters(#MDA,"GSH,"SOD),
`NP–#infarctvolume,#motordeficit,#oxidative
`
`endothelinreceptor.
`
`via#oxidativestressandindependentof
`stressparameters(#MDA,"GSH,"SOD)NP
`NP–#infarctvolume,#motordeficit,#oxidative
`NP–#infarctvolumeandimprovesfunctional
`
`outcome
`
`Mainoutcomes(NP–Neuroprotective)
`
`TDS–14days
`
`u.l.tMCAO(90)
`
`nimodipine
`
`OD–14days
`15
`
`b.l.tCCAO(30)
`u.l.tMCAO(60)
`
`glimepiride
`
`30
`
`u.l.MCAO(60)
`
`Ex-9-39
`
`OD–7days
`
`u.l.tMCAO(90)
`
`IntraperitonealEx-4,shRNA
`
`30
`
`u.l.tMCAO(60)
`
`Ex-4,Ex-9-39,shRNA
`
`TDS–14days
`
`u.l.tMCAO(90)
`
`nimodipine,insulin
`
`OD–7days
`
`u.l.tMCAO(90)
`
`&hypotension
`
`OD–14days
`
`b.ltCCAO(10)
`
`OD–14days
`
`u.l.pMCAO
`
`Non-diabetic,insulin
`
`BD–7days
`
`u.l.pMCAO
`
`15
`
`u.ltMCAO(60)
`
`ischemia
`Post-
`
`ischemia
`Pre-
`
`Administrationtime(min)
`
`(time/min)
`induction
`Stroke
`
`i.p.
`
`rhGLP-1
`
`DM
`
`2018Rat
`
`Fangetal.39
`
`i.p.
`i.c.v.
`
`i.c.v
`
`Lixisenatide
`OXM
`
`DM
`
`Abdel-latifetal.422018Rat
`Lietal.48
`2017Rat
`
`Ex-4
`
`2017Rat
`
`Kimetal.23
`
`i.n.
`
`Ex-4
`
`2016Mouse
`
`Zhangetal.22
`
`p.o.
`
`DMB
`
`2016Mouse
`
`Zhangetal.46
`
`i.p.
`
`rhGLP-1
`
`DM
`
`2016Rat
`
`Jiangetal.38
`
`shRNA
`
`i.p.
`
`Pro-GLP-1
`
`2015Mouse
`
`Zhangetal.45
`
`Non-diabetic,Ex-4
`
`s.c.
`
`PEx-4
`
`DM
`
`2015Rat
`
`Chienetal.44
`
`s.c.
`
`Liraglutide
`
`DM
`
`2014Rat
`
`Briyaletal.29
`
`BQ123
`
`i.p.
`
`Ex-4
`
`2012Rat
`
`Briyaletal.19
`
`Lietal.18
`GLP-1RAadministeredpriortostrokeonset(includingchronicpre-treatment)
`
`2009Rat
`
`i.c.v.
`
`Ex-4
`
`Sub-studies
`
`route
`Admin
`
`modelCo-morb.Drug
`Animal
`
`Year
`
`Author
`
`Table1.Overviewofthepre-clinicalstudies.
`
`Novo Nordisk Exhibit 2104
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00004
`
`

`

`18
`
`Journal of Cerebral Blood Flow & Metabolism 41(1)
`
`(continued)
`
`youngandoldanimalmodels
`tered4.5hrsafterstrokeonset.NPinboth
`administration.NotNPwhenEx-4adminis-
`kg.StrokevolumenotaffectedbyEx-4
`50mg/kgat90/180minandat90minfor5mg/
`inflammatorymarkers.NPinnon-DM/DMat
`Nonstatisticallysignificantreductioninpro-
`
`NP–"neuronalsurvival,"M2microglialmarkers
`#oxidativestress,"VEGFincortexbutnotin
`
`raisedintracellularcAMPlevels
`GLP-1Ractivation).NPprobablymediatedvia
`
`NP–#infarctvolume,#neurologicaldeficit,
`
`thestriatum.
`
`activation),"intracellularcAMPlevels(dueto
`#oxidativestress,#inflammation(#microglial
`
`NP–#infarctvolume,#neurologicaldeficit,
`
`NPbetweenDM&non-DM
`signallingpathway)Nosignificantdifferencein
`
`(#MPO),"Nrf2,"HO-1(antioxidativestress
`#oxidativestress("SOD),#inflammation
`NP-#infarctvolume,#neurologicaldeficit,
`
`dosedependent.
`proliferation&neuroblastformation.NPis
`
`NP–#infarctvolume,#neurologicaldeficit,
`
`andnaloxone
`blockedbyEx-9-39,b-endorphinanti-serum
`"hippocampalb-endorphinexpression,NP
`
`0
`
`u.l.tMCAO(120)
`
`nimodipine
`
`thenOD1week
`
`90,180or270
`
`14monthobese/DMmice
`
`u.l.tMCAO(30)
`
`60
`
`u.l.tMCAO(90)
`
`0,60,180
`
`u.ltMCAO(60)
`
`BD–7days
`
`BD–7days
`
`u.l.pMCAO
`
`Non-DM
`
`0
`
`15
`
`u.l.tMCAO(60)
`
`butmarginaleffectonactivation,"stemcell
`4weekspostMCAO),#microglialinfiltration
`NP–#infarctvolume(nodifferencebetween2&
`activity,#microglialactivation
`NP–#neurologicaldeficit,"GLP-1Rimmunore-
`
`BD–4weeksBD–2–4weeks
`
`u.l.tMCAO(90)
`
`60
`
`120
`
`b.l.tCCAO(5)
`
`cemiccontrolamelioration(MetforminnotNP)
`non-DMmodel,NPnotassociatedwithgly-
`
`NP–#oxidativestressparameters(#MDA,
`
`Lixisenatide"NP
`#IL-1b,#TNF-a),"viablehippocampalneu-
`#caspase-3),#inflammatorymarkers(#MPO,
`"GSH,"SOD),#apoptosis("Bcl-2,#Bax,
`
`ronsonhistologicalstaining.Higherdose
`
`NP–#infarctvolume,#neurologicaldeficitonlyin
`
`Mainoutcomes(NP–Neuroprotective)
`
`OD–14days
`
`OD–7days
`
`u.l.tMCAO(30)
`
`Non-diabetic,metformin
`
`ischemia
`Post-
`
`ischemia
`Pre-
`
`Administrationtime(min)
`
`(time/min)
`induction
`Stroke
`
`i.p.
`
`rhGLP-1
`
`DM
`
`2015Rat
`
`Zhaoetal.40
`
`2monthold&
`
`i.p.
`
`Ex-4
`
`2014MouseDM
`
`Darsaliaetal.25
`
`i.p.
`
`Liraglutide
`
`2013Rat
`
`Satoetal.32
`
`Teramotoetal.242011Mouse
`GLP-1RAadministeredfollowingstrokeonset(includingdelayedadministration)
`
`Ex-4
`
`t.v.
`
`i.p.
`
`Liraglutide
`
`DM
`
`2018Rat
`
`Dengetal.30
`
`Ex-9-39
`
`i.c.v.
`
`Ex-4/Catapol
`
`2015Rat
`
`Jiaetal.21
`
`0.1,2or5mg/kgofEx-4
`
`i.p.
`
`i.p.
`
`Ex-4
`
`DM
`
`2012Rat
`
`Darsaliaetal.20
`
`HyunLeeetal.172011Gerbil
`GLP-1RAadministeredpriortoandfollowingstrokeonset
`
`Ex-4
`
`0.7and7nmol/kglixisenatideb.l.tCCAO(60)
`
`i.p.
`
`Lixisenatide
`
`2020Rat
`
`Gadetal.51
`
`s.c.
`
`Liraglutide
`
`DM
`
`2018Rat
`
`Filchenkoetal.31
`
`Sub-studies
`
`route
`Admin
`
`modelCo-morb.Drug
`Animal
`
`Year
`
`Author
`
`Table1.Continued.
`
`Novo Nordisk Exhibit 2104
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00005
`
`

`

`19
`
`(continued)
`
`ingGLP-1Rindependentpathway.
`inhibitedbyGLP-1RantagonistEx-39suggest-
`
`#inflammation(TNF-a)–theseeffectsnot
`(#GSH,#MDA,#catalase,#NO),
`#apoptosis(caspase-3),#oxidativestress
`NP–#infarctvolume,#neurologicaldeficit,
`
`delayedby1day
`
`("NeuN,"GFAP,"vWF).NPwhenfirstdose
`"neurovascularremodelling
`"glucosemetabolisminischemicpenumbra
`NP–#neurologicaldeficit,"GLP-1Rexpression,
`#stressrelatedhyperglycemiawithoutcausing
`"Bcl-2,#Bax),#DNAdamage(#TUNEL),
`#apoptosis(#ROS,#caspase-3,-8,-9,#PARP,
`
`(18-FDG-PET)
`
`hypoglycemia.
`
`#inflammation(#ICAM-1,NF-2jB,p50,p65).
`#TUNEL,#Bax,#PARP,"Bcl-2),
`#ROS,"SOD),#apoptosis(#caspase-3,
`#cerebraloedema,#oxidativestress(#DHE,
`dysfunction,"cerebralmicrocirculation.
`
`notNP.
`microglia,NPinhyperglycemicmodel.Insulin
`
`NP–#neurologicaldeficitincluding#bladder
`
`NP–#infarctvolume,#neurologicaldeficit,
`NP–#DNAdamage("APE1,#TUNEL)
`
`1&24hrs
`
`b.l.tCCAO(30)
`
`Ex-9-39,pentoxyphylline
`
`i.p.
`
`Lixisenatide
`
`Abdel-latifetal.412018Rat
`
`for4weeks
`24hrs,thenOD
`
`1,3and7days
`1hr,thenODfor
`
`0&24hrs
`
`u.l.MCAO(90)
`
`s.c.
`
`Liraglutide
`
`2017Rat
`
`Dongetal.34
`
`u.lpMCAO
`
`b.l.tCCAO(60)
`
`u.l.tMCAO&
`
`s.c.
`
`t.v.
`
`Liraglutide
`
`2016Rat
`
`Zhuetal.33
`
`Ex-4
`
`2016Rat
`
`Yangetal.28
`
`0,3,6or12hrs
`
`u.l.tMCAO(60)
`
`Ex-4orliraglutidei.p.
`
`2016MouseDM
`
`Lietal.50
`
`Maskery et al.
`
`(#TNF-a),#MMP-9activation,#Iba-1positive
`oedema,#oxidativestress,#inflammation
`tion.Ex-4NP–#infarctvolume,#cerebral
`"cerebraloedema,"hemorrhagictransforma-
`Hyperglycemiaassociatedwith"infarctvolume,
`
`hemorrhagictransformation.
`inhibitor.Amelioratedwarfarinassociated
`
`HHE),#inflammation,NPblockedbyP13K
`integrity,#oxidativestress(#8-OHdG,#
`NP–#infarctvolume,#neurologicaldeficit,"BBB
`
`damage(iNOS).DAmoreNPthanSA.
`
`coseinhighdosegroup(nohypoglycemia)
`Statisticallysignificantreductioninbloodglu-
`
`#apoptosis("Bcl-2,#Bax),#nitricoxide
`NP–#infarctvolume,#neurologicaldeficit,
`
`#nitricoxidedamage(iNOS,eNOS)
`#oxidativestress(#MDA,"GSH,"SOD),
`
`NP–#infarctvolume,"neuronalsurvival,
`
`Mainoutcomes(NP–Neuroprotective)
`
`60
`
`0
`
`60
`
`u.l.tMCAO(60)
`
`insulin
`
`i.p.
`
`Ex-4
`
`2016MouseDM
`
`Kurokietal.27
`
`u.l.tMCAO(45)
`
`P13Kinhibitor
`
`t.v.
`
`Warfarin&Ex-4
`
`2016Mouse
`
`Chenetal.26
`
`u.l.tMCAO(60)
`
`GLP-1RA(SA)
`
`i.p.
`
`GLP-1/GIP(DA)
`
`2016Rat
`
`Hanetal.47
`
`ischemia
`Post-
`
`ischemia
`Pre-
`
`Administrationtime(min)
`
`(time/min)
`induction
`Stroke
`
`Sub-studies
`
`route
`Admin
`
`modelCo-morb.Drug
`Animal
`
`Year
`
`Author
`
`Table1.Continued.
`
`Novo Nordisk Exhibit 2104
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00006
`
`

`

`20
`
`Journal of Cerebral Blood Flow & Metabolism 41(1)
`
`fibrillaryacidicprotein(glialmarker);vWF:vonWillebrandfactor(endothelialmarker);b-tub3:betatubulinIII;GSK-3:glycogensynthasekinase3;IL:interleukin.
`NOS:nitricoxidesynthase;BBB:bloodbrainbarrier;MMP-9:matrixmetalloproteinase-9;ICAM-1:intracellularadhesionmolecule-1;NeuN:neuronalnuclearprotein(neuronalmarker);GFAP:Glial
`myelinbasicprotein;COX-2:cyclo-oxygenase-2;PE2:ProstaglandinE2;EAAT2:Excitatoryaminoacidtransporter-2;MPO:myeloperoxidase;HO-1:hemeoxygenase-1;Nrf2:nuclearfactorerythroid-2;
`protein4;Caspase-3;PARP:poly-ADPribosepolymerase;TUNEL:terminaldeoxynucleotidyltransferasedUTPnickendlabeling;S100b:s100-calciumbindingproteinB;NSE:neuronspecificenolase;MBP:
`MDA:malondialdehyde;GSH:glutathione;8-OHdG:8-hydroxy-2-deoxyguanosine;HHE:4-hydroxyhexenal;DHE:dihydroethidium;ROS:reactiveoxygenspecies;Bcl-2:Bcelllymphoma2;Bax:Bcl-2-like
`diabetesmellitus(healthy);u.l.:unilateral;b.l.:bilateral;MCAO:middlecerebralarteryocclusion;CCAO:commoncarotidarteryocclusion;(t/p)MCAO:transient/permanent;SOD:superoxidedismutase;
`compound;i.p.:intraperitoneal;s.c.:subcutaneous;p.o.:oral;i.n.:intranasal;t.v.:transvenous;i.c.v.:intracerebroventricular;OD/BD/TDS:once/twice/threetimesdaily;DM:diabetesmellitus;Non-DM:non
`6,7-dichloro-2-methyl-sulfonyl-3-N-tert-butylaminoquinoxaline;OXM:oxyntomodulin;shRNA:smallhairpinRNA(targetsGLP-1R);BQ123-endothelinreceptorantagonist;P7C3-aminopropylcarbazole
`SA:singleagonist;DA:dualagonist;Ex-4-exendin-4;PEx-4:Exendin-4loadedpoly-microspheres;Ex-9-39:GLP-1Rantagonist;pro-GLP-1:longactingGLP-1RA;rhGLP-1:recombinanthumanGLP-1;DMB:
`
`effects.
`COX-2)P13kinhibitorpartiallyreversedNP
`
`#inflammation(#TNF-a,IL-18,IL-1b,IL-6,
`#apoptosis("Bcl-2,#caspase-3,#TUNEL),
`
`NP–#infarctvolume,#cerebraloedema,
`NP–#neurologicaldeficit,#oxidativestress
`
`glycemiainnormoglycemicmodel.
`
`factorsignalling("ERK1,"ERS-1),Nohypo-
`#caspase-3),"neurogenesis,improvedgrowth
`#neuronalloss,#apoptosis("Bcl-2,#Bax,
`
`NP–#infarctvolume,#neurologicaldeficit,
`
`dependent.
`
`manner).NPwhenfirstdosedelayedby1day
`releaseisinablood-glucosedependent
`andcontrols(asexpected,stimulatedinsulin
`enceinbloodglucoselevelsbetweenliraglutide
`
`"adamts20,#GSK-3)NPisGLP-1R
`("doublecortin,"b-tub3,"adam11,
`NP–#neurologicaldeficit,"neurogenesis
`
`0thenOD
`ODfor7days
`
`u.lpMCAO
`u.l.pCCAO
`
`P13kinhibitor
`
`i.p.
`i.p.
`
`Liraglutide
`Liraglutide
`
`2020Rat
`2019Mice
`
`Zengetal.37
`Heetal.36
`
`days1,7,14or21days
`
`2hrs,thenalternate
`
`u.l.pMCAO
`
`Bloodglucosemonitoring
`
`i.p.
`
`Semaglutide
`
`2019Rat
`
`Yangetal.43
`
`120
`
`u.l.tMCAO(40)
`
`Ex-9-39
`
`t.v.
`
`P7C3
`
`2018Mouse
`
`Wangetal.49
`
`"angiogenesis("VEGF)Nosignificantdiffer-
`
`24hrs,thenODfor14daysNP–#infarctvolume,#neurologicaldeficit,
`
`Mainoutcomes(NP–Neuroprotective)
`
`ischemia
`Post-
`
`ischemia
`Pre-
`
`Administrationtime(min)
`
`u.l.pMCAO
`
`(time/min)
`induction
`Stroke
`
`i.p.
`
`Liraglutide
`
`2018Mouse
`
`Chenetal.35
`
`Sub-studies
`
`route
`Admin
`
`modelCo-morb.Drug
`Animal
`
`Year
`
`Author
`
`Table1.Continued.
`
`Novo Nordisk Exhibit 2104
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00007
`
`

`

`Maskery et al.
`
`21
`
`between 14 days prior and 15 minutes prior to stroke
`onset, for one to three times daily.
`A further four studies administered GLP-1RAs
`prior to and following stroke onset for between 0 and
`four weeks – with dosing schedules of up to twice
`daily.17,20,21,30
`Most closely aligned with the proposed clinical
`application of administering treatment to GLP-1RA
`naı¨ ve patients in the hyper-acute AIS setting, 17 studies
`only administered the medication following stroke
`onset.24–28,32–37,40,41,43,47,49,50 First dose was delayed
`between 0 min and 24 h post-onset and continued for
`up to four weeks.
`Alterations in physiological parameters such as
`body temperature have the potential to impact the out-
`come of stroke. Li et al. reported no significant change
`in body temperature when measured before and after
`treatment with exendin-4.18 We did not identify any
`GLP-1RA study of AIS which varied temperature
`between groups. Most studies regulated body temper-
`ature within normal physiological parameters during
`MCAO surgery and animals were housed within a
`temperature-controlled environment.
`
`Quality of included studies. Pre-clinical study meth-
`odology was appraised according to the STAIR 2009
`criteria, with a maximum available score of 7. Median
`score was 3 (range 2–7). Inclusion/exclusion criteria
`and reporting of potential conflicts of interest/funding
`were consistently reported in 100% and 94% of studies,
`respectively. Fewer studies commented on randomisa-
`tion (54%), allocation concealment (17%) and blinded
`assessment of outcome (43%). Only four studies (11%)
`reported performing a sample size calculation.
`Reporting of pre-clinical studies was also assessed
`using the ARRIVE guidelines, with a maximum score
`of 36. Median score was 22 (range 14–29). Methods,
`statistical analysis, outcomes and confidence intervals
`were well reported, as were ethical and funding state-
`ments, but the justification for animal models, transla-
`tion to human biology, limitations and adverse events
`were frequently not reported.
`
`Impact of hyperglycemia in stroke models. Twelve
`studies were based on hyperglycemic rodents, with or
`without a normoglycemic control group.
`Kuroki et al. demonstrated that hyperglycemia was
`associated with an increase in infarct volume, cerebral
`edema and hemorrhagic transformation in a mouse
`model that underwent transient, unilateral occlusion
`of the MCAO for 60 min.27 They reported that intra-
`peritoneal administration of exendin-4 60 min after
`stroke onset was associated with a reduction in these
`parameters which was not
`replicated by insulin
`monotherapy.
`
`Briyal et al. reported that liraglutide reduced infarct
`volume by a similar amount in both diabetic and nor-
`moglyemic models, but this neuroprotection was again
`not reproduced in the insulin treatment arm despite
`resolution of hyperglycemia.29 Deng et al. further con-
`firmed neuroprotection was independent of glycemic
`status prior to stroke onset.30 Jiang et al. reported
`that stroke infarct volume in the rhGLP-1 group was
`significantly reduced when compared to insulin treat-
`ment.38 Metformin was also shown to ameliorate
`hyperglycemia but did not confer the additional neuro-
`protective outcomes associated with liraglutide.31
`One study demonstrated similar neuroprotective
`outcomes between 2-month-old healthy mice and 14-
`month-old overweight, diabetic mice treated with exen-
`din-4.25
`Whilst GLP-1RAs were associated with a reduction
`in blood glucose in hyperglycemic models,40 no study
`reported hypoglycemia when GLP-1RAs were admin-
`istered to normoglycemic models. This is to be expected
`as GLP-1RA-associated insulin secretion from the pan-
`creas is glucose dependent.35 Indeed, Zhang et al. con-
`cluded
`that
`neuroprotection was
`not
`glucose
`dependent.45
`Yang et al. monitored blood glucose in rats which
`underwent intraperitoneal administration of semaglu-
`tide starting 2 h following unilateral permanent MCAO
`occlusion, further demonstrating neuroprotection in
`the absence of hypoglycemic episodes.43
`
`Infarct volume and neuronal survival. Administration
`of GLP-1RAs prior to, at the point of, or delayed fol-
`lowing stroke onset were associated with reduction in
`infarct volume. In total, nine studies demonstrated a
`reduction in infarct volume with exendin-4,18–24,26,27
`eight with liraglutide,29–33,35–37 three with rhGLP-1,
`three with lixisenatide and one with each of PEx-4,
`proGLP-1, DMB, OXM, GLP-1/GIP DA and sema-
`glutide. Liraglutide was associated with a reduction in
`infarct volume when the first dose was delayed by up to
`1 day.35 Kim et al. reported a reduction in infarct
`volume by up to 75% in a rat model of transient
`MCAO. Darsalia et al. reported that exendin-4 admin-
`istration was associated with a reduction in stroke
`volume when administered for four weeks prior to,
`following stroke onset.20
`and two to four weeks
`However, in a later study, whereby they only adminis-
`tered exendin-4 following stroke onset, it did not sig-
`nificantly reduce infarct volume but did reduce overall
`neuronal loss.25 Reduction in neuronal loss was also
`rhGLP-140
`semaglutide,43
`and
`reported with
`lixisenatide.51
`
`Cellular function. Apoptosis represents a chain of
`enzymatic
`events
`resulting
`in programmed cell
`
`Novo Nordisk Exhibit 2104
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00008
`
`

`

`22
`
`Journal of Cerebral Blood Flow & Metabolism 41(1)
`
`death.52 Whilst controlled apoptosis is essential for the
`maintenance of homeostasis, dysregulated apoptosis,
`for example in within the ischemic penumbra, can
`in increased cell death.53 Bcl-2 is an anti-
`result
`apoptotic protein which functions at least in part by
`reducing cytochrome C release from the mitochondria.
`Conversely, Bax is a pro-apoptotic protein and
`increases cytochrome C levels.52 Increased Bcl-2 and
`reduced Bax levels, contributing to an increased
`Bcl-2:Bax ratio and representing reduced levels of apo-
`ptosis, have been reported in three studies of liraglu-
`tide29,33,37 and one study for each pro-GLP-1,45
`rhGLP-1,39 DMB,22 exendin-4,50 lixisenatide,51 GLP-
`1/GIP DA47 and semaglutide.43 Critically, this reduc-
`tion of apoptosis was reproduced in normoglycemic
`models when administered after stroke onset.33,37,43,47
`Caspase proteins are also pro-apoptotic and
`increased levels are associated with higher rates of
`cell death.54 Liraglutide,
`exendin-4,
`lixisenatide,
`rhGLP-1 and semaglutide have been shown to reduce
`levels
`of
`caspase-3
`in
`pre-clinical models
`of
`stroke.33,37,39,41–43,46,50,51 Zhu et al. have also demon-
`strated reduced levels of caspase 8 and 9 in models
`administered liraglutide.33
`Cleavage of PARP by caspases is a signal of apo-
`ptosis and has been implicated in cerebral ischemia.53
`Reduction in PARP has been demonstrated with both
`liraglutide and exendin-4 treatment.33,50
`TUNEL assays detect DNA degradation during the
`later
`stages of apoptosis and have demonstrated
`reduced apoptotic activity with liraglutide and exen-
`din-4.37,50,55
`GLP-1RA administration following stroke induc-
`tion has been associated with an anti-inflammatory
`effect.26 Tumor necrosis factor alpha (TNF-a) is a cyto-
`kine synthesised by many cell lines, but particularly by
`macrophages and microglia.56 TNF-a is involved with
`the inflammatory response following stroke onset.56
`Five studies reported a reduction in TNF-a when com-
`pared to controls, three with lixisenatide41,42,51 and one
`with each of liraglutide37 and exendin-4.27 Indeed, two
`reported reduced microglial activation17,24
`studies
`whilst a further study by Darsalia et al. reported
`reduced microglial infiltration, but a marginal effect
`on activation.20 Reduced levels of other markers of
`inflammation, such as myeloperoxidase and interleu-
`kin, have also been reported.30,37,51
`One study reported a non-statistically significant
`reduction in pro-inflammatory markers.25
`There is also deterioration in markers of oxidative
`stress following AIS. Twelve studies reported an
`improvement in oxidative stress parameters following
`GLP-1RA administration in AIS – five studies of exen-
`din-4,19,24,26,27,50 four studies of liraglutide,29,30,32,36
`lixisenatide41,42
`rhGLP.40
`two of
`and one of
`
`reported reduced levels of
`Predominantly, studies
`malondialdehyde and increased concentrations of glu-
`tathione and superoxide dismutase.
`Dong et al. performed 18-FDG PET imaging in a
`rat model of unilateral transient MCAO. In the ani-
`mals treated with subcutaneous liraglutide, there was
`radiological evidence of increased glucose metabolism
`within the ischemic penumbra.34
`
`function. The neurovascular unit
`Neurovascular
`incorporates both cellular and extracellular compo-
`nents involved in the regulation of cerebral blood
`flow and blood-brain barrier function – it is involved
`with the maintenance of cerebral homeostasis and con-
`trol of cerebral blood flow.57
`GLP-1RAs have been shown to increase cerebral
`blood flow following AIS when compared with con-
`trols.44 Li et al. reported that cerebral microcirculation
`is reduced following AIS, but improved to a similar
`degree within 4–12 h after MCAO in diabetic mice
`treated with either liraglutide or exendin-4 after stroke
`induction.50 Blood–brain barrier integrity has also been
`shown to improve with GLP-1RA treatment.26
`is
`Vascular endothelial growth factor
`(VEGF)
`known to promote angiogenesis and protect ischemic
`neurons from injury, demonstrating a crucial role in the
`neurovascular remodelling post-AIS.58 Chen et al.
`demonstrated that intraperitoneal liraglutide therapy,
`administere

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