CCJM delivers practical clinical articles relevant to internists, cardiologists, endocrinologists, and other specialists, all written by known experts.

Theme
medstat_ccjm
Top Sections
CME
Reviews
1-Minute Consult
The Clinical Picture
Smart Testing
Symptoms to Diagnosis
ccjm
Main menu
CCJM Main Menu
Explore menu
CCJM Explore Menu
Proclivity ID
18804001
Unpublish
Negative Keywords
gaming
gambling
compulsive behaviors
ammunition
assault rifle
black jack
Boko Haram
bondage
child abuse
cocaine
Daech
drug paraphernalia
explosion
gun
human trafficking
ISIL
ISIS
Islamic caliphate
Islamic state
mixed martial arts
MMA
molestation
national rifle association
NRA
nsfw
pedophile
pedophilia
poker
porn
pornography
psychedelic drug
recreational drug
sex slave rings
slot machine
terrorism
terrorist
Texas hold 'em
UFC
substance abuse
abuseed
abuseer
abusees
abuseing
abusely
abuses
aeolus
aeolused
aeoluser
aeoluses
aeolusing
aeolusly
aeoluss
ahole
aholeed
aholeer
aholees
aholeing
aholely
aholes
alcohol
alcoholed
alcoholer
alcoholes
alcoholing
alcoholly
alcohols
allman
allmaned
allmaner
allmanes
allmaning
allmanly
allmans
alted
altes
alting
altly
alts
analed
analer
anales
analing
anally
analprobe
analprobeed
analprobeer
analprobees
analprobeing
analprobely
analprobes
anals
anilingus
anilingused
anilinguser
anilinguses
anilingusing
anilingusly
anilinguss
anus
anused
anuser
anuses
anusing
anusly
anuss
areola
areolaed
areolaer
areolaes
areolaing
areolaly
areolas
areole
areoleed
areoleer
areolees
areoleing
areolely
areoles
arian
arianed
arianer
arianes
arianing
arianly
arians
aryan
aryaned
aryaner
aryanes
aryaning
aryanly
aryans
asiaed
asiaer
asiaes
asiaing
asialy
asias
ass
ass hole
ass lick
ass licked
ass licker
ass lickes
ass licking
ass lickly
ass licks
assbang
assbanged
assbangeded
assbangeder
assbangedes
assbangeding
assbangedly
assbangeds
assbanger
assbanges
assbanging
assbangly
assbangs
assbangsed
assbangser
assbangses
assbangsing
assbangsly
assbangss
assed
asser
asses
assesed
asseser
asseses
assesing
assesly
assess
assfuck
assfucked
assfucker
assfuckered
assfuckerer
assfuckeres
assfuckering
assfuckerly
assfuckers
assfuckes
assfucking
assfuckly
assfucks
asshat
asshated
asshater
asshates
asshating
asshatly
asshats
assholeed
assholeer
assholees
assholeing
assholely
assholes
assholesed
assholeser
assholeses
assholesing
assholesly
assholess
assing
assly
assmaster
assmastered
assmasterer
assmasteres
assmastering
assmasterly
assmasters
assmunch
assmunched
assmuncher
assmunches
assmunching
assmunchly
assmunchs
asss
asswipe
asswipeed
asswipeer
asswipees
asswipeing
asswipely
asswipes
asswipesed
asswipeser
asswipeses
asswipesing
asswipesly
asswipess
azz
azzed
azzer
azzes
azzing
azzly
azzs
babeed
babeer
babees
babeing
babely
babes
babesed
babeser
babeses
babesing
babesly
babess
ballsac
ballsaced
ballsacer
ballsaces
ballsacing
ballsack
ballsacked
ballsacker
ballsackes
ballsacking
ballsackly
ballsacks
ballsacly
ballsacs
ballsed
ballser
ballses
ballsing
ballsly
ballss
barf
barfed
barfer
barfes
barfing
barfly
barfs
bastard
bastarded
bastarder
bastardes
bastarding
bastardly
bastards
bastardsed
bastardser
bastardses
bastardsing
bastardsly
bastardss
bawdy
bawdyed
bawdyer
bawdyes
bawdying
bawdyly
bawdys
beaner
beanered
beanerer
beaneres
beanering
beanerly
beaners
beardedclam
beardedclamed
beardedclamer
beardedclames
beardedclaming
beardedclamly
beardedclams
beastiality
beastialityed
beastialityer
beastialityes
beastialitying
beastialityly
beastialitys
beatch
beatched
beatcher
beatches
beatching
beatchly
beatchs
beater
beatered
beaterer
beateres
beatering
beaterly
beaters
beered
beerer
beeres
beering
beerly
beeyotch
beeyotched
beeyotcher
beeyotches
beeyotching
beeyotchly
beeyotchs
beotch
beotched
beotcher
beotches
beotching
beotchly
beotchs
biatch
biatched
biatcher
biatches
biatching
biatchly
biatchs
big tits
big titsed
big titser
big titses
big titsing
big titsly
big titss
bigtits
bigtitsed
bigtitser
bigtitses
bigtitsing
bigtitsly
bigtitss
bimbo
bimboed
bimboer
bimboes
bimboing
bimboly
bimbos
bisexualed
bisexualer
bisexuales
bisexualing
bisexually
bisexuals
bitch
bitched
bitcheded
bitcheder
bitchedes
bitcheding
bitchedly
bitcheds
bitcher
bitches
bitchesed
bitcheser
bitcheses
bitchesing
bitchesly
bitchess
bitching
bitchly
bitchs
bitchy
bitchyed
bitchyer
bitchyes
bitchying
bitchyly
bitchys
bleached
bleacher
bleaches
bleaching
bleachly
bleachs
blow job
blow jobed
blow jober
blow jobes
blow jobing
blow jobly
blow jobs
blowed
blower
blowes
blowing
blowjob
blowjobed
blowjober
blowjobes
blowjobing
blowjobly
blowjobs
blowjobsed
blowjobser
blowjobses
blowjobsing
blowjobsly
blowjobss
blowly
blows
boink
boinked
boinker
boinkes
boinking
boinkly
boinks
bollock
bollocked
bollocker
bollockes
bollocking
bollockly
bollocks
bollocksed
bollockser
bollockses
bollocksing
bollocksly
bollockss
bollok
bolloked
bolloker
bollokes
bolloking
bollokly
bolloks
boner
bonered
bonerer
boneres
bonering
bonerly
boners
bonersed
bonerser
bonerses
bonersing
bonersly
bonerss
bong
bonged
bonger
bonges
bonging
bongly
bongs
boob
boobed
boober
boobes
boobies
boobiesed
boobieser
boobieses
boobiesing
boobiesly
boobiess
boobing
boobly
boobs
boobsed
boobser
boobses
boobsing
boobsly
boobss
booby
boobyed
boobyer
boobyes
boobying
boobyly
boobys
booger
boogered
boogerer
boogeres
boogering
boogerly
boogers
bookie
bookieed
bookieer
bookiees
bookieing
bookiely
bookies
bootee
booteeed
booteeer
booteees
booteeing
booteely
bootees
bootie
bootieed
bootieer
bootiees
bootieing
bootiely
booties
booty
bootyed
bootyer
bootyes
bootying
bootyly
bootys
boozeed
boozeer
boozees
boozeing
boozely
boozer
boozered
boozerer
boozeres
boozering
boozerly
boozers
boozes
boozy
boozyed
boozyer
boozyes
boozying
boozyly
boozys
bosomed
bosomer
bosomes
bosoming
bosomly
bosoms
bosomy
bosomyed
bosomyer
bosomyes
bosomying
bosomyly
bosomys
bugger
buggered
buggerer
buggeres
buggering
buggerly
buggers
bukkake
bukkakeed
bukkakeer
bukkakees
bukkakeing
bukkakely
bukkakes
bull shit
bull shited
bull shiter
bull shites
bull shiting
bull shitly
bull shits
bullshit
bullshited
bullshiter
bullshites
bullshiting
bullshitly
bullshits
bullshitsed
bullshitser
bullshitses
bullshitsing
bullshitsly
bullshitss
bullshitted
bullshitteded
bullshitteder
bullshittedes
bullshitteding
bullshittedly
bullshitteds
bullturds
bullturdsed
bullturdser
bullturdses
bullturdsing
bullturdsly
bullturdss
bung
bunged
bunger
bunges
bunging
bungly
bungs
busty
bustyed
bustyer
bustyes
bustying
bustyly
bustys
butt
butt fuck
butt fucked
butt fucker
butt fuckes
butt fucking
butt fuckly
butt fucks
butted
buttes
buttfuck
buttfucked
buttfucker
buttfuckered
buttfuckerer
buttfuckeres
buttfuckering
buttfuckerly
buttfuckers
buttfuckes
buttfucking
buttfuckly
buttfucks
butting
buttly
buttplug
buttpluged
buttpluger
buttpluges
buttpluging
buttplugly
buttplugs
butts
caca
cacaed
cacaer
cacaes
cacaing
cacaly
cacas
cahone
cahoneed
cahoneer
cahonees
cahoneing
cahonely
cahones
cameltoe
cameltoeed
cameltoeer
cameltoees
cameltoeing
cameltoely
cameltoes
carpetmuncher
carpetmunchered
carpetmuncherer
carpetmuncheres
carpetmunchering
carpetmuncherly
carpetmunchers
cawk
cawked
cawker
cawkes
cawking
cawkly
cawks
chinc
chinced
chincer
chinces
chincing
chincly
chincs
chincsed
chincser
chincses
chincsing
chincsly
chincss
chink
chinked
chinker
chinkes
chinking
chinkly
chinks
chode
chodeed
chodeer
chodees
chodeing
chodely
chodes
chodesed
chodeser
chodeses
chodesing
chodesly
chodess
clit
clited
cliter
clites
cliting
clitly
clitoris
clitorised
clitoriser
clitorises
clitorising
clitorisly
clitoriss
clitorus
clitorused
clitoruser
clitoruses
clitorusing
clitorusly
clitoruss
clits
clitsed
clitser
clitses
clitsing
clitsly
clitss
clitty
clittyed
clittyer
clittyes
clittying
clittyly
clittys
cocain
cocaine
cocained
cocaineed
cocaineer
cocainees
cocaineing
cocainely
cocainer
cocaines
cocaining
cocainly
cocains
cock
cock sucker
cock suckered
cock suckerer
cock suckeres
cock suckering
cock suckerly
cock suckers
cockblock
cockblocked
cockblocker
cockblockes
cockblocking
cockblockly
cockblocks
cocked
cocker
cockes
cockholster
cockholstered
cockholsterer
cockholsteres
cockholstering
cockholsterly
cockholsters
cocking
cockknocker
cockknockered
cockknockerer
cockknockeres
cockknockering
cockknockerly
cockknockers
cockly
cocks
cocksed
cockser
cockses
cocksing
cocksly
cocksmoker
cocksmokered
cocksmokerer
cocksmokeres
cocksmokering
cocksmokerly
cocksmokers
cockss
cocksucker
cocksuckered
cocksuckerer
cocksuckeres
cocksuckering
cocksuckerly
cocksuckers
coital
coitaled
coitaler
coitales
coitaling
coitally
coitals
commie
commieed
commieer
commiees
commieing
commiely
commies
condomed
condomer
condomes
condoming
condomly
condoms
coon
cooned
cooner
coones
cooning
coonly
coons
coonsed
coonser
coonses
coonsing
coonsly
coonss
corksucker
corksuckered
corksuckerer
corksuckeres
corksuckering
corksuckerly
corksuckers
cracked
crackwhore
crackwhoreed
crackwhoreer
crackwhorees
crackwhoreing
crackwhorely
crackwhores
crap
craped
craper
crapes
craping
craply
crappy
crappyed
crappyer
crappyes
crappying
crappyly
crappys
cum
cumed
cumer
cumes
cuming
cumly
cummin
cummined
cumminer
cummines
cumming
cumminged
cumminger
cumminges
cumminging
cummingly
cummings
cummining
cumminly
cummins
cums
cumshot
cumshoted
cumshoter
cumshotes
cumshoting
cumshotly
cumshots
cumshotsed
cumshotser
cumshotses
cumshotsing
cumshotsly
cumshotss
cumslut
cumsluted
cumsluter
cumslutes
cumsluting
cumslutly
cumsluts
cumstain
cumstained
cumstainer
cumstaines
cumstaining
cumstainly
cumstains
cunilingus
cunilingused
cunilinguser
cunilinguses
cunilingusing
cunilingusly
cunilinguss
cunnilingus
cunnilingused
cunnilinguser
cunnilinguses
cunnilingusing
cunnilingusly
cunnilinguss
cunny
cunnyed
cunnyer
cunnyes
cunnying
cunnyly
cunnys
cunt
cunted
cunter
cuntes
cuntface
cuntfaceed
cuntfaceer
cuntfacees
cuntfaceing
cuntfacely
cuntfaces
cunthunter
cunthuntered
cunthunterer
cunthunteres
cunthuntering
cunthunterly
cunthunters
cunting
cuntlick
cuntlicked
cuntlicker
cuntlickered
cuntlickerer
cuntlickeres
cuntlickering
cuntlickerly
cuntlickers
cuntlickes
cuntlicking
cuntlickly
cuntlicks
cuntly
cunts
cuntsed
cuntser
cuntses
cuntsing
cuntsly
cuntss
dago
dagoed
dagoer
dagoes
dagoing
dagoly
dagos
dagosed
dagoser
dagoses
dagosing
dagosly
dagoss
dammit
dammited
dammiter
dammites
dammiting
dammitly
dammits
damn
damned
damneded
damneder
damnedes
damneding
damnedly
damneds
damner
damnes
damning
damnit
damnited
damniter
damnites
damniting
damnitly
damnits
damnly
damns
dick
dickbag
dickbaged
dickbager
dickbages
dickbaging
dickbagly
dickbags
dickdipper
dickdippered
dickdipperer
dickdipperes
dickdippering
dickdipperly
dickdippers
dicked
dicker
dickes
dickface
dickfaceed
dickfaceer
dickfacees
dickfaceing
dickfacely
dickfaces
dickflipper
dickflippered
dickflipperer
dickflipperes
dickflippering
dickflipperly
dickflippers
dickhead
dickheaded
dickheader
dickheades
dickheading
dickheadly
dickheads
dickheadsed
dickheadser
dickheadses
dickheadsing
dickheadsly
dickheadss
dicking
dickish
dickished
dickisher
dickishes
dickishing
dickishly
dickishs
dickly
dickripper
dickrippered
dickripperer
dickripperes
dickrippering
dickripperly
dickrippers
dicks
dicksipper
dicksippered
dicksipperer
dicksipperes
dicksippering
dicksipperly
dicksippers
dickweed
dickweeded
dickweeder
dickweedes
dickweeding
dickweedly
dickweeds
dickwhipper
dickwhippered
dickwhipperer
dickwhipperes
dickwhippering
dickwhipperly
dickwhippers
dickzipper
dickzippered
dickzipperer
dickzipperes
dickzippering
dickzipperly
dickzippers
diddle
diddleed
diddleer
diddlees
diddleing
diddlely
diddles
dike
dikeed
dikeer
dikees
dikeing
dikely
dikes
dildo
dildoed
dildoer
dildoes
dildoing
dildoly
dildos
dildosed
dildoser
dildoses
dildosing
dildosly
dildoss
diligaf
diligafed
diligafer
diligafes
diligafing
diligafly
diligafs
dillweed
dillweeded
dillweeder
dillweedes
dillweeding
dillweedly
dillweeds
dimwit
dimwited
dimwiter
dimwites
dimwiting
dimwitly
dimwits
dingle
dingleed
dingleer
dinglees
dingleing
dinglely
dingles
dipship
dipshiped
dipshiper
dipshipes
dipshiping
dipshiply
dipships
dizzyed
dizzyer
dizzyes
dizzying
dizzyly
dizzys
doggiestyleed
doggiestyleer
doggiestylees
doggiestyleing
doggiestylely
doggiestyles
doggystyleed
doggystyleer
doggystylees
doggystyleing
doggystylely
doggystyles
dong
donged
donger
donges
donging
dongly
dongs
doofus
doofused
doofuser
doofuses
doofusing
doofusly
doofuss
doosh
dooshed
doosher
dooshes
dooshing
dooshly
dooshs
dopeyed
dopeyer
dopeyes
dopeying
dopeyly
dopeys
douchebag
douchebaged
douchebager
douchebages
douchebaging
douchebagly
douchebags
douchebagsed
douchebagser
douchebagses
douchebagsing
douchebagsly
douchebagss
doucheed
doucheer
douchees
doucheing
douchely
douches
douchey
doucheyed
doucheyer
doucheyes
doucheying
doucheyly
doucheys
drunk
drunked
drunker
drunkes
drunking
drunkly
drunks
dumass
dumassed
dumasser
dumasses
dumassing
dumassly
dumasss
dumbass
dumbassed
dumbasser
dumbasses
dumbassesed
dumbasseser
dumbasseses
dumbassesing
dumbassesly
dumbassess
dumbassing
dumbassly
dumbasss
dummy
dummyed
dummyer
dummyes
dummying
dummyly
dummys
dyke
dykeed
dykeer
dykees
dykeing
dykely
dykes
dykesed
dykeser
dykeses
dykesing
dykesly
dykess
erotic
eroticed
eroticer
erotices
eroticing
eroticly
erotics
extacy
extacyed
extacyer
extacyes
extacying
extacyly
extacys
extasy
extasyed
extasyer
extasyes
extasying
extasyly
extasys
fack
facked
facker
fackes
facking
fackly
facks
fag
faged
fager
fages
fagg
fagged
faggeded
faggeder
faggedes
faggeding
faggedly
faggeds
fagger
fagges
fagging
faggit
faggited
faggiter
faggites
faggiting
faggitly
faggits
faggly
faggot
faggoted
faggoter
faggotes
faggoting
faggotly
faggots
faggs
faging
fagly
fagot
fagoted
fagoter
fagotes
fagoting
fagotly
fagots
fags
fagsed
fagser
fagses
fagsing
fagsly
fagss
faig
faiged
faiger
faiges
faiging
faigly
faigs
faigt
faigted
faigter
faigtes
faigting
faigtly
faigts
fannybandit
fannybandited
fannybanditer
fannybandites
fannybanditing
fannybanditly
fannybandits
farted
farter
fartes
farting
fartknocker
fartknockered
fartknockerer
fartknockeres
fartknockering
fartknockerly
fartknockers
fartly
farts
felch
felched
felcher
felchered
felcherer
felcheres
felchering
felcherly
felchers
felches
felching
felchinged
felchinger
felchinges
felchinging
felchingly
felchings
felchly
felchs
fellate
fellateed
fellateer
fellatees
fellateing
fellately
fellates
fellatio
fellatioed
fellatioer
fellatioes
fellatioing
fellatioly
fellatios
feltch
feltched
feltcher
feltchered
feltcherer
feltcheres
feltchering
feltcherly
feltchers
feltches
feltching
feltchly
feltchs
feom
feomed
feomer
feomes
feoming
feomly
feoms
fisted
fisteded
fisteder
fistedes
fisteding
fistedly
fisteds
fisting
fistinged
fistinger
fistinges
fistinging
fistingly
fistings
fisty
fistyed
fistyer
fistyes
fistying
fistyly
fistys
floozy
floozyed
floozyer
floozyes
floozying
floozyly
floozys
foad
foaded
foader
foades
foading
foadly
foads
fondleed
fondleer
fondlees
fondleing
fondlely
fondles
foobar
foobared
foobarer
foobares
foobaring
foobarly
foobars
freex
freexed
freexer
freexes
freexing
freexly
freexs
frigg
frigga
friggaed
friggaer
friggaes
friggaing
friggaly
friggas
frigged
frigger
frigges
frigging
friggly
friggs
fubar
fubared
fubarer
fubares
fubaring
fubarly
fubars
fuck
fuckass
fuckassed
fuckasser
fuckasses
fuckassing
fuckassly
fuckasss
fucked
fuckeded
fuckeder
fuckedes
fuckeding
fuckedly
fuckeds
fucker
fuckered
fuckerer
fuckeres
fuckering
fuckerly
fuckers
fuckes
fuckface
fuckfaceed
fuckfaceer
fuckfacees
fuckfaceing
fuckfacely
fuckfaces
fuckin
fuckined
fuckiner
fuckines
fucking
fuckinged
fuckinger
fuckinges
fuckinging
fuckingly
fuckings
fuckining
fuckinly
fuckins
fuckly
fucknugget
fucknuggeted
fucknuggeter
fucknuggetes
fucknuggeting
fucknuggetly
fucknuggets
fucknut
fucknuted
fucknuter
fucknutes
fucknuting
fucknutly
fucknuts
fuckoff
fuckoffed
fuckoffer
fuckoffes
fuckoffing
fuckoffly
fuckoffs
fucks
fucksed
fuckser
fuckses
fucksing
fucksly
fuckss
fucktard
fucktarded
fucktarder
fucktardes
fucktarding
fucktardly
fucktards
fuckup
fuckuped
fuckuper
fuckupes
fuckuping
fuckuply
fuckups
fuckwad
fuckwaded
fuckwader
fuckwades
fuckwading
fuckwadly
fuckwads
fuckwit
fuckwited
fuckwiter
fuckwites
fuckwiting
fuckwitly
fuckwits
fudgepacker
fudgepackered
fudgepackerer
fudgepackeres
fudgepackering
fudgepackerly
fudgepackers
fuk
fuked
fuker
fukes
fuking
fukly
fuks
fvck
fvcked
fvcker
fvckes
fvcking
fvckly
fvcks
fxck
fxcked
fxcker
fxckes
fxcking
fxckly
fxcks
gae
gaeed
gaeer
gaees
gaeing
gaely
gaes
gai
gaied
gaier
gaies
gaiing
gaily
gais
ganja
ganjaed
ganjaer
ganjaes
ganjaing
ganjaly
ganjas
gayed
gayer
gayes
gaying
gayly
gays
gaysed
gayser
gayses
gaysing
gaysly
gayss
gey
geyed
geyer
geyes
geying
geyly
geys
gfc
gfced
gfcer
gfces
gfcing
gfcly
gfcs
gfy
gfyed
gfyer
gfyes
gfying
gfyly
gfys
ghay
ghayed
ghayer
ghayes
ghaying
ghayly
ghays
ghey
gheyed
gheyer
gheyes
gheying
gheyly
gheys
gigolo
gigoloed
gigoloer
gigoloes
gigoloing
gigololy
gigolos
goatse
goatseed
goatseer
goatsees
goatseing
goatsely
goatses
godamn
godamned
godamner
godamnes
godamning
godamnit
godamnited
godamniter
godamnites
godamniting
godamnitly
godamnits
godamnly
godamns
goddam
goddamed
goddamer
goddames
goddaming
goddamly
goddammit
goddammited
goddammiter
goddammites
goddammiting
goddammitly
goddammits
goddamn
goddamned
goddamner
goddamnes
goddamning
goddamnly
goddamns
goddams
goldenshower
goldenshowered
goldenshowerer
goldenshoweres
goldenshowering
goldenshowerly
goldenshowers
gonad
gonaded
gonader
gonades
gonading
gonadly
gonads
gonadsed
gonadser
gonadses
gonadsing
gonadsly
gonadss
gook
gooked
gooker
gookes
gooking
gookly
gooks
gooksed
gookser
gookses
gooksing
gooksly
gookss
gringo
gringoed
gringoer
gringoes
gringoing
gringoly
gringos
gspot
gspoted
gspoter
gspotes
gspoting
gspotly
gspots
gtfo
gtfoed
gtfoer
gtfoes
gtfoing
gtfoly
gtfos
guido
guidoed
guidoer
guidoes
guidoing
guidoly
guidos
handjob
handjobed
handjober
handjobes
handjobing
handjobly
handjobs
hard on
hard oned
hard oner
hard ones
hard oning
hard only
hard ons
hardknight
hardknighted
hardknighter
hardknightes
hardknighting
hardknightly
hardknights
hebe
hebeed
hebeer
hebees
hebeing
hebely
hebes
heeb
heebed
heeber
heebes
heebing
heebly
heebs
hell
helled
heller
helles
helling
hellly
hells
hemp
hemped
hemper
hempes
hemping
hemply
hemps
heroined
heroiner
heroines
heroining
heroinly
heroins
herp
herped
herper
herpes
herpesed
herpeser
herpeses
herpesing
herpesly
herpess
herping
herply
herps
herpy
herpyed
herpyer
herpyes
herpying
herpyly
herpys
hitler
hitlered
hitlerer
hitleres
hitlering
hitlerly
hitlers
hived
hiver
hives
hiving
hivly
hivs
hobag
hobaged
hobager
hobages
hobaging
hobagly
hobags
homey
homeyed
homeyer
homeyes
homeying
homeyly
homeys
homo
homoed
homoer
homoes
homoey
homoeyed
homoeyer
homoeyes
homoeying
homoeyly
homoeys
homoing
homoly
homos
honky
honkyed
honkyer
honkyes
honkying
honkyly
honkys
hooch
hooched
hoocher
hooches
hooching
hoochly
hoochs
hookah
hookahed
hookaher
hookahes
hookahing
hookahly
hookahs
hooker
hookered
hookerer
hookeres
hookering
hookerly
hookers
hoor
hoored
hoorer
hoores
hooring
hoorly
hoors
hootch
hootched
hootcher
hootches
hootching
hootchly
hootchs
hooter
hootered
hooterer
hooteres
hootering
hooterly
hooters
hootersed
hooterser
hooterses
hootersing
hootersly
hooterss
horny
hornyed
hornyer
hornyes
hornying
hornyly
hornys
houstoned
houstoner
houstones
houstoning
houstonly
houstons
hump
humped
humpeded
humpeder
humpedes
humpeding
humpedly
humpeds
humper
humpes
humping
humpinged
humpinger
humpinges
humpinging
humpingly
humpings
humply
humps
husbanded
husbander
husbandes
husbanding
husbandly
husbands
hussy
hussyed
hussyer
hussyes
hussying
hussyly
hussys
hymened
hymener
hymenes
hymening
hymenly
hymens
inbred
inbreded
inbreder
inbredes
inbreding
inbredly
inbreds
incest
incested
incester
incestes
incesting
incestly
incests
injun
injuned
injuner
injunes
injuning
injunly
injuns
jackass
jackassed
jackasser
jackasses
jackassing
jackassly
jackasss
jackhole
jackholeed
jackholeer
jackholees
jackholeing
jackholely
jackholes
jackoff
jackoffed
jackoffer
jackoffes
jackoffing
jackoffly
jackoffs
jap
japed
japer
japes
japing
japly
japs
japsed
japser
japses
japsing
japsly
japss
jerkoff
jerkoffed
jerkoffer
jerkoffes
jerkoffing
jerkoffly
jerkoffs
jerks
jism
jismed
jismer
jismes
jisming
jismly
jisms
jiz
jized
jizer
jizes
jizing
jizly
jizm
jizmed
jizmer
jizmes
jizming
jizmly
jizms
jizs
jizz
jizzed
jizzeded
jizzeder
jizzedes
jizzeding
jizzedly
jizzeds
jizzer
jizzes
jizzing
jizzly
jizzs
junkie
junkieed
junkieer
junkiees
junkieing
junkiely
junkies
junky
junkyed
junkyer
junkyes
junkying
junkyly
junkys
kike
kikeed
kikeer
kikees
kikeing
kikely
kikes
kikesed
kikeser
kikeses
kikesing
kikesly
kikess
killed
killer
killes
killing
killly
kills
kinky
kinkyed
kinkyer
kinkyes
kinkying
kinkyly
kinkys
kkk
kkked
kkker
kkkes
kkking
kkkly
kkks
klan
klaned
klaner
klanes
klaning
klanly
klans
knobend
knobended
knobender
knobendes
knobending
knobendly
knobends
kooch
kooched
koocher
kooches
koochesed
koocheser
koocheses
koochesing
koochesly
koochess
kooching
koochly
koochs
kootch
kootched
kootcher
kootches
kootching
kootchly
kootchs
kraut
krauted
krauter
krautes
krauting
krautly
krauts
kyke
kykeed
kykeer
kykees
kykeing
kykely
kykes
lech
leched
lecher
leches
leching
lechly
lechs
leper
lepered
leperer
leperes
lepering
leperly
lepers
lesbiansed
lesbianser
lesbianses
lesbiansing
lesbiansly
lesbianss
lesbo
lesboed
lesboer
lesboes
lesboing
lesboly
lesbos
lesbosed
lesboser
lesboses
lesbosing
lesbosly
lesboss
lez
lezbianed
lezbianer
lezbianes
lezbianing
lezbianly
lezbians
lezbiansed
lezbianser
lezbianses
lezbiansing
lezbiansly
lezbianss
lezbo
lezboed
lezboer
lezboes
lezboing
lezboly
lezbos
lezbosed
lezboser
lezboses
lezbosing
lezbosly
lezboss
lezed
lezer
lezes
lezing
lezly
lezs
lezzie
lezzieed
lezzieer
lezziees
lezzieing
lezziely
lezzies
lezziesed
lezzieser
lezzieses
lezziesing
lezziesly
lezziess
lezzy
lezzyed
lezzyer
lezzyes
lezzying
lezzyly
lezzys
lmaoed
lmaoer
lmaoes
lmaoing
lmaoly
lmaos
lmfao
lmfaoed
lmfaoer
lmfaoes
lmfaoing
lmfaoly
lmfaos
loined
loiner
loines
loining
loinly
loins
loinsed
loinser
loinses
loinsing
loinsly
loinss
lubeed
lubeer
lubees
lubeing
lubely
lubes
lusty
lustyed
lustyer
lustyes
lustying
lustyly
lustys
massa
massaed
massaer
massaes
massaing
massaly
massas
masterbate
masterbateed
masterbateer
masterbatees
masterbateing
masterbately
masterbates
masterbating
masterbatinged
masterbatinger
masterbatinges
masterbatinging
masterbatingly
masterbatings
masterbation
masterbationed
masterbationer
masterbationes
masterbationing
masterbationly
masterbations
masturbate
masturbateed
masturbateer
masturbatees
masturbateing
masturbately
masturbates
masturbating
masturbatinged
masturbatinger
masturbatinges
masturbatinging
masturbatingly
masturbatings
masturbation
masturbationed
masturbationer
masturbationes
masturbationing
masturbationly
masturbations
methed
mether
methes
mething
methly
meths
militaryed
militaryer
militaryes
militarying
militaryly
militarys
mofo
mofoed
mofoer
mofoes
mofoing
mofoly
mofos
molest
molested
molester
molestes
molesting
molestly
molests
moolie
moolieed
moolieer
mooliees
moolieing
mooliely
moolies
moron
moroned
moroner
morones
moroning
moronly
morons
motherfucka
motherfuckaed
motherfuckaer
motherfuckaes
motherfuckaing
motherfuckaly
motherfuckas
motherfucker
motherfuckered
motherfuckerer
motherfuckeres
motherfuckering
motherfuckerly
motherfuckers
motherfucking
motherfuckinged
motherfuckinger
motherfuckinges
motherfuckinging
motherfuckingly
motherfuckings
mtherfucker
mtherfuckered
mtherfuckerer
mtherfuckeres
mtherfuckering
mtherfuckerly
mtherfuckers
mthrfucker
mthrfuckered
mthrfuckerer
mthrfuckeres
mthrfuckering
mthrfuckerly
mthrfuckers
mthrfucking
mthrfuckinged
mthrfuckinger
mthrfuckinges
mthrfuckinging
mthrfuckingly
mthrfuckings
muff
muffdiver
muffdivered
muffdiverer
muffdiveres
muffdivering
muffdiverly
muffdivers
muffed
muffer
muffes
muffing
muffly
muffs
murdered
murderer
murderes
murdering
murderly
murders
muthafuckaz
muthafuckazed
muthafuckazer
muthafuckazes
muthafuckazing
muthafuckazly
muthafuckazs
muthafucker
muthafuckered
muthafuckerer
muthafuckeres
muthafuckering
muthafuckerly
muthafuckers
mutherfucker
mutherfuckered
mutherfuckerer
mutherfuckeres
mutherfuckering
mutherfuckerly
mutherfuckers
mutherfucking
mutherfuckinged
mutherfuckinger
mutherfuckinges
mutherfuckinging
mutherfuckingly
mutherfuckings
muthrfucking
muthrfuckinged
muthrfuckinger
muthrfuckinges
muthrfuckinging
muthrfuckingly
muthrfuckings
nad
naded
nader
nades
nading
nadly
nads
nadsed
nadser
nadses
nadsing
nadsly
nadss
nakeded
nakeder
nakedes
nakeding
nakedly
nakeds
napalm
napalmed
napalmer
napalmes
napalming
napalmly
napalms
nappy
nappyed
nappyer
nappyes
nappying
nappyly
nappys
nazi
nazied
nazier
nazies
naziing
nazily
nazis
nazism
nazismed
nazismer
nazismes
nazisming
nazismly
nazisms
negro
negroed
negroer
negroes
negroing
negroly
negros
nigga
niggaed
niggaer
niggaes
niggah
niggahed
niggaher
niggahes
niggahing
niggahly
niggahs
niggaing
niggaly
niggas
niggased
niggaser
niggases
niggasing
niggasly
niggass
niggaz
niggazed
niggazer
niggazes
niggazing
niggazly
niggazs
nigger
niggered
niggerer
niggeres
niggering
niggerly
niggers
niggersed
niggerser
niggerses
niggersing
niggersly
niggerss
niggle
niggleed
niggleer
nigglees
niggleing
nigglely
niggles
niglet
nigleted
nigleter
nigletes
nigleting
nigletly
niglets
nimrod
nimroded
nimroder
nimrodes
nimroding
nimrodly
nimrods
ninny
ninnyed
ninnyer
ninnyes
ninnying
ninnyly
ninnys
nooky
nookyed
nookyer
nookyes
nookying
nookyly
nookys
nuccitelli
nuccitellied
nuccitellier
nuccitellies
nuccitelliing
nuccitellily
nuccitellis
nympho
nymphoed
nymphoer
nymphoes
nymphoing
nympholy
nymphos
opium
opiumed
opiumer
opiumes
opiuming
opiumly
opiums
orgies
orgiesed
orgieser
orgieses
orgiesing
orgiesly
orgiess
orgy
orgyed
orgyer
orgyes
orgying
orgyly
orgys
paddy
paddyed
paddyer
paddyes
paddying
paddyly
paddys
paki
pakied
pakier
pakies
pakiing
pakily
pakis
pantie
pantieed
pantieer
pantiees
pantieing
pantiely
panties
pantiesed
pantieser
pantieses
pantiesing
pantiesly
pantiess
panty
pantyed
pantyer
pantyes
pantying
pantyly
pantys
pastie
pastieed
pastieer
pastiees
pastieing
pastiely
pasties
pasty
pastyed
pastyer
pastyes
pastying
pastyly
pastys
pecker
peckered
peckerer
peckeres
peckering
peckerly
peckers
pedo
pedoed
pedoer
pedoes
pedoing
pedoly
pedophile
pedophileed
pedophileer
pedophilees
pedophileing
pedophilely
pedophiles
pedophilia
pedophiliac
pedophiliaced
pedophiliacer
pedophiliaces
pedophiliacing
pedophiliacly
pedophiliacs
pedophiliaed
pedophiliaer
pedophiliaes
pedophiliaing
pedophilialy
pedophilias
pedos
penial
penialed
penialer
peniales
penialing
penially
penials
penile
penileed
penileer
penilees
penileing
penilely
peniles
penis
penised
peniser
penises
penising
penisly
peniss
perversion
perversioned
perversioner
perversiones
perversioning
perversionly
perversions
peyote
peyoteed
peyoteer
peyotees
peyoteing
peyotely
peyotes
phuck
phucked
phucker
phuckes
phucking
phuckly
phucks
pillowbiter
pillowbitered
pillowbiterer
pillowbiteres
pillowbitering
pillowbiterly
pillowbiters
pimp
pimped
pimper
pimpes
pimping
pimply
pimps
pinko
pinkoed
pinkoer
pinkoes
pinkoing
pinkoly
pinkos
pissed
pisseded
pisseder
pissedes
pisseding
pissedly
pisseds
pisser
pisses
pissing
pissly
pissoff
pissoffed
pissoffer
pissoffes
pissoffing
pissoffly
pissoffs
pisss
polack
polacked
polacker
polackes
polacking
polackly
polacks
pollock
pollocked
pollocker
pollockes
pollocking
pollockly
pollocks
poon
pooned
pooner
poones
pooning
poonly
poons
poontang
poontanged
poontanger
poontanges
poontanging
poontangly
poontangs
porn
porned
porner
pornes
porning
pornly
porno
pornoed
pornoer
pornoes
pornography
pornographyed
pornographyer
pornographyes
pornographying
pornographyly
pornographys
pornoing
pornoly
pornos
porns
prick
pricked
pricker
prickes
pricking
prickly
pricks
prig
priged
priger
priges
priging
prigly
prigs
prostitute
prostituteed
prostituteer
prostitutees
prostituteing
prostitutely
prostitutes
prude
prudeed
prudeer
prudees
prudeing
prudely
prudes
punkass
punkassed
punkasser
punkasses
punkassing
punkassly
punkasss
punky
punkyed
punkyer
punkyes
punkying
punkyly
punkys
puss
pussed
pusser
pusses
pussies
pussiesed
pussieser
pussieses
pussiesing
pussiesly
pussiess
pussing
pussly
pusss
pussy
pussyed
pussyer
pussyes
pussying
pussyly
pussypounder
pussypoundered
pussypounderer
pussypounderes
pussypoundering
pussypounderly
pussypounders
pussys
puto
putoed
putoer
putoes
putoing
putoly
putos
queaf
queafed
queafer
queafes
queafing
queafly
queafs
queef
queefed
queefer
queefes
queefing
queefly
queefs
queer
queered
queerer
queeres
queering
queerly
queero
queeroed
queeroer
queeroes
queeroing
queeroly
queeros
queers
queersed
queerser
queerses
queersing
queersly
queerss
quicky
quickyed
quickyer
quickyes
quickying
quickyly
quickys
quim
quimed
quimer
quimes
quiming
quimly
quims
racy
racyed
racyer
racyes
racying
racyly
racys
rape
raped
rapeded
rapeder
rapedes
rapeding
rapedly
rapeds
rapeed
rapeer
rapees
rapeing
rapely
raper
rapered
raperer
raperes
rapering
raperly
rapers
rapes
rapist
rapisted
rapister
rapistes
rapisting
rapistly
rapists
raunch
raunched
rauncher
raunches
raunching
raunchly
raunchs
rectus
rectused
rectuser
rectuses
rectusing
rectusly
rectuss
reefer
reefered
reeferer
reeferes
reefering
reeferly
reefers
reetard
reetarded
reetarder
reetardes
reetarding
reetardly
reetards
reich
reiched
reicher
reiches
reiching
reichly
reichs
retard
retarded
retardeded
retardeder
retardedes
retardeding
retardedly
retardeds
retarder
retardes
retarding
retardly
retards
rimjob
rimjobed
rimjober
rimjobes
rimjobing
rimjobly
rimjobs
ritard
ritarded
ritarder
ritardes
ritarding
ritardly
ritards
rtard
rtarded
rtarder
rtardes
rtarding
rtardly
rtards
rum
rumed
rumer
rumes
ruming
rumly
rump
rumped
rumper
rumpes
rumping
rumply
rumprammer
rumprammered
rumprammerer
rumprammeres
rumprammering
rumprammerly
rumprammers
rumps
rums
ruski
ruskied
ruskier
ruskies
ruskiing
ruskily
ruskis
sadism
sadismed
sadismer
sadismes
sadisming
sadismly
sadisms
sadist
sadisted
sadister
sadistes
sadisting
sadistly
sadists
scag
scaged
scager
scages
scaging
scagly
scags
scantily
scantilyed
scantilyer
scantilyes
scantilying
scantilyly
scantilys
schlong
schlonged
schlonger
schlonges
schlonging
schlongly
schlongs
scrog
scroged
scroger
scroges
scroging
scrogly
scrogs
scrot
scrote
scroted
scroteed
scroteer
scrotees
scroteing
scrotely
scroter
scrotes
scroting
scrotly
scrots
scrotum
scrotumed
scrotumer
scrotumes
scrotuming
scrotumly
scrotums
scrud
scruded
scruder
scrudes
scruding
scrudly
scruds
scum
scumed
scumer
scumes
scuming
scumly
scums
seaman
seamaned
seamaner
seamanes
seamaning
seamanly
seamans
seamen
seamened
seamener
seamenes
seamening
seamenly
seamens
seduceed
seduceer
seducees
seduceing
seducely
seduces
semen
semened
semener
semenes
semening
semenly
semens
shamedame
shamedameed
shamedameer
shamedamees
shamedameing
shamedamely
shamedames
shit
shite
shiteater
shiteatered
shiteaterer
shiteateres
shiteatering
shiteaterly
shiteaters
shited
shiteed
shiteer
shitees
shiteing
shitely
shiter
shites
shitface
shitfaceed
shitfaceer
shitfacees
shitfaceing
shitfacely
shitfaces
shithead
shitheaded
shitheader
shitheades
shitheading
shitheadly
shitheads
shithole
shitholeed
shitholeer
shitholees
shitholeing
shitholely
shitholes
shithouse
shithouseed
shithouseer
shithousees
shithouseing
shithousely
shithouses
shiting
shitly
shits
shitsed
shitser
shitses
shitsing
shitsly
shitss
shitt
shitted
shitteded
shitteder
shittedes
shitteding
shittedly
shitteds
shitter
shittered
shitterer
shitteres
shittering
shitterly
shitters
shittes
shitting
shittly
shitts
shitty
shittyed
shittyer
shittyes
shittying
shittyly
shittys
shiz
shized
shizer
shizes
shizing
shizly
shizs
shooted
shooter
shootes
shooting
shootly
shoots
sissy
sissyed
sissyer
sissyes
sissying
sissyly
sissys
skag
skaged
skager
skages
skaging
skagly
skags
skank
skanked
skanker
skankes
skanking
skankly
skanks
slave
slaveed
slaveer
slavees
slaveing
slavely
slaves
sleaze
sleazeed
sleazeer
sleazees
sleazeing
sleazely
sleazes
sleazy
sleazyed
sleazyer
sleazyes
sleazying
sleazyly
sleazys
slut
slutdumper
slutdumpered
slutdumperer
slutdumperes
slutdumpering
slutdumperly
slutdumpers
sluted
sluter
slutes
sluting
slutkiss
slutkissed
slutkisser
slutkisses
slutkissing
slutkissly
slutkisss
slutly
sluts
slutsed
slutser
slutses
slutsing
slutsly
slutss
smegma
smegmaed
smegmaer
smegmaes
smegmaing
smegmaly
smegmas
smut
smuted
smuter
smutes
smuting
smutly
smuts
smutty
smuttyed
smuttyer
smuttyes
smuttying
smuttyly
smuttys
snatch
snatched
snatcher
snatches
snatching
snatchly
snatchs
sniper
snipered
sniperer
sniperes
snipering
sniperly
snipers
snort
snorted
snorter
snortes
snorting
snortly
snorts
snuff
snuffed
snuffer
snuffes
snuffing
snuffly
snuffs
sodom
sodomed
sodomer
sodomes
sodoming
sodomly
sodoms
spic
spiced
spicer
spices
spicing
spick
spicked
spicker
spickes
spicking
spickly
spicks
spicly
spics
spik
spoof
spoofed
spoofer
spoofes
spoofing
spoofly
spoofs
spooge
spoogeed
spoogeer
spoogees
spoogeing
spoogely
spooges
spunk
spunked
spunker
spunkes
spunking
spunkly
spunks
steamyed
steamyer
steamyes
steamying
steamyly
steamys
stfu
stfued
stfuer
stfues
stfuing
stfuly
stfus
stiffy
stiffyed
stiffyer
stiffyes
stiffying
stiffyly
stiffys
stoneded
stoneder
stonedes
stoneding
stonedly
stoneds
stupided
stupider
stupides
stupiding
stupidly
stupids
suckeded
suckeder
suckedes
suckeding
suckedly
suckeds
sucker
suckes
sucking
suckinged
suckinger
suckinges
suckinging
suckingly
suckings
suckly
sucks
sumofabiatch
sumofabiatched
sumofabiatcher
sumofabiatches
sumofabiatching
sumofabiatchly
sumofabiatchs
tard
tarded
tarder
tardes
tarding
tardly
tards
tawdry
tawdryed
tawdryer
tawdryes
tawdrying
tawdryly
tawdrys
teabagging
teabagginged
teabagginger
teabagginges
teabagginging
teabaggingly
teabaggings
terd
terded
terder
terdes
terding
terdly
terds
teste
testee
testeed
testeeed
testeeer
testeees
testeeing
testeely
testeer
testees
testeing
testely
testes
testesed
testeser
testeses
testesing
testesly
testess
testicle
testicleed
testicleer
testiclees
testicleing
testiclely
testicles
testis
testised
testiser
testises
testising
testisly
testiss
thrusted
thruster
thrustes
thrusting
thrustly
thrusts
thug
thuged
thuger
thuges
thuging
thugly
thugs
tinkle
tinkleed
tinkleer
tinklees
tinkleing
tinklely
tinkles
tit
tited
titer
tites
titfuck
titfucked
titfucker
titfuckes
titfucking
titfuckly
titfucks
titi
titied
titier
tities
titiing
titily
titing
titis
titly
tits
titsed
titser
titses
titsing
titsly
titss
tittiefucker
tittiefuckered
tittiefuckerer
tittiefuckeres
tittiefuckering
tittiefuckerly
tittiefuckers
titties
tittiesed
tittieser
tittieses
tittiesing
tittiesly
tittiess
titty
tittyed
tittyer
tittyes
tittyfuck
tittyfucked
tittyfucker
tittyfuckered
tittyfuckerer
tittyfuckeres
tittyfuckering
tittyfuckerly
tittyfuckers
tittyfuckes
tittyfucking
tittyfuckly
tittyfucks
tittying
tittyly
tittys
toke
tokeed
tokeer
tokees
tokeing
tokely
tokes
toots
tootsed
tootser
tootses
tootsing
tootsly
tootss
tramp
tramped
tramper
trampes
tramping
tramply
tramps
transsexualed
transsexualer
transsexuales
transsexualing
transsexually
transsexuals
trashy
trashyed
trashyer
trashyes
trashying
trashyly
trashys
tubgirl
tubgirled
tubgirler
tubgirles
tubgirling
tubgirlly
tubgirls
turd
turded
turder
turdes
turding
turdly
turds
tush
tushed
tusher
tushes
tushing
tushly
tushs
twat
twated
twater
twates
twating
twatly
twats
twatsed
twatser
twatses
twatsing
twatsly
twatss
undies
undiesed
undieser
undieses
undiesing
undiesly
undiess
unweded
unweder
unwedes
unweding
unwedly
unweds
uzi
uzied
uzier
uzies
uziing
uzily
uzis
vag
vaged
vager
vages
vaging
vagly
vags
valium
valiumed
valiumer
valiumes
valiuming
valiumly
valiums
venous
virgined
virginer
virgines
virgining
virginly
virgins
vixen
vixened
vixener
vixenes
vixening
vixenly
vixens
vodkaed
vodkaer
vodkaes
vodkaing
vodkaly
vodkas
voyeur
voyeured
voyeurer
voyeures
voyeuring
voyeurly
voyeurs
vulgar
vulgared
vulgarer
vulgares
vulgaring
vulgarly
vulgars
wang
wanged
wanger
wanges
wanging
wangly
wangs
wank
wanked
wanker
wankered
wankerer
wankeres
wankering
wankerly
wankers
wankes
wanking
wankly
wanks
wazoo
wazooed
wazooer
wazooes
wazooing
wazooly
wazoos
wedgie
wedgieed
wedgieer
wedgiees
wedgieing
wedgiely
wedgies
weeded
weeder
weedes
weeding
weedly
weeds
weenie
weenieed
weenieer
weeniees
weenieing
weeniely
weenies
weewee
weeweeed
weeweeer
weeweees
weeweeing
weeweely
weewees
weiner
weinered
weinerer
weineres
weinering
weinerly
weiners
weirdo
weirdoed
weirdoer
weirdoes
weirdoing
weirdoly
weirdos
wench
wenched
wencher
wenches
wenching
wenchly
wenchs
wetback
wetbacked
wetbacker
wetbackes
wetbacking
wetbackly
wetbacks
whitey
whiteyed
whiteyer
whiteyes
whiteying
whiteyly
whiteys
whiz
whized
whizer
whizes
whizing
whizly
whizs
whoralicious
whoralicioused
whoraliciouser
whoraliciouses
whoraliciousing
whoraliciously
whoraliciouss
whore
whorealicious
whorealicioused
whorealiciouser
whorealiciouses
whorealiciousing
whorealiciously
whorealiciouss
whored
whoreded
whoreder
whoredes
whoreding
whoredly
whoreds
whoreed
whoreer
whorees
whoreface
whorefaceed
whorefaceer
whorefacees
whorefaceing
whorefacely
whorefaces
whorehopper
whorehoppered
whorehopperer
whorehopperes
whorehoppering
whorehopperly
whorehoppers
whorehouse
whorehouseed
whorehouseer
whorehousees
whorehouseing
whorehousely
whorehouses
whoreing
whorely
whores
whoresed
whoreser
whoreses
whoresing
whoresly
whoress
whoring
whoringed
whoringer
whoringes
whoringing
whoringly
whorings
wigger
wiggered
wiggerer
wiggeres
wiggering
wiggerly
wiggers
woody
woodyed
woodyer
woodyes
woodying
woodyly
woodys
wop
woped
woper
wopes
woping
woply
wops
wtf
wtfed
wtfer
wtfes
wtfing
wtfly
wtfs
xxx
xxxed
xxxer
xxxes
xxxing
xxxly
xxxs
yeasty
yeastyed
yeastyer
yeastyes
yeastying
yeastyly
yeastys
yobbo
yobboed
yobboer
yobboes
yobboing
yobboly
yobbos
zoophile
zoophileed
zoophileer
zoophilees
zoophileing
zoophilely
zoophiles
anal
ass
ass lick
balls
ballsac
bisexual
bleach
causas
cheap
cost of miracles
cunt
display network stats
fart
fda and death
fda AND warn
fda AND warning
fda AND warns
feom
fuck
gfc
humira AND expensive
illegal
madvocate
masturbation
nuccitelli
overdose
porn
shit
snort
texarkana
direct\-acting antivirals
assistance
ombitasvir
support path
harvoni
abbvie
direct-acting antivirals
paritaprevir
advocacy
ledipasvir
vpak
ritonavir with dasabuvir
program
gilead
greedy
financial
needy
fake-ovir
viekira pak
v pak
sofosbuvir
support
oasis
discount
dasabuvir
protest
ritonavir
Negative Keywords Excluded Elements
header[@id='header']
section[contains(@class, 'nav-hidden')]
footer[@id='footer']
div[contains(@class, 'pane-pub-article-cleveland-clinic')]
div[contains(@class, 'pane-pub-home-cleveland-clinic')]
div[contains(@class, 'pane-pub-topic-cleveland-clinic')]
div[contains(@class, 'panel-panel-inner')]
div[contains(@class, 'pane-node-field-article-topics')]
section[contains(@class, 'footer-nav-section-wrapper')]
Altmetric
DSM Affiliated
Display in offset block
Disqus Exclude
Best Practices
CE/CME
Education Center
Medical Education Library
Enable Disqus
Display Author and Disclosure Link
Publication Type
Society
Slot System
Featured Buckets
Disable Sticky Ads
Disable Ad Block Mitigation
Featured Buckets Admin
LayerRx MD-IQ Id
773
Show Ads on this Publication's Homepage
Consolidated Pub
Show Article Page Numbers on TOC
Use larger logo size
Off
publication_blueconic_enabled
Off
Show More Destinations Menu
Disable Adhesion on Publication
Off
Restore Menu Label on Mobile Navigation
Disable Facebook Pixel from Publication
Exclude this publication from publication selection on articles and quiz

Role of the incretin pathway in the pathogenesis of type 2 diabetes mellitus

Article Type
Changed
Tue, 10/02/2018 - 11:45
Display Headline
Role of the incretin pathway in the pathogenesis of type 2 diabetes mellitus

It has long been understood that the pathophysiology of type 2 diabetes mellitus (T2DM) is based on the triad of progressive decline in insulin-producing pancreatic beta cells, an increase in insulin resistance, and increased hepatic glucose production.1,2 It is now evident that other factors, including defective actions of the gastrointestinal (GI) incretin hormones glucagon-like peptide–1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), also play significant roles.2–5 The uncontrolled hyperglycemia resulting from such defects may lead to microvascular complications, including retinopathy, neuropathy, microangiopathy, and nephropathy, and macrovascular complications, such as coronary artery disease and peripheral vascular disease.

This review explores the growing understanding of the role of the incretins in normal insulin secretion, as well as in the pathogenesis of T2DM, and examines the pathophysiologic basis for the benefits and therapeutic application of incretin-based therapies in T2DM.1,2

THE GI SYSTEM AND GLUCOSE HOMEOSTASIS IN THE HEALTHY STATE

The GI system plays an integral role in glucose homeostasis.6 The observation that orally administered glucose provides a stronger insulinotropic stimulus than an intravenous glucose challenge provided insight into the regulation of plasma glucose by the GI system of healthy individuals.7 The incretin effect, as this is termed, may be responsible for 50% to 70% of the total insulin secreted following oral glucose intake.8

Two GI peptide hormones (the incretins)—GLP-1 and GIP—were found to exert major glucoregulatory actions.3,9,10 Within minutes of nutrient ingestion, GLP-1 is secreted from intestinal L cells in the distal ileum and colon, while GIP is released by intestinal K cells in the duodenum and jejunum.3 GLP-1 and GIP trigger their insulinotropic actions by binding beta-cell receptors.3 GLP-1 receptors are expressed on pancreatic glucagon-containing alpha and delta cells as well as on beta cells, whereas GIP receptors are expressed primarily on beta cells.3,8 GLP-1 receptors are also expressed in the central nervous system (CNS), peripheral nervous system, lung, heart, and GI tract, while GIP receptors are expressed in adipose tissue and the CNS.3 GLP-1 inhibits glucose-dependent glucagon secretion from alpha cells.3 In healthy individuals, fasting glucose is managed by tonic insulin/glucagon secretion, but excursions of postprandial glucose (PPG) are controlled by insulin and the incretin hormones.11

Additionally, in animal studies, GLP-1 has been shown to induce the transcriptional activation of the insulin gene and insulin biosynthesis, thus increasing beta-cell proliferation and decreasing beta-cell apoptosis.12 GLP-1 stimulates a CNS-mediated pathway of insulin secretion, slows gastric emptying, increases CNS-mediated satiety leading to reduced food intake, indirectly increases insulin sensitivity and nutrient uptake in skeletal muscle and adipose tissue, and exerts neuroprotective effects.8

Reprinted, with permission, from European Journal of Endocrinology (Van Gaal LF, et al. Eur J Endocrinol 2008; 158:773–784),13 Copyright © 2008 European Society of Endocrinology.
Figure 1. Biologic actions of GIP and GLP-1 in relation to the pathophysiology of type 2 diabetes mellitus. GIP = glucose-dependent insulinotropic polypeptide, GLP-1 = glucagon-like peptide–1; PP = postprandial; solid arrows = potentially beneficial actions; dashed arrows = potentially harmful actions; slashed arrows = actions with no effect.
Along with its insulinotropic action, GIP has been shown in animal studies to inhibit gastric acid secretion, bioregulate fat metabolism in adipocytes, increase glucagon secretion and fat deposition, increase beta-cell replication, and decrease beta-cell apoptosis.8Figure 1 illustrates the biologic actions of GLP-1 and GIP.13

Both GLP-1 and GIP are rapidly degraded by the serine protease dipeptidyl peptidase–4 (DPP-4), which is widely expressed in bound and free forms.14 A recent study in healthy adults showed that GLP-1 concentration declined even during maximal DPP-4 inhibition, suggesting that there may be pathways of GLP-1 elimination other than DPP-4 enzymatic degradation.15

INCRETINS AND THE PATHOGENESIS OF T2DM

Studies have shown that incretin pathways play a role in the progression of T2DM.3,16 The significant reduction in the incretin effect seen in patients with T2DM has been attributed to several factors, including impaired secretion of GLP-1, accelerated metabolism of GLP-1 and GIP, and defective responsiveness to both hormones.16 Many patients with T2DM also have accelerated gastric emptying that may contribute to deterioration of their glycemic control.17

While GIP concentration is normal or modestly increased in patients with T2DM,16,18 the insulinotropic actions of GIP are significantly diminished.19 Thus, patients with T2DM have an impaired responsiveness to GIP with a possible link to GIP-receptor downregulation or desensitization.20

Are secretory defects a cause or result of T2DM?

In contrast to GIP, the secretion of GLP-1 has been shown to be deficient in patients with T2DM.18 As with GIP, it is unknown to what degree this defect is a cause or consequence of T2DM. In a study of identical twins, defective GLP-1 secretion was observed only in the one sibling with T2DM, suggesting that GLP-1 secretory deficits may be secondary to the development of T2DM.21 Despite the diminished secretion of GLP-1 in patients with T2DM, the insulinotropic actions of GLP-1 are preserved.19 It has also been shown that the effects of GLP-1 on gastric emptying and glucagon secretion are maintained in patients with T2DM.19,22,23

Whether this incretin dysregulation is responsible for or is the end result of hyperglycemia remains a subject of continued investigation. A recent study confirmed that the incretin effect is reduced in patients with T2DM, but advanced the concept that it may be a consequence of the diabetic state.16,24 Notably, impaired actions of GLP-1 and GIP and diminished concentrations of GLP-1 may be partially restored by improved glycemic control.24

Recent preclinical and clinical studies continue to clarify the roles of incretin hormones in T2DM. The findings from a study of obese diabetic mice suggest that the effect of GLP-1 therapy on the long-term remission of diabetes may be caused by improvements in beta-cell function and insulin sensitivity, as well as by a reduction in gluconeogenesis in the liver.25

Incretin effect and glucose tolerance, body mass index

Another study was conducted to evaluate quantitatively the separate impacts of obesity and hyperglycemia on the incretin effect in patients with T2DM, patients with impaired glucose tolerance, and patients with normal glucose tolerance.26 There was a significant (P ≤ .05) reduction in the incretin effect in terms of total insulin secretion, beta-cell glucose sensitivity, and the GLP-1 response to oral glucose in patients with T2DM compared with individuals whose glucose tolerance was normal or impaired. Each manifestation of the incretin effect was inversely related to both glucose tolerance and body mass index in an independent, additive manner (P ≤ .05); thus, glucose tolerance and obesity attenuate the incretin effect on beta-cell function and GLP-1 response independently of each other.

Exogenous GLP-1 has been shown to restore the regulation of blood glucose to near-normal concentrations in patients with T2DM.27 Several studies of patients with T2DM have shown that synthetic GLP-1 administration induces insulin secretion,19,27 slows gastric emptying (which is accelerated in patients with T2DM), and decreases inappropriately elevated glucagon secretion.19,23,28 Acute GLP-1 infusion studies showed that GLP-1 improved fasting plasma glucose (FPG) and PPG concentrations23,27; long-term studies showed that this hormone exerts euglycemic effects, leading to improvements in glycosylated hemoglobin (HbA1c), and induces weight loss.29

 

 

TARGETING FUNDAMENTAL DEFECTS OF T2DM WITH INCRETIN-BASED THERAPIES

Recognition and a better understanding of the role of the incretins and the enzyme involved in their degradation have led to the development of two incretin-based treatments: the GLP-1 receptor agonists, which possess many of the glucoregulatory actions of incretin peptides, and the DPP-4 inhibitors.5 Both the GLP-1 receptor agonists and the DPP-4 inhibitors have demonstrated safety and efficacy in the management of hyperglycemia in patients with T2DM.

GLP-1 receptor agonists

The GLP-1 receptor agonist exenatide is a synthetic form of exendin-4 and has a unique amino acid sequence that renders it resistant to degradation by DPP-4, making its actions longer lasting than endogenous GLP-1.5,30 Exenatide has a half-life of 2.4 hours and is detectable for up to 10 hours after subcutaneous (SC) injection.5,30 It is administered BID and has been approved as monotherapy or an adjunct therapy in patients with T2DM who have inadequate glycemic control following treatment with metformin, a sulfonylurea, a thiazolidinedione (TZD), or metformin in combination with a sulfonylurea or a TZD.31–35

In both human and animal studies, exenatide has been shown to enhance glucose-dependent insulin secretion and suppress inappropriate glucagon secretion in a glucose-dependent manner, reduce food intake and body weight, and acutely improve beta-cell function by enhancing first- and second-phase insulin secretion.5,36,37

In a small study involving 17 patients with T2DM, exenatide was shown to slow gastric emptying, which could be an important mechanism contributing to its beneficial effects on PPG concentration.38 Exenatide also has been shown to attenuate postprandial hyper­glycemia, a risk factor for cardiovascular disease (CVD),  by reducing endogenous glucose production by about 50% in patients with T2DM.39 Another mechanism for glycemic control may exist, as a recent animal study has shown that exenatide, similar to endogenous GLP-1, lowers blood glucose concentration independent of changes in pancreatic islet hormone secretion or delayed gastric emptying.40

A formulation of exenatide that is administered once weekly—exenatide long-acting release (LAR)—is in clinical evaluation and under review by the US Food and Drug Administration (FDA). In a short-term study, exenatide-LAR (0.8 mg or 2.0 mg) was administered once weekly for 15 weeks to patients with T2DM whose glycemia was suboptimally controlled with metformin alone or in combination with diet and exercise. Compared with placebo, treatment with exenatide once weekly was associated with markedly reduced HbA1c, FPG, PPG and body weight.41 In a larger, 30-week, phase 3 trial, Diabetes Therapy Utilization: Researching Changes in A1C, Weight and Other Factors Through Intervention with Exenatide ONce Weekly (DURATION-1), exenatide-LAR 2 mg once weekly was compared with exenatide 10 mg BID in patients with T2DM. Exenatide-LAR once weekly was associated with a significantly greater reduction in HbA1c (–1.9% vs –1.5%, P = .0023), and with a similar low risk of hypoglycemia and reduction in body weight (–3.7 kg vs –3.6 kg, P = .89) compared with the BID formulation.42

Liraglutide, recently approved in the European Union for T2DM and also under regulatory review in the United States, is a DPP-4–resistant human analogue GLP-1 receptor agonist in clinical development that has a 97% homology to native GLP-1.43–45 In contrast to exenatide, the acetylated liraglutide molecule allows binding to serum albumin and provides resistance to DPP-4 degradation, thus prolonging the half-life of liraglutide to approximately 12 hours. Liraglutide is administered SC QD as monotherapy or in combination with other antidiabetes agents such as metformin or sulfonylurea to patients with T2DM.44–47 Liraglutide has been shown to reduce HbA1c, decrease body weight, and lead to a lower incidence of hypoglycemia compared with the sulfonylurea glimepiride.

DPP-4 inhibitors

Sitagliptin is a DPP-4 inhibitor indicated as monotherapy or in combination with metformin or a TZD in patients with T2DM with inadequate glycemic control.48–51 Given orally, sitagliptin does not bind to the GLP-1 receptor agonist and has been shown to inhibit circulating DPP-4 activity by about 80%.52,53 Sitagliptin has been associated with an approximate twofold increase in postprandial GLP-1 plasma concentrations compared with placebo in healthy human subjects and in patients with T2DM.53 Saxagliptin, another potent DPP-4 inhibitor, significantly reduced HbA1c and FPG concentrations in patients with T2DM54 with a neutral effect on weight; it was recently approved by the FDA for treatment of T2DM.55

The DPP-4 inhibitor vildagliptin is currently being used in the European Union and Latin America but has yet to receive regulatory approval in the United States.54 Alogliptin, a novel, high-affinity, high-specificity DPP-4 inhibitor currently in development, provides rapid and sustained DPP-4 inhibition and significantly reduces HbA1c, FPG, and PPG concentrations with no change in body weight in patients with T2DM.56,57

Incretin-based therapies compared

In a recent head-to-head crossover trial between the GLP-1 receptor agonist exenatide and the DPP-4 inhibitor sitagliptin, exenatide had a greater effect in reducing 2-hour PPG.52 Patients with T2DM who switched from sitagliptin to exenatide showed a further reduction in 2-hour PPG concentration. Exenatide was also more potent than sitagliptin in increasing insulin secretion, reducing postprandial glucagon secretion, and decreasing triglycerides.52 Finally, exenatide slowed gastric emptying and reduced caloric intake. The differences between the two incretin-based therapies and their effects on glycemic control could be attributed to the pharmacologic concentration of the GLP-1 receptor agonist exenatide that is available for GLP-1 receptor activation compared with the twofold rise in endogenous GLP-1 concentration seen with the DPP-4 inhibitor sitagliptin.52

A comparison of the actions of the GLP-1 receptor agonists and DPP-4 inhibitors in patients with T2DM is provided in Table 1,52,58 and an overview of incretin-based therapies is presented in Table 2.45,54,59 GLP-1 receptor agonists induce weight loss in patients with T2DM, while DPP-4 inhibitors are weight neutral.3,52,58,60 The GLP-1 receptor agonists are associated with a much higher incidence of adverse GI effects such as nausea and vomiting, presumably also attributable to the pharmacologic levels achieved.

 

 

Effects of incretin-based therapies

The number of people with T2DM, overweight/obesity, or CVD, alone or in combination, is approaching epidemic proportions, with the mechanisms of these conditions interrelated. Approximately 24 million Americans have diabetes, and T2DM accounts for more than 90% of these cases.61 Most patients with T2DM are not achieving HbA1c targets.62–64 About 60% of deaths among patients with T2DM are caused by CVD.65 Compounding the problem, overweight/obesity enhances the risk for CV-related morbidities in patients with diabetes.66 A cluster of metabolic disorders referred to as the metabolic syndrome (which includes hyperglycemia, measures of central obesity, and a series of significant CV risk factors) is common in patients with T2DM and CVD.67 Unfortunately, many antidiabetes drugs that successfully manage glycemic control also cause weight gain, which in theory may increase CV risk in patients with T2DM.68

Data from studies of patients with T2DM show that exenatide improves glycemic control and reduces body weight. Exenatide administered BID significantly reduced HbA1c (–0.40% to –0.86%) and weight (–1.6 kg to –2.8 kg) relative to baseline in three 30-week, placebo-controlled clinical trials.31,33,34 In subsequent 2-year, open-label extension studies, exenatide produced significant reductions from baseline in HbA1c (–20.9% at 30 weeks) and weight (–2.1 kg at 30 weeks). Both decreases were sustained through 2 years (HbA1c –1.1%, weight –4.7 kg) with a low incidence of hypoglycemia.31 Further post hoc analysis of the open-label extension of the 30-week trials followed patients treated with exenatide BID for 3 years or longer.69 In addition to markedly decreasing HbA1c from baseline levels (–1.1% at 3 years and –0.8% at up to 3.5 years; P < .0001), adjunctive exenatide produced significant reductions in body weight—up to –5.3 kg after 3.5 years of therapy.31,69 At 3.5 years, continued exenatide therapy resulted in a –6% reduction in low-density lipoprotein cholesterol, a 24% mean increase in high-density lipoprotein cholesterol, and a mean reduction in blood pressure of –2% to –4% from baseline levels. Improvements in hepatic biomarkers and homeostasis model assessment-B, a measure of beta-cell function, were seen after 2 and 3 years of exenatide treatment.31 Hypoglycemia was generally mild and transient.

In comparative head-to-head studies, exenatide BID and insulin analogues reduced HbA1c by similar magnitudes; yet exenatide treatment resulted in better control in terms of PPG and weight loss, while insulin glargine and insulin aspart produced weight gain.70–73

Mechanisms of cardioprotective effects

Although the mechanisms for the potential cardioprotective effects of GLP-1 and its receptor agonists remain to be fully elucidated, a recent study suggested that two novel pathways could be involved—one that is dependent on the known GLP-1 receptor pathway, and one that is independent of the GLP-1 receptor pathway.74 Correlating with observations of a potential cardioprotective effect, an infusion of recombinant GLP-1 in patients with acute myocardial infarction, when added to standard therapy, resulted in improved left ventricular function and was associated with reduced mortality.75 Evidence continues to accumulate for potential cardioprotective effects of the GLP-1 receptor agonists, indicating that they may have a positive impact on macro­vascular complications in patients with T2DM.

CONCLUSION

T2DM, which is often associated with overweight and obesity, remains a significant challenge worldwide. The broad spectrum of glucoregulatory actions of the incretin hormones GLP-1 and GIP, and their importance in maintaining glucose homeostasis, have been recognized and correlated with the pathogenesis of T2DM. An improved understanding of the roles played by GLP-1 and GIP in the pathogenesis of T2DM may provide clinicians with important details regarding the therapeutic application of incretin-based therapies, including the GLP-1 receptor agonist exenatide and the DPP-4 inhibitors sitagliptin and saxagliptin. Antidiabetes agents whose development is based on the multiple pharmacologic effects of incretin hormones can address the multifaceted nature of T2DM and overcome some current limitations of traditional therapies, especially those related to weight. This becomes more compelling given the close link among T2DM, obesity, and increased CV risk.

References
  1. Boyle PJ, Freeman JS. Application of incretin mimetics and dipeptidyl peptidase IV inhibitors in managing type 2 diabetes mellitus. J Am Osteopath Assoc 2007; 107(suppl 3):S10–S16.
  2. Freeman JS. The pathophysiologic role of incretins. J Am Osteopath Assoc 2007; 107(suppl 3):S6–S9.
  3. Drucker DJ, Nauck MA. The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 2006; 368:1696–1705.
  4. Nauck MA, Baller B, Meier JJ. Gastric inhibitory polypeptide and glucagon-like peptide-1 in the pathogenesis of type 2 diabetes. Diabetes 2004; 53(suppl 3):S190–S196.
  5. Stonehouse A, Okerson T, Kendall D, Maggs D. Emerging incretin based therapies for type 2 diabetes: incretin mimetics and DPP-4 inhibitors. Curr Diabetes Rev 2008; 4:101–109.
  6. Huda MS, Wilding JP, Pinkney JH. Gut peptides and the regulation of appetite. Obes Rev 2006; 7:163–182.
  7. Elrick H, Stimmler L, Hlad CJ Jr, Arai Y. Plasma insulin response to oral and intravenous glucose administration. J Clin Endocrinol Metab 1964; 24:1076–1082.
  8. Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP. Gastroenterology 2007; 132:2131–2157.
  9. Brown JC, Dryburgh JR, Ross SA, Dupré J. Identification and actions of gastric inhibitory polypeptide. Recent Prog Horm Res 1975; 31:487–532.
  10. Kreymann B, Williams G, Ghatei MA, Bloom SR. Glucagon-like peptide-1 7-36: a physiological incretin in man. Lancet 1987; 2:1300–1304.
  11. Nauck MA, Homberger E, Siegel EG, et al. Incretin effects of increasing glucose loads in man calculated from venous insulin and C-peptide responses. J Clin Endocrinol Metab 1986; 63:492–498.
  12. Farilla L, Bulotta A, Hirshberg B, et al. Glucagon-like peptide 1 inhibits cell apoptosis and improves glucose responsiveness of freshly isolated human islets. Endocrinology 2003; 144:5149–5158.
  13. Van Gaal LF, Gutkin SW, Nauck MA. Exploiting the antidiabetic properties of incretins to treat type 2 diabetes mellitus: glucagon-like peptide 1 receptor agonists or insulin for patients with inadequate glycemic control? Eur J Endocrinol 2008; 158:773–784.
  14. Deacon CF, Nauck MA, Toft-Nielsen M, Pridal L, Willms B, Holst JJ. Both subcutaneously and intravenously administered glucagon-like peptide I are rapidly degraded from the NH2-terminus in type II diabetic patients and in healthy subjects. Diabetes 1995; 44:1126–1131.
  15. Dai H, Gustavson SM, Preston GM, Eskra JD, Calle R, Hirshberg B. Non-linear increase in GLP-1 levels in response to DPP-IV inhibition in healthy adult subjects. Diabetes Obes Metab 2008; 10:506–513.
  16. Nauck MA, Stöckmann F, Ebert R, Creutzfeldt W. Reduced incretin effect in type 2 (non-insulin-dependent) diabetes. Diabetologia 1986; 29:46–52.
  17. Phillips WT, Schwartz JG, McMahan CA. Rapid gastric emptying of an oral glucose solution in type 2 diabetic patients. J Nucl Med 1992; 33:1496–1500.
  18. Toft-Nielsen MB, Damholt MB, Madsbad S, et al. Determinants of the impaired secretion of glucagon-like peptide-1 in type 2 diabetic patients. J Clin Endocrinol Metab 2001; 86:3717–3723.
  19. Nauck MA, Heimesaat MM, Orskov C, Holst JJ, Ebert R, Creutzfeldt W. Preserved incretin activity of glucagon-like peptide 1 [7-36 amide] but not of synthetic human gastric inhibitory polypeptide in patients with type-2 diabetes mellitus. J Clin Invest 1993; 91:301–307.
  20. Lynn FC, Thompson SA, Pospisilik JA, et al. A novel pathway for regulation of glucose-dependent insulinotropic polypeptide (GIP) receptor expression in beta cells. FASEB J 2003; 17:91–93.
  21. Vaag AA, Holst JJ, Vølund A, Beck-Nielsen HB. Gut incretin hormones in identical twins discordant for non-insulin-dependent diabetes mellitus (NIDDM)—evidence for decreased glucagon-like peptide-1 secretion during oral glucose ingestion in NIDDM twins. Eur J Endocrinol 1996; 135:425–432.
  22. Meier JJ, Gallwitz B, Salmen S, et al. Normalization of glucose concentrations and deceleration of gastric emptying after solid meals during intravenous glucagon-like peptide 1 in patients with type 2 diabetes. J Clin Endocrinol Metab 2003; 88:2719–2725.
  23. Nauck MA, Kleine N, Orskov C, Holst JJ, Willms B, Creutzfeldt W. Normalization of fasting hyperglycaemia by exogenous glucagon-like peptide 1 (7-36 amide) in type 2 (non-insulin-dependent) diabetic patients. Diabetologia 1993; 36:741–744.
  24. Knop FK, Vilsbøll T, Højberg PV, et al. Reduced incretin effect in type 2 diabetes: cause or consequence of the diabetic state? Diabetes 2007; 56:1951–1959.
  25. Lee YS, Shin S, Shigihara T, et al. Glucagon-like peptide-1 gene therapy in obese diabetic mice results in long-term cure of diabetes by improving insulin sensitivity and reducing hepatic gluconeogenesis. Diabetes 2007; 56:1671–1679.
  26. Muscelli E, Mari A, Casolaro A, et al. Separate impact of obesity and glucose tolerance on the incretin effect in normal subjects and type 2 diabetic patients. Diabetes 2008; 57:1340–1348.
  27. Nathan DM, Schreiber E, Fogel H, Mojsov S, Habener JF. Insulinotropic action of glucagonlike peptide-I-(7-37) in diabetic and nondiabetic subjects. Diabetes Care 1992; 15:270–276.
  28. Kolterman OG, Buse JB, Fineman MS, et al. Synthetic exendin-4 (exenatide) significantly reduces postprandial and fasting plasma glucose in subjects with type 2 diabetes. J Clin Endocrinol Metab 2003; 88:3082–3089.
  29. Zander M, Madsbad S, Madsen JL, Holst JJ. Effect of 6-week course of glucagon-like peptide 1 on glycaemic control, insulin sensitivity, and beta-cell function in type 2 diabetes: a parallel-group study. Lancet 2002; 359:824–830.
  30. Kolterman OG, Kim DD, Shen L, et al. Pharmacokinetics, pharmacodynamics, and safety of exenatide in patients with type 2 diabetes mellitus. Am J Health Syst Pharm 2005; 62:173–181.
  31. Buse JB, Henry RR, Han J, et al. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in sulfonylurea-treated patients with type 2 diabetes. Diabetes Care 2004; 27:2628–2635.
  32. Byetta [package insert]. San Diego, CA: Amylin Pharmaceuticals, Inc.; 2009.
  33. DeFronzo RA, Ratner RE, Han J, Kim DD, Fineman MS, Baron AD. Effects of exenatide (exendin-4) on glycemic control and weight over 30 weeks in metformin-treated patients with type 2 diabetes. Diabetes Care 2005; 28:1092–1100.
  34. Kendall DM, Riddle MC, Rosenstock J, et al. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in patients with type 2 diabetes treated with metformin and a sulfonylurea. Diabetes Care 2005; 28:1083–1091.
  35. Zinman B, Hoogwerf BJ, Durán García S, et al. The effect of adding exenatide to a thiazolidinedione in suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med 2007; 146:477–485.
  36. Fehse F, Trautmann M, Holst JJ, et al. Exenatide augments first- and second-phase insulin secretion in response to intravenous glucose in subjects with type 2 diabetes. J Clin Endocrinol Metab 2005; 90:5991–5997.
  37. Parkes DG, Pittner R, Jodka C, Smith P, Young A. Insulinotropic actions of exendin-4 and glucagon-like peptide-1 in vivo and in vitro. Metabolism 2001; 50:583–589.
  38. Linnebjerg H, Park S, Kothare PA, et al. Effect of exenatide on gastric emptying and relationship to postprandial glycemia in type 2 diabetes. Regul Pept 2008; 151:123–129.
  39. Cervera A, Wajcberg E, Sriwijitkamol A, et al. Mechanism of action of exenatide to reduce postprandial hyperglycemia in type 2 diabetes. Am J Physiol Endocrinol Metab 2008; 294:E846–E852.
  40. Ionut V, Zheng D, Stefanovski D, Bergman RN. Exenatide can reduce glucose independent of islet hormones or gastric emptying. Am J Physiol Endocrinol Metab 2008; 295:E269–E277.
  41. Kim D, MacConell L, Zhuang D, et al. Effects of once-weekly dosing of a long-acting release formulation of exenatide on glucose control and body weight in subjects with type 2 diabetes. Diabetes Care 2007; 30:1487–1493.
  42. Drucker DJ, Buse JB, Taylor K, et al. Exenatide once weekly versus twice daily for the treatment of type 2 diabetes: a randomised, open-label, non-inferiority study. Lancet 2008; 372:1240–1250.
  43. Knudsen LB, Nielsen PF, Huusfeldt PO, et al. Potent derivatives of glucagon-like peptide-1 with pharmacokinetic properties suitable for once daily administration. J Med Chem 2000; 43:1664–1669.
  44. Nauck M, Frid A, Hermansen K, et al; for the LEAD-2 Study Group. Efficacy and safety comparison of liraglutide, glimepiride, and placebo, all in combination with metformin, in type 2 diabetes: the LEAD (liraglutide effect and action in diabetes)-2 study. Diabetes Care 2009; 32:84–90.
  45. Committee for Medicinal Products for Human Use: Summary of Positive Opinion for Victoza. European Medicines Agency Web site. http://www.emea.europa.eu/pdfs/human/opinion/Victoza_14168909en.pdf. Published April 23, 2009. Accessed September 21, 2009.
  46. Garber A, Henry R, Ratner R, et al. Liraglutide versus glimepiride monotherapy for type 2 diabetes (LEAD-3 Mono): a randomised, 52-week, phase III, double-blind, parallel-treatment trial. Lancet 2009; 373:473–481.
  47. Marre M, Shaw J, Brändle M, et al. Liraglutide, a once-daily human GLP-1 analogue, added to a sulphonylurea over 26 weeks produces greater improvements in glycaemic and weight control compared with adding rosiglitazone or placebo in subjects with type 2 diabetes (LEAD-1 SU). Diabet Med 2009; 26:268–278.
  48. Aschner P, Kipnes MS, Lunceford JK, et al; for the Sitagliptin 021 Study Group. Effect of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy on glycemic control in patients with type 2 diabetes. Diabetes Care 2006; 29:2632–2637.
  49. Charbonnel B, Karasik A, Liu J, Wu M, Meininger G; for the Sitagliptin Study 020 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin added to ongoing metformin therapy in patients with type 2 diabetes inadequately controlled with metformin alone. Diabetes Care 2006; 29:2638–2643.
  50. Raz I, Hanefeld M, Xu L, et al. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy in patients with type 2 diabetes mellitus. Diabetologia 2006; 49:2564–2571.
  51. Rosenstock J, Brazg R, Andryuk PJ, Lu K, Stein P; for the Sitagliptin Study 019 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin added to ongoing pioglitazone therapy in patients with type 2 diabetes: a 24-week, multicenter, randomized, double-blind, placebo-controlled, parallel-group study. Clin Ther 2006; 28:1556–1568.
  52. DeFronzo RA, Okerson T, Viswanathan P, Guan X, Holcombe JH, MacConell L. Effects of exenatide versus sitagliptin on postprandial glucose, insulin and glucagon secretion, gastric emptying, and caloric intake: a randomized, cross-over study. Curr Med Res Opin 2008; 24:2943–2952.
  53. Herman GA, Stevens C, Van Dyck K, et al. Pharmacokinetics and pharmacodynamics of sitagliptin, an inhibitor of dipeptidyl peptidase IV, in healthy subjects: results from two randomized, double-blind, placebo-controlled studies with single oral doses. Clin Pharmacol Ther 2005; 78:675–688.
  54. Baggio LL, Drucker DJ, Maida A, Lamont BJ. ADA 2008: incretin-based therapeutics. MedscapeCME Web site. http://www.medscape.com/viewprogram/15786. Accessed September 18, 2009.
  55. US Department of Health and Human Services. FDA approves new drug treatment for type 2 diabetes. US Food and Drug Administration Web site. http://www.fda.gov/newsevents/newsroom/pressannouncements/ucm174780.htm. Published July 31, 2009. Accessed September 18, 2009.
  56. Covington P, Christopher R, Davenport M, et al. Pharmacokinetic, pharmacodynamic, and tolerability profiles of the dipeptidyl peptidase-4 inhibitor alogliptin: a randomized, double-blind, placebo-controlled, multiple-dose study in adult patients with type 2 diabetes. Clin Ther 2008; 30:499–512.
  57. DeFronzo RA, Fleck PR, Wilson CA, Mekki Q; on behalf of the Alogliptin Study 010 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor alogliptin in patients with type 2 diabetes and inadequate glycemic control: a randomized, double-blind, placebo-controlled study. Diabetes Care 2008; 31:2315–2317.
  58. Triplitt CL, McGill JB, Porte D Jr, Conner CS. The changing landscape of type 2 diabetes: the role of incretin-based therapies in managed care outcomes. J Manag Care Pharm 2007; 13(9 suppl C):S2–S16.
  59. Garber AJ, Spann SJ. An overview of incretin clinical trials. J Fam Pract 2008; 57(9 suppl):S10–S18.
  60. Henry RR. Evolving concepts of type 2 diabetes management with oral medications: new approaches to an old disease. Curr Med Res Opin 2008; 24:2189–2202.
  61. Centers for Disease Control and Prevention. National diabetes fact sheet: general information and national estimates on diabetes in the United States, 2007. Centers for Disease Control and Prevention Web site. http://www.cdc.gov/diabetes/pubs/pdf/ndfs_2007.pdf. Published 2008. Accessed September 21, 2009.
  62. Ong KL, Cheung BM, Wong LY, Wt NM, Tan KC, Lam KS. Prevalence, treatment, and control of diagnosed diabetes in the U.S. National Health and Nutrition Examination Survey 1999–2004. Ann Epidemiol 2008; 18:222–229.
  63. Sanders CL, Yesupriya AJ, Curtin LR. Analysis of population structure and stratification in NHANES III self-reported race/ethnicities. Centers for Disease Control and Prevention Web site. http://www.cdc.gov/genomics/events/file/print/10year/08_pop_struct_ab.pdf. Accessed September 21, 2009.
  64. Koro CE, Bowlin SJ, Bourgeois N, Fedder DO. Glycemic control from 1988 to 2000 among US adults diagnosed with type 2 diabetes: a preliminary report. Diabetes Care 2004; 27:17–20.
  65. Haffner SM, Lehto S, Rönnemaa T, Pyörälä K, Laakso M. Mortality from coronary heart disease in subjects with type 2 diabetes and in nondiabetic subjects with and without prior myocardial infarction. N Engl J Med 1998; 339:229–234.
  66. Lloyd-Jones D, Adams R, Carnethon M, et al. Heart disease and stroke statistics—2009 update: a report from the American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Circulation 2009; 119:480–486.
  67. Stonehouse AH, Holcombe JH, Kendall DM. GLP-1 analogues, DPP-IV inhibitors and the metabolic syndrome. In: Fonseca V, ed. Therapeutic Strategies in Metabolic Syndrome. Oxford, UK: Atlas Medical Publishing Ltd; 2008: 137–157.
  68. Purnell JQ, Weyer C. Weight effect of current and experimental drugs for diabetes mellitus: from promotion to alleviation of obesity. Treat Endocrinol 2003; 2:33–47.
  69. Klonoff DC, Buse JB, Nielsen LL, et al. Exenatide effects on diabetes, obesity, cardiovascular risk factors and hepatic biomarkers in patients with type 2 diabetes treated for at least 3 years. Curr Med Res Opin 2008; 24:275–286.
  70. Barnett AH, Burger J, Johns D, et al. Tolerability and efficacy of exenatide and titrated insulin glargine in adult patients with type 2 diabetes previously uncontrolled with metformin or a sulfonylurea: a multinational, randomized, open-label, two-period, crossover noninferiority trial. Clin Ther 2007; 29:2333–2348.
  71. Glass LC, Qu Y, Lenox S, et al. Effects of exenatide versus insulin analogues on weight change in subjects with type 2 diabetes: a pooled post-hoc analysis. Curr Med Res Opin 2008; 24:639–644.
  72. Heine RJ, Van Gaal LF, Johns D, Mihm MJ, Widel MH, Brodows RG; for the GWAA Study Group. Exenatide versus insulin glargine in patients with suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med 2005; 143:559–569.
  73. Nauck MA, Duran S, Kim D, et al. A comparison of twice-daily exenatide and biphasic insulin aspart in patients with type 2 diabetes who were suboptimally controlled with sulfonylurea and metformin: a non-inferiority study. Diabetologia 2007; 50:259–267.
  74. Ban K, Noyan-Ashraf MH, Hoefer J, Bolz SS, Drucker DJ, Husain M. Cardioprotective and vasodilatory actions of glucagon-like peptide 1 receptor are mediated through both glucagon-like peptide 1 receptor-dependent and -independent pathways. Circulation 2008; 117:2340–2350.
  75. Nikolaidis LA, Mankad S, Sokos GG, et al. Effects of glucagon-like peptide-1 in patients with acute myocardial infarction and left ventricular dysfunction after successful reperfusion. Circulation 2004; 109:962–965.
Article PDF
Author and Disclosure Information

Jeffrey S. Freeman, DO
Professor of Internal Medicine and Chairman, Division of Endocrinology and Metabolism, Philadelphia College of Osteopathic Medicine, Philadelphia, PA

Correspondence: Jeffrey S. Freeman, DO, Professor of Internal Medicine and Chairman, Division of Endocrinology and Metabolism, Philadelphia College of Osteopathic Medicine, 4190 City Ave., Suite 324, Philadelphia, PA 19131-1626; [email protected]

Dr. Freeman reported that he has received speakers’ bureau fees from Glaxo­SmithKline, Merck & Co., Inc., and Novo Nordisk Inc. He reported that he did not receive an honorarium for writing this article.

Dr. Freeman reported that he wrote this article and received no assistance with content development from unnamed contributors. He reported that BlueSpark Healthcare Communications, a medical communications company, assisted with preliminary literature searches, reference verification, proofing for grammar and style, table and figure rendering based on author instructions, copyright permission requests, and identification of topical overlap with other manuscripts in this supplement.

Publications
Page Number
S12-S19
Author and Disclosure Information

Jeffrey S. Freeman, DO
Professor of Internal Medicine and Chairman, Division of Endocrinology and Metabolism, Philadelphia College of Osteopathic Medicine, Philadelphia, PA

Correspondence: Jeffrey S. Freeman, DO, Professor of Internal Medicine and Chairman, Division of Endocrinology and Metabolism, Philadelphia College of Osteopathic Medicine, 4190 City Ave., Suite 324, Philadelphia, PA 19131-1626; [email protected]

Dr. Freeman reported that he has received speakers’ bureau fees from Glaxo­SmithKline, Merck & Co., Inc., and Novo Nordisk Inc. He reported that he did not receive an honorarium for writing this article.

Dr. Freeman reported that he wrote this article and received no assistance with content development from unnamed contributors. He reported that BlueSpark Healthcare Communications, a medical communications company, assisted with preliminary literature searches, reference verification, proofing for grammar and style, table and figure rendering based on author instructions, copyright permission requests, and identification of topical overlap with other manuscripts in this supplement.

Author and Disclosure Information

Jeffrey S. Freeman, DO
Professor of Internal Medicine and Chairman, Division of Endocrinology and Metabolism, Philadelphia College of Osteopathic Medicine, Philadelphia, PA

Correspondence: Jeffrey S. Freeman, DO, Professor of Internal Medicine and Chairman, Division of Endocrinology and Metabolism, Philadelphia College of Osteopathic Medicine, 4190 City Ave., Suite 324, Philadelphia, PA 19131-1626; [email protected]

Dr. Freeman reported that he has received speakers’ bureau fees from Glaxo­SmithKline, Merck & Co., Inc., and Novo Nordisk Inc. He reported that he did not receive an honorarium for writing this article.

Dr. Freeman reported that he wrote this article and received no assistance with content development from unnamed contributors. He reported that BlueSpark Healthcare Communications, a medical communications company, assisted with preliminary literature searches, reference verification, proofing for grammar and style, table and figure rendering based on author instructions, copyright permission requests, and identification of topical overlap with other manuscripts in this supplement.

Article PDF
Article PDF
Related Articles

It has long been understood that the pathophysiology of type 2 diabetes mellitus (T2DM) is based on the triad of progressive decline in insulin-producing pancreatic beta cells, an increase in insulin resistance, and increased hepatic glucose production.1,2 It is now evident that other factors, including defective actions of the gastrointestinal (GI) incretin hormones glucagon-like peptide–1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), also play significant roles.2–5 The uncontrolled hyperglycemia resulting from such defects may lead to microvascular complications, including retinopathy, neuropathy, microangiopathy, and nephropathy, and macrovascular complications, such as coronary artery disease and peripheral vascular disease.

This review explores the growing understanding of the role of the incretins in normal insulin secretion, as well as in the pathogenesis of T2DM, and examines the pathophysiologic basis for the benefits and therapeutic application of incretin-based therapies in T2DM.1,2

THE GI SYSTEM AND GLUCOSE HOMEOSTASIS IN THE HEALTHY STATE

The GI system plays an integral role in glucose homeostasis.6 The observation that orally administered glucose provides a stronger insulinotropic stimulus than an intravenous glucose challenge provided insight into the regulation of plasma glucose by the GI system of healthy individuals.7 The incretin effect, as this is termed, may be responsible for 50% to 70% of the total insulin secreted following oral glucose intake.8

Two GI peptide hormones (the incretins)—GLP-1 and GIP—were found to exert major glucoregulatory actions.3,9,10 Within minutes of nutrient ingestion, GLP-1 is secreted from intestinal L cells in the distal ileum and colon, while GIP is released by intestinal K cells in the duodenum and jejunum.3 GLP-1 and GIP trigger their insulinotropic actions by binding beta-cell receptors.3 GLP-1 receptors are expressed on pancreatic glucagon-containing alpha and delta cells as well as on beta cells, whereas GIP receptors are expressed primarily on beta cells.3,8 GLP-1 receptors are also expressed in the central nervous system (CNS), peripheral nervous system, lung, heart, and GI tract, while GIP receptors are expressed in adipose tissue and the CNS.3 GLP-1 inhibits glucose-dependent glucagon secretion from alpha cells.3 In healthy individuals, fasting glucose is managed by tonic insulin/glucagon secretion, but excursions of postprandial glucose (PPG) are controlled by insulin and the incretin hormones.11

Additionally, in animal studies, GLP-1 has been shown to induce the transcriptional activation of the insulin gene and insulin biosynthesis, thus increasing beta-cell proliferation and decreasing beta-cell apoptosis.12 GLP-1 stimulates a CNS-mediated pathway of insulin secretion, slows gastric emptying, increases CNS-mediated satiety leading to reduced food intake, indirectly increases insulin sensitivity and nutrient uptake in skeletal muscle and adipose tissue, and exerts neuroprotective effects.8

Reprinted, with permission, from European Journal of Endocrinology (Van Gaal LF, et al. Eur J Endocrinol 2008; 158:773–784),13 Copyright © 2008 European Society of Endocrinology.
Figure 1. Biologic actions of GIP and GLP-1 in relation to the pathophysiology of type 2 diabetes mellitus. GIP = glucose-dependent insulinotropic polypeptide, GLP-1 = glucagon-like peptide–1; PP = postprandial; solid arrows = potentially beneficial actions; dashed arrows = potentially harmful actions; slashed arrows = actions with no effect.
Along with its insulinotropic action, GIP has been shown in animal studies to inhibit gastric acid secretion, bioregulate fat metabolism in adipocytes, increase glucagon secretion and fat deposition, increase beta-cell replication, and decrease beta-cell apoptosis.8Figure 1 illustrates the biologic actions of GLP-1 and GIP.13

Both GLP-1 and GIP are rapidly degraded by the serine protease dipeptidyl peptidase–4 (DPP-4), which is widely expressed in bound and free forms.14 A recent study in healthy adults showed that GLP-1 concentration declined even during maximal DPP-4 inhibition, suggesting that there may be pathways of GLP-1 elimination other than DPP-4 enzymatic degradation.15

INCRETINS AND THE PATHOGENESIS OF T2DM

Studies have shown that incretin pathways play a role in the progression of T2DM.3,16 The significant reduction in the incretin effect seen in patients with T2DM has been attributed to several factors, including impaired secretion of GLP-1, accelerated metabolism of GLP-1 and GIP, and defective responsiveness to both hormones.16 Many patients with T2DM also have accelerated gastric emptying that may contribute to deterioration of their glycemic control.17

While GIP concentration is normal or modestly increased in patients with T2DM,16,18 the insulinotropic actions of GIP are significantly diminished.19 Thus, patients with T2DM have an impaired responsiveness to GIP with a possible link to GIP-receptor downregulation or desensitization.20

Are secretory defects a cause or result of T2DM?

In contrast to GIP, the secretion of GLP-1 has been shown to be deficient in patients with T2DM.18 As with GIP, it is unknown to what degree this defect is a cause or consequence of T2DM. In a study of identical twins, defective GLP-1 secretion was observed only in the one sibling with T2DM, suggesting that GLP-1 secretory deficits may be secondary to the development of T2DM.21 Despite the diminished secretion of GLP-1 in patients with T2DM, the insulinotropic actions of GLP-1 are preserved.19 It has also been shown that the effects of GLP-1 on gastric emptying and glucagon secretion are maintained in patients with T2DM.19,22,23

Whether this incretin dysregulation is responsible for or is the end result of hyperglycemia remains a subject of continued investigation. A recent study confirmed that the incretin effect is reduced in patients with T2DM, but advanced the concept that it may be a consequence of the diabetic state.16,24 Notably, impaired actions of GLP-1 and GIP and diminished concentrations of GLP-1 may be partially restored by improved glycemic control.24

Recent preclinical and clinical studies continue to clarify the roles of incretin hormones in T2DM. The findings from a study of obese diabetic mice suggest that the effect of GLP-1 therapy on the long-term remission of diabetes may be caused by improvements in beta-cell function and insulin sensitivity, as well as by a reduction in gluconeogenesis in the liver.25

Incretin effect and glucose tolerance, body mass index

Another study was conducted to evaluate quantitatively the separate impacts of obesity and hyperglycemia on the incretin effect in patients with T2DM, patients with impaired glucose tolerance, and patients with normal glucose tolerance.26 There was a significant (P ≤ .05) reduction in the incretin effect in terms of total insulin secretion, beta-cell glucose sensitivity, and the GLP-1 response to oral glucose in patients with T2DM compared with individuals whose glucose tolerance was normal or impaired. Each manifestation of the incretin effect was inversely related to both glucose tolerance and body mass index in an independent, additive manner (P ≤ .05); thus, glucose tolerance and obesity attenuate the incretin effect on beta-cell function and GLP-1 response independently of each other.

Exogenous GLP-1 has been shown to restore the regulation of blood glucose to near-normal concentrations in patients with T2DM.27 Several studies of patients with T2DM have shown that synthetic GLP-1 administration induces insulin secretion,19,27 slows gastric emptying (which is accelerated in patients with T2DM), and decreases inappropriately elevated glucagon secretion.19,23,28 Acute GLP-1 infusion studies showed that GLP-1 improved fasting plasma glucose (FPG) and PPG concentrations23,27; long-term studies showed that this hormone exerts euglycemic effects, leading to improvements in glycosylated hemoglobin (HbA1c), and induces weight loss.29

 

 

TARGETING FUNDAMENTAL DEFECTS OF T2DM WITH INCRETIN-BASED THERAPIES

Recognition and a better understanding of the role of the incretins and the enzyme involved in their degradation have led to the development of two incretin-based treatments: the GLP-1 receptor agonists, which possess many of the glucoregulatory actions of incretin peptides, and the DPP-4 inhibitors.5 Both the GLP-1 receptor agonists and the DPP-4 inhibitors have demonstrated safety and efficacy in the management of hyperglycemia in patients with T2DM.

GLP-1 receptor agonists

The GLP-1 receptor agonist exenatide is a synthetic form of exendin-4 and has a unique amino acid sequence that renders it resistant to degradation by DPP-4, making its actions longer lasting than endogenous GLP-1.5,30 Exenatide has a half-life of 2.4 hours and is detectable for up to 10 hours after subcutaneous (SC) injection.5,30 It is administered BID and has been approved as monotherapy or an adjunct therapy in patients with T2DM who have inadequate glycemic control following treatment with metformin, a sulfonylurea, a thiazolidinedione (TZD), or metformin in combination with a sulfonylurea or a TZD.31–35

In both human and animal studies, exenatide has been shown to enhance glucose-dependent insulin secretion and suppress inappropriate glucagon secretion in a glucose-dependent manner, reduce food intake and body weight, and acutely improve beta-cell function by enhancing first- and second-phase insulin secretion.5,36,37

In a small study involving 17 patients with T2DM, exenatide was shown to slow gastric emptying, which could be an important mechanism contributing to its beneficial effects on PPG concentration.38 Exenatide also has been shown to attenuate postprandial hyper­glycemia, a risk factor for cardiovascular disease (CVD),  by reducing endogenous glucose production by about 50% in patients with T2DM.39 Another mechanism for glycemic control may exist, as a recent animal study has shown that exenatide, similar to endogenous GLP-1, lowers blood glucose concentration independent of changes in pancreatic islet hormone secretion or delayed gastric emptying.40

A formulation of exenatide that is administered once weekly—exenatide long-acting release (LAR)—is in clinical evaluation and under review by the US Food and Drug Administration (FDA). In a short-term study, exenatide-LAR (0.8 mg or 2.0 mg) was administered once weekly for 15 weeks to patients with T2DM whose glycemia was suboptimally controlled with metformin alone or in combination with diet and exercise. Compared with placebo, treatment with exenatide once weekly was associated with markedly reduced HbA1c, FPG, PPG and body weight.41 In a larger, 30-week, phase 3 trial, Diabetes Therapy Utilization: Researching Changes in A1C, Weight and Other Factors Through Intervention with Exenatide ONce Weekly (DURATION-1), exenatide-LAR 2 mg once weekly was compared with exenatide 10 mg BID in patients with T2DM. Exenatide-LAR once weekly was associated with a significantly greater reduction in HbA1c (–1.9% vs –1.5%, P = .0023), and with a similar low risk of hypoglycemia and reduction in body weight (–3.7 kg vs –3.6 kg, P = .89) compared with the BID formulation.42

Liraglutide, recently approved in the European Union for T2DM and also under regulatory review in the United States, is a DPP-4–resistant human analogue GLP-1 receptor agonist in clinical development that has a 97% homology to native GLP-1.43–45 In contrast to exenatide, the acetylated liraglutide molecule allows binding to serum albumin and provides resistance to DPP-4 degradation, thus prolonging the half-life of liraglutide to approximately 12 hours. Liraglutide is administered SC QD as monotherapy or in combination with other antidiabetes agents such as metformin or sulfonylurea to patients with T2DM.44–47 Liraglutide has been shown to reduce HbA1c, decrease body weight, and lead to a lower incidence of hypoglycemia compared with the sulfonylurea glimepiride.

DPP-4 inhibitors

Sitagliptin is a DPP-4 inhibitor indicated as monotherapy or in combination with metformin or a TZD in patients with T2DM with inadequate glycemic control.48–51 Given orally, sitagliptin does not bind to the GLP-1 receptor agonist and has been shown to inhibit circulating DPP-4 activity by about 80%.52,53 Sitagliptin has been associated with an approximate twofold increase in postprandial GLP-1 plasma concentrations compared with placebo in healthy human subjects and in patients with T2DM.53 Saxagliptin, another potent DPP-4 inhibitor, significantly reduced HbA1c and FPG concentrations in patients with T2DM54 with a neutral effect on weight; it was recently approved by the FDA for treatment of T2DM.55

The DPP-4 inhibitor vildagliptin is currently being used in the European Union and Latin America but has yet to receive regulatory approval in the United States.54 Alogliptin, a novel, high-affinity, high-specificity DPP-4 inhibitor currently in development, provides rapid and sustained DPP-4 inhibition and significantly reduces HbA1c, FPG, and PPG concentrations with no change in body weight in patients with T2DM.56,57

Incretin-based therapies compared

In a recent head-to-head crossover trial between the GLP-1 receptor agonist exenatide and the DPP-4 inhibitor sitagliptin, exenatide had a greater effect in reducing 2-hour PPG.52 Patients with T2DM who switched from sitagliptin to exenatide showed a further reduction in 2-hour PPG concentration. Exenatide was also more potent than sitagliptin in increasing insulin secretion, reducing postprandial glucagon secretion, and decreasing triglycerides.52 Finally, exenatide slowed gastric emptying and reduced caloric intake. The differences between the two incretin-based therapies and their effects on glycemic control could be attributed to the pharmacologic concentration of the GLP-1 receptor agonist exenatide that is available for GLP-1 receptor activation compared with the twofold rise in endogenous GLP-1 concentration seen with the DPP-4 inhibitor sitagliptin.52

A comparison of the actions of the GLP-1 receptor agonists and DPP-4 inhibitors in patients with T2DM is provided in Table 1,52,58 and an overview of incretin-based therapies is presented in Table 2.45,54,59 GLP-1 receptor agonists induce weight loss in patients with T2DM, while DPP-4 inhibitors are weight neutral.3,52,58,60 The GLP-1 receptor agonists are associated with a much higher incidence of adverse GI effects such as nausea and vomiting, presumably also attributable to the pharmacologic levels achieved.

 

 

Effects of incretin-based therapies

The number of people with T2DM, overweight/obesity, or CVD, alone or in combination, is approaching epidemic proportions, with the mechanisms of these conditions interrelated. Approximately 24 million Americans have diabetes, and T2DM accounts for more than 90% of these cases.61 Most patients with T2DM are not achieving HbA1c targets.62–64 About 60% of deaths among patients with T2DM are caused by CVD.65 Compounding the problem, overweight/obesity enhances the risk for CV-related morbidities in patients with diabetes.66 A cluster of metabolic disorders referred to as the metabolic syndrome (which includes hyperglycemia, measures of central obesity, and a series of significant CV risk factors) is common in patients with T2DM and CVD.67 Unfortunately, many antidiabetes drugs that successfully manage glycemic control also cause weight gain, which in theory may increase CV risk in patients with T2DM.68

Data from studies of patients with T2DM show that exenatide improves glycemic control and reduces body weight. Exenatide administered BID significantly reduced HbA1c (–0.40% to –0.86%) and weight (–1.6 kg to –2.8 kg) relative to baseline in three 30-week, placebo-controlled clinical trials.31,33,34 In subsequent 2-year, open-label extension studies, exenatide produced significant reductions from baseline in HbA1c (–20.9% at 30 weeks) and weight (–2.1 kg at 30 weeks). Both decreases were sustained through 2 years (HbA1c –1.1%, weight –4.7 kg) with a low incidence of hypoglycemia.31 Further post hoc analysis of the open-label extension of the 30-week trials followed patients treated with exenatide BID for 3 years or longer.69 In addition to markedly decreasing HbA1c from baseline levels (–1.1% at 3 years and –0.8% at up to 3.5 years; P < .0001), adjunctive exenatide produced significant reductions in body weight—up to –5.3 kg after 3.5 years of therapy.31,69 At 3.5 years, continued exenatide therapy resulted in a –6% reduction in low-density lipoprotein cholesterol, a 24% mean increase in high-density lipoprotein cholesterol, and a mean reduction in blood pressure of –2% to –4% from baseline levels. Improvements in hepatic biomarkers and homeostasis model assessment-B, a measure of beta-cell function, were seen after 2 and 3 years of exenatide treatment.31 Hypoglycemia was generally mild and transient.

In comparative head-to-head studies, exenatide BID and insulin analogues reduced HbA1c by similar magnitudes; yet exenatide treatment resulted in better control in terms of PPG and weight loss, while insulin glargine and insulin aspart produced weight gain.70–73

Mechanisms of cardioprotective effects

Although the mechanisms for the potential cardioprotective effects of GLP-1 and its receptor agonists remain to be fully elucidated, a recent study suggested that two novel pathways could be involved—one that is dependent on the known GLP-1 receptor pathway, and one that is independent of the GLP-1 receptor pathway.74 Correlating with observations of a potential cardioprotective effect, an infusion of recombinant GLP-1 in patients with acute myocardial infarction, when added to standard therapy, resulted in improved left ventricular function and was associated with reduced mortality.75 Evidence continues to accumulate for potential cardioprotective effects of the GLP-1 receptor agonists, indicating that they may have a positive impact on macro­vascular complications in patients with T2DM.

CONCLUSION

T2DM, which is often associated with overweight and obesity, remains a significant challenge worldwide. The broad spectrum of glucoregulatory actions of the incretin hormones GLP-1 and GIP, and their importance in maintaining glucose homeostasis, have been recognized and correlated with the pathogenesis of T2DM. An improved understanding of the roles played by GLP-1 and GIP in the pathogenesis of T2DM may provide clinicians with important details regarding the therapeutic application of incretin-based therapies, including the GLP-1 receptor agonist exenatide and the DPP-4 inhibitors sitagliptin and saxagliptin. Antidiabetes agents whose development is based on the multiple pharmacologic effects of incretin hormones can address the multifaceted nature of T2DM and overcome some current limitations of traditional therapies, especially those related to weight. This becomes more compelling given the close link among T2DM, obesity, and increased CV risk.

It has long been understood that the pathophysiology of type 2 diabetes mellitus (T2DM) is based on the triad of progressive decline in insulin-producing pancreatic beta cells, an increase in insulin resistance, and increased hepatic glucose production.1,2 It is now evident that other factors, including defective actions of the gastrointestinal (GI) incretin hormones glucagon-like peptide–1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), also play significant roles.2–5 The uncontrolled hyperglycemia resulting from such defects may lead to microvascular complications, including retinopathy, neuropathy, microangiopathy, and nephropathy, and macrovascular complications, such as coronary artery disease and peripheral vascular disease.

This review explores the growing understanding of the role of the incretins in normal insulin secretion, as well as in the pathogenesis of T2DM, and examines the pathophysiologic basis for the benefits and therapeutic application of incretin-based therapies in T2DM.1,2

THE GI SYSTEM AND GLUCOSE HOMEOSTASIS IN THE HEALTHY STATE

The GI system plays an integral role in glucose homeostasis.6 The observation that orally administered glucose provides a stronger insulinotropic stimulus than an intravenous glucose challenge provided insight into the regulation of plasma glucose by the GI system of healthy individuals.7 The incretin effect, as this is termed, may be responsible for 50% to 70% of the total insulin secreted following oral glucose intake.8

Two GI peptide hormones (the incretins)—GLP-1 and GIP—were found to exert major glucoregulatory actions.3,9,10 Within minutes of nutrient ingestion, GLP-1 is secreted from intestinal L cells in the distal ileum and colon, while GIP is released by intestinal K cells in the duodenum and jejunum.3 GLP-1 and GIP trigger their insulinotropic actions by binding beta-cell receptors.3 GLP-1 receptors are expressed on pancreatic glucagon-containing alpha and delta cells as well as on beta cells, whereas GIP receptors are expressed primarily on beta cells.3,8 GLP-1 receptors are also expressed in the central nervous system (CNS), peripheral nervous system, lung, heart, and GI tract, while GIP receptors are expressed in adipose tissue and the CNS.3 GLP-1 inhibits glucose-dependent glucagon secretion from alpha cells.3 In healthy individuals, fasting glucose is managed by tonic insulin/glucagon secretion, but excursions of postprandial glucose (PPG) are controlled by insulin and the incretin hormones.11

Additionally, in animal studies, GLP-1 has been shown to induce the transcriptional activation of the insulin gene and insulin biosynthesis, thus increasing beta-cell proliferation and decreasing beta-cell apoptosis.12 GLP-1 stimulates a CNS-mediated pathway of insulin secretion, slows gastric emptying, increases CNS-mediated satiety leading to reduced food intake, indirectly increases insulin sensitivity and nutrient uptake in skeletal muscle and adipose tissue, and exerts neuroprotective effects.8

Reprinted, with permission, from European Journal of Endocrinology (Van Gaal LF, et al. Eur J Endocrinol 2008; 158:773–784),13 Copyright © 2008 European Society of Endocrinology.
Figure 1. Biologic actions of GIP and GLP-1 in relation to the pathophysiology of type 2 diabetes mellitus. GIP = glucose-dependent insulinotropic polypeptide, GLP-1 = glucagon-like peptide–1; PP = postprandial; solid arrows = potentially beneficial actions; dashed arrows = potentially harmful actions; slashed arrows = actions with no effect.
Along with its insulinotropic action, GIP has been shown in animal studies to inhibit gastric acid secretion, bioregulate fat metabolism in adipocytes, increase glucagon secretion and fat deposition, increase beta-cell replication, and decrease beta-cell apoptosis.8Figure 1 illustrates the biologic actions of GLP-1 and GIP.13

Both GLP-1 and GIP are rapidly degraded by the serine protease dipeptidyl peptidase–4 (DPP-4), which is widely expressed in bound and free forms.14 A recent study in healthy adults showed that GLP-1 concentration declined even during maximal DPP-4 inhibition, suggesting that there may be pathways of GLP-1 elimination other than DPP-4 enzymatic degradation.15

INCRETINS AND THE PATHOGENESIS OF T2DM

Studies have shown that incretin pathways play a role in the progression of T2DM.3,16 The significant reduction in the incretin effect seen in patients with T2DM has been attributed to several factors, including impaired secretion of GLP-1, accelerated metabolism of GLP-1 and GIP, and defective responsiveness to both hormones.16 Many patients with T2DM also have accelerated gastric emptying that may contribute to deterioration of their glycemic control.17

While GIP concentration is normal or modestly increased in patients with T2DM,16,18 the insulinotropic actions of GIP are significantly diminished.19 Thus, patients with T2DM have an impaired responsiveness to GIP with a possible link to GIP-receptor downregulation or desensitization.20

Are secretory defects a cause or result of T2DM?

In contrast to GIP, the secretion of GLP-1 has been shown to be deficient in patients with T2DM.18 As with GIP, it is unknown to what degree this defect is a cause or consequence of T2DM. In a study of identical twins, defective GLP-1 secretion was observed only in the one sibling with T2DM, suggesting that GLP-1 secretory deficits may be secondary to the development of T2DM.21 Despite the diminished secretion of GLP-1 in patients with T2DM, the insulinotropic actions of GLP-1 are preserved.19 It has also been shown that the effects of GLP-1 on gastric emptying and glucagon secretion are maintained in patients with T2DM.19,22,23

Whether this incretin dysregulation is responsible for or is the end result of hyperglycemia remains a subject of continued investigation. A recent study confirmed that the incretin effect is reduced in patients with T2DM, but advanced the concept that it may be a consequence of the diabetic state.16,24 Notably, impaired actions of GLP-1 and GIP and diminished concentrations of GLP-1 may be partially restored by improved glycemic control.24

Recent preclinical and clinical studies continue to clarify the roles of incretin hormones in T2DM. The findings from a study of obese diabetic mice suggest that the effect of GLP-1 therapy on the long-term remission of diabetes may be caused by improvements in beta-cell function and insulin sensitivity, as well as by a reduction in gluconeogenesis in the liver.25

Incretin effect and glucose tolerance, body mass index

Another study was conducted to evaluate quantitatively the separate impacts of obesity and hyperglycemia on the incretin effect in patients with T2DM, patients with impaired glucose tolerance, and patients with normal glucose tolerance.26 There was a significant (P ≤ .05) reduction in the incretin effect in terms of total insulin secretion, beta-cell glucose sensitivity, and the GLP-1 response to oral glucose in patients with T2DM compared with individuals whose glucose tolerance was normal or impaired. Each manifestation of the incretin effect was inversely related to both glucose tolerance and body mass index in an independent, additive manner (P ≤ .05); thus, glucose tolerance and obesity attenuate the incretin effect on beta-cell function and GLP-1 response independently of each other.

Exogenous GLP-1 has been shown to restore the regulation of blood glucose to near-normal concentrations in patients with T2DM.27 Several studies of patients with T2DM have shown that synthetic GLP-1 administration induces insulin secretion,19,27 slows gastric emptying (which is accelerated in patients with T2DM), and decreases inappropriately elevated glucagon secretion.19,23,28 Acute GLP-1 infusion studies showed that GLP-1 improved fasting plasma glucose (FPG) and PPG concentrations23,27; long-term studies showed that this hormone exerts euglycemic effects, leading to improvements in glycosylated hemoglobin (HbA1c), and induces weight loss.29

 

 

TARGETING FUNDAMENTAL DEFECTS OF T2DM WITH INCRETIN-BASED THERAPIES

Recognition and a better understanding of the role of the incretins and the enzyme involved in their degradation have led to the development of two incretin-based treatments: the GLP-1 receptor agonists, which possess many of the glucoregulatory actions of incretin peptides, and the DPP-4 inhibitors.5 Both the GLP-1 receptor agonists and the DPP-4 inhibitors have demonstrated safety and efficacy in the management of hyperglycemia in patients with T2DM.

GLP-1 receptor agonists

The GLP-1 receptor agonist exenatide is a synthetic form of exendin-4 and has a unique amino acid sequence that renders it resistant to degradation by DPP-4, making its actions longer lasting than endogenous GLP-1.5,30 Exenatide has a half-life of 2.4 hours and is detectable for up to 10 hours after subcutaneous (SC) injection.5,30 It is administered BID and has been approved as monotherapy or an adjunct therapy in patients with T2DM who have inadequate glycemic control following treatment with metformin, a sulfonylurea, a thiazolidinedione (TZD), or metformin in combination with a sulfonylurea or a TZD.31–35

In both human and animal studies, exenatide has been shown to enhance glucose-dependent insulin secretion and suppress inappropriate glucagon secretion in a glucose-dependent manner, reduce food intake and body weight, and acutely improve beta-cell function by enhancing first- and second-phase insulin secretion.5,36,37

In a small study involving 17 patients with T2DM, exenatide was shown to slow gastric emptying, which could be an important mechanism contributing to its beneficial effects on PPG concentration.38 Exenatide also has been shown to attenuate postprandial hyper­glycemia, a risk factor for cardiovascular disease (CVD),  by reducing endogenous glucose production by about 50% in patients with T2DM.39 Another mechanism for glycemic control may exist, as a recent animal study has shown that exenatide, similar to endogenous GLP-1, lowers blood glucose concentration independent of changes in pancreatic islet hormone secretion or delayed gastric emptying.40

A formulation of exenatide that is administered once weekly—exenatide long-acting release (LAR)—is in clinical evaluation and under review by the US Food and Drug Administration (FDA). In a short-term study, exenatide-LAR (0.8 mg or 2.0 mg) was administered once weekly for 15 weeks to patients with T2DM whose glycemia was suboptimally controlled with metformin alone or in combination with diet and exercise. Compared with placebo, treatment with exenatide once weekly was associated with markedly reduced HbA1c, FPG, PPG and body weight.41 In a larger, 30-week, phase 3 trial, Diabetes Therapy Utilization: Researching Changes in A1C, Weight and Other Factors Through Intervention with Exenatide ONce Weekly (DURATION-1), exenatide-LAR 2 mg once weekly was compared with exenatide 10 mg BID in patients with T2DM. Exenatide-LAR once weekly was associated with a significantly greater reduction in HbA1c (–1.9% vs –1.5%, P = .0023), and with a similar low risk of hypoglycemia and reduction in body weight (–3.7 kg vs –3.6 kg, P = .89) compared with the BID formulation.42

Liraglutide, recently approved in the European Union for T2DM and also under regulatory review in the United States, is a DPP-4–resistant human analogue GLP-1 receptor agonist in clinical development that has a 97% homology to native GLP-1.43–45 In contrast to exenatide, the acetylated liraglutide molecule allows binding to serum albumin and provides resistance to DPP-4 degradation, thus prolonging the half-life of liraglutide to approximately 12 hours. Liraglutide is administered SC QD as monotherapy or in combination with other antidiabetes agents such as metformin or sulfonylurea to patients with T2DM.44–47 Liraglutide has been shown to reduce HbA1c, decrease body weight, and lead to a lower incidence of hypoglycemia compared with the sulfonylurea glimepiride.

DPP-4 inhibitors

Sitagliptin is a DPP-4 inhibitor indicated as monotherapy or in combination with metformin or a TZD in patients with T2DM with inadequate glycemic control.48–51 Given orally, sitagliptin does not bind to the GLP-1 receptor agonist and has been shown to inhibit circulating DPP-4 activity by about 80%.52,53 Sitagliptin has been associated with an approximate twofold increase in postprandial GLP-1 plasma concentrations compared with placebo in healthy human subjects and in patients with T2DM.53 Saxagliptin, another potent DPP-4 inhibitor, significantly reduced HbA1c and FPG concentrations in patients with T2DM54 with a neutral effect on weight; it was recently approved by the FDA for treatment of T2DM.55

The DPP-4 inhibitor vildagliptin is currently being used in the European Union and Latin America but has yet to receive regulatory approval in the United States.54 Alogliptin, a novel, high-affinity, high-specificity DPP-4 inhibitor currently in development, provides rapid and sustained DPP-4 inhibition and significantly reduces HbA1c, FPG, and PPG concentrations with no change in body weight in patients with T2DM.56,57

Incretin-based therapies compared

In a recent head-to-head crossover trial between the GLP-1 receptor agonist exenatide and the DPP-4 inhibitor sitagliptin, exenatide had a greater effect in reducing 2-hour PPG.52 Patients with T2DM who switched from sitagliptin to exenatide showed a further reduction in 2-hour PPG concentration. Exenatide was also more potent than sitagliptin in increasing insulin secretion, reducing postprandial glucagon secretion, and decreasing triglycerides.52 Finally, exenatide slowed gastric emptying and reduced caloric intake. The differences between the two incretin-based therapies and their effects on glycemic control could be attributed to the pharmacologic concentration of the GLP-1 receptor agonist exenatide that is available for GLP-1 receptor activation compared with the twofold rise in endogenous GLP-1 concentration seen with the DPP-4 inhibitor sitagliptin.52

A comparison of the actions of the GLP-1 receptor agonists and DPP-4 inhibitors in patients with T2DM is provided in Table 1,52,58 and an overview of incretin-based therapies is presented in Table 2.45,54,59 GLP-1 receptor agonists induce weight loss in patients with T2DM, while DPP-4 inhibitors are weight neutral.3,52,58,60 The GLP-1 receptor agonists are associated with a much higher incidence of adverse GI effects such as nausea and vomiting, presumably also attributable to the pharmacologic levels achieved.

 

 

Effects of incretin-based therapies

The number of people with T2DM, overweight/obesity, or CVD, alone or in combination, is approaching epidemic proportions, with the mechanisms of these conditions interrelated. Approximately 24 million Americans have diabetes, and T2DM accounts for more than 90% of these cases.61 Most patients with T2DM are not achieving HbA1c targets.62–64 About 60% of deaths among patients with T2DM are caused by CVD.65 Compounding the problem, overweight/obesity enhances the risk for CV-related morbidities in patients with diabetes.66 A cluster of metabolic disorders referred to as the metabolic syndrome (which includes hyperglycemia, measures of central obesity, and a series of significant CV risk factors) is common in patients with T2DM and CVD.67 Unfortunately, many antidiabetes drugs that successfully manage glycemic control also cause weight gain, which in theory may increase CV risk in patients with T2DM.68

Data from studies of patients with T2DM show that exenatide improves glycemic control and reduces body weight. Exenatide administered BID significantly reduced HbA1c (–0.40% to –0.86%) and weight (–1.6 kg to –2.8 kg) relative to baseline in three 30-week, placebo-controlled clinical trials.31,33,34 In subsequent 2-year, open-label extension studies, exenatide produced significant reductions from baseline in HbA1c (–20.9% at 30 weeks) and weight (–2.1 kg at 30 weeks). Both decreases were sustained through 2 years (HbA1c –1.1%, weight –4.7 kg) with a low incidence of hypoglycemia.31 Further post hoc analysis of the open-label extension of the 30-week trials followed patients treated with exenatide BID for 3 years or longer.69 In addition to markedly decreasing HbA1c from baseline levels (–1.1% at 3 years and –0.8% at up to 3.5 years; P < .0001), adjunctive exenatide produced significant reductions in body weight—up to –5.3 kg after 3.5 years of therapy.31,69 At 3.5 years, continued exenatide therapy resulted in a –6% reduction in low-density lipoprotein cholesterol, a 24% mean increase in high-density lipoprotein cholesterol, and a mean reduction in blood pressure of –2% to –4% from baseline levels. Improvements in hepatic biomarkers and homeostasis model assessment-B, a measure of beta-cell function, were seen after 2 and 3 years of exenatide treatment.31 Hypoglycemia was generally mild and transient.

In comparative head-to-head studies, exenatide BID and insulin analogues reduced HbA1c by similar magnitudes; yet exenatide treatment resulted in better control in terms of PPG and weight loss, while insulin glargine and insulin aspart produced weight gain.70–73

Mechanisms of cardioprotective effects

Although the mechanisms for the potential cardioprotective effects of GLP-1 and its receptor agonists remain to be fully elucidated, a recent study suggested that two novel pathways could be involved—one that is dependent on the known GLP-1 receptor pathway, and one that is independent of the GLP-1 receptor pathway.74 Correlating with observations of a potential cardioprotective effect, an infusion of recombinant GLP-1 in patients with acute myocardial infarction, when added to standard therapy, resulted in improved left ventricular function and was associated with reduced mortality.75 Evidence continues to accumulate for potential cardioprotective effects of the GLP-1 receptor agonists, indicating that they may have a positive impact on macro­vascular complications in patients with T2DM.

CONCLUSION

T2DM, which is often associated with overweight and obesity, remains a significant challenge worldwide. The broad spectrum of glucoregulatory actions of the incretin hormones GLP-1 and GIP, and their importance in maintaining glucose homeostasis, have been recognized and correlated with the pathogenesis of T2DM. An improved understanding of the roles played by GLP-1 and GIP in the pathogenesis of T2DM may provide clinicians with important details regarding the therapeutic application of incretin-based therapies, including the GLP-1 receptor agonist exenatide and the DPP-4 inhibitors sitagliptin and saxagliptin. Antidiabetes agents whose development is based on the multiple pharmacologic effects of incretin hormones can address the multifaceted nature of T2DM and overcome some current limitations of traditional therapies, especially those related to weight. This becomes more compelling given the close link among T2DM, obesity, and increased CV risk.

References
  1. Boyle PJ, Freeman JS. Application of incretin mimetics and dipeptidyl peptidase IV inhibitors in managing type 2 diabetes mellitus. J Am Osteopath Assoc 2007; 107(suppl 3):S10–S16.
  2. Freeman JS. The pathophysiologic role of incretins. J Am Osteopath Assoc 2007; 107(suppl 3):S6–S9.
  3. Drucker DJ, Nauck MA. The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 2006; 368:1696–1705.
  4. Nauck MA, Baller B, Meier JJ. Gastric inhibitory polypeptide and glucagon-like peptide-1 in the pathogenesis of type 2 diabetes. Diabetes 2004; 53(suppl 3):S190–S196.
  5. Stonehouse A, Okerson T, Kendall D, Maggs D. Emerging incretin based therapies for type 2 diabetes: incretin mimetics and DPP-4 inhibitors. Curr Diabetes Rev 2008; 4:101–109.
  6. Huda MS, Wilding JP, Pinkney JH. Gut peptides and the regulation of appetite. Obes Rev 2006; 7:163–182.
  7. Elrick H, Stimmler L, Hlad CJ Jr, Arai Y. Plasma insulin response to oral and intravenous glucose administration. J Clin Endocrinol Metab 1964; 24:1076–1082.
  8. Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP. Gastroenterology 2007; 132:2131–2157.
  9. Brown JC, Dryburgh JR, Ross SA, Dupré J. Identification and actions of gastric inhibitory polypeptide. Recent Prog Horm Res 1975; 31:487–532.
  10. Kreymann B, Williams G, Ghatei MA, Bloom SR. Glucagon-like peptide-1 7-36: a physiological incretin in man. Lancet 1987; 2:1300–1304.
  11. Nauck MA, Homberger E, Siegel EG, et al. Incretin effects of increasing glucose loads in man calculated from venous insulin and C-peptide responses. J Clin Endocrinol Metab 1986; 63:492–498.
  12. Farilla L, Bulotta A, Hirshberg B, et al. Glucagon-like peptide 1 inhibits cell apoptosis and improves glucose responsiveness of freshly isolated human islets. Endocrinology 2003; 144:5149–5158.
  13. Van Gaal LF, Gutkin SW, Nauck MA. Exploiting the antidiabetic properties of incretins to treat type 2 diabetes mellitus: glucagon-like peptide 1 receptor agonists or insulin for patients with inadequate glycemic control? Eur J Endocrinol 2008; 158:773–784.
  14. Deacon CF, Nauck MA, Toft-Nielsen M, Pridal L, Willms B, Holst JJ. Both subcutaneously and intravenously administered glucagon-like peptide I are rapidly degraded from the NH2-terminus in type II diabetic patients and in healthy subjects. Diabetes 1995; 44:1126–1131.
  15. Dai H, Gustavson SM, Preston GM, Eskra JD, Calle R, Hirshberg B. Non-linear increase in GLP-1 levels in response to DPP-IV inhibition in healthy adult subjects. Diabetes Obes Metab 2008; 10:506–513.
  16. Nauck MA, Stöckmann F, Ebert R, Creutzfeldt W. Reduced incretin effect in type 2 (non-insulin-dependent) diabetes. Diabetologia 1986; 29:46–52.
  17. Phillips WT, Schwartz JG, McMahan CA. Rapid gastric emptying of an oral glucose solution in type 2 diabetic patients. J Nucl Med 1992; 33:1496–1500.
  18. Toft-Nielsen MB, Damholt MB, Madsbad S, et al. Determinants of the impaired secretion of glucagon-like peptide-1 in type 2 diabetic patients. J Clin Endocrinol Metab 2001; 86:3717–3723.
  19. Nauck MA, Heimesaat MM, Orskov C, Holst JJ, Ebert R, Creutzfeldt W. Preserved incretin activity of glucagon-like peptide 1 [7-36 amide] but not of synthetic human gastric inhibitory polypeptide in patients with type-2 diabetes mellitus. J Clin Invest 1993; 91:301–307.
  20. Lynn FC, Thompson SA, Pospisilik JA, et al. A novel pathway for regulation of glucose-dependent insulinotropic polypeptide (GIP) receptor expression in beta cells. FASEB J 2003; 17:91–93.
  21. Vaag AA, Holst JJ, Vølund A, Beck-Nielsen HB. Gut incretin hormones in identical twins discordant for non-insulin-dependent diabetes mellitus (NIDDM)—evidence for decreased glucagon-like peptide-1 secretion during oral glucose ingestion in NIDDM twins. Eur J Endocrinol 1996; 135:425–432.
  22. Meier JJ, Gallwitz B, Salmen S, et al. Normalization of glucose concentrations and deceleration of gastric emptying after solid meals during intravenous glucagon-like peptide 1 in patients with type 2 diabetes. J Clin Endocrinol Metab 2003; 88:2719–2725.
  23. Nauck MA, Kleine N, Orskov C, Holst JJ, Willms B, Creutzfeldt W. Normalization of fasting hyperglycaemia by exogenous glucagon-like peptide 1 (7-36 amide) in type 2 (non-insulin-dependent) diabetic patients. Diabetologia 1993; 36:741–744.
  24. Knop FK, Vilsbøll T, Højberg PV, et al. Reduced incretin effect in type 2 diabetes: cause or consequence of the diabetic state? Diabetes 2007; 56:1951–1959.
  25. Lee YS, Shin S, Shigihara T, et al. Glucagon-like peptide-1 gene therapy in obese diabetic mice results in long-term cure of diabetes by improving insulin sensitivity and reducing hepatic gluconeogenesis. Diabetes 2007; 56:1671–1679.
  26. Muscelli E, Mari A, Casolaro A, et al. Separate impact of obesity and glucose tolerance on the incretin effect in normal subjects and type 2 diabetic patients. Diabetes 2008; 57:1340–1348.
  27. Nathan DM, Schreiber E, Fogel H, Mojsov S, Habener JF. Insulinotropic action of glucagonlike peptide-I-(7-37) in diabetic and nondiabetic subjects. Diabetes Care 1992; 15:270–276.
  28. Kolterman OG, Buse JB, Fineman MS, et al. Synthetic exendin-4 (exenatide) significantly reduces postprandial and fasting plasma glucose in subjects with type 2 diabetes. J Clin Endocrinol Metab 2003; 88:3082–3089.
  29. Zander M, Madsbad S, Madsen JL, Holst JJ. Effect of 6-week course of glucagon-like peptide 1 on glycaemic control, insulin sensitivity, and beta-cell function in type 2 diabetes: a parallel-group study. Lancet 2002; 359:824–830.
  30. Kolterman OG, Kim DD, Shen L, et al. Pharmacokinetics, pharmacodynamics, and safety of exenatide in patients with type 2 diabetes mellitus. Am J Health Syst Pharm 2005; 62:173–181.
  31. Buse JB, Henry RR, Han J, et al. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in sulfonylurea-treated patients with type 2 diabetes. Diabetes Care 2004; 27:2628–2635.
  32. Byetta [package insert]. San Diego, CA: Amylin Pharmaceuticals, Inc.; 2009.
  33. DeFronzo RA, Ratner RE, Han J, Kim DD, Fineman MS, Baron AD. Effects of exenatide (exendin-4) on glycemic control and weight over 30 weeks in metformin-treated patients with type 2 diabetes. Diabetes Care 2005; 28:1092–1100.
  34. Kendall DM, Riddle MC, Rosenstock J, et al. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in patients with type 2 diabetes treated with metformin and a sulfonylurea. Diabetes Care 2005; 28:1083–1091.
  35. Zinman B, Hoogwerf BJ, Durán García S, et al. The effect of adding exenatide to a thiazolidinedione in suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med 2007; 146:477–485.
  36. Fehse F, Trautmann M, Holst JJ, et al. Exenatide augments first- and second-phase insulin secretion in response to intravenous glucose in subjects with type 2 diabetes. J Clin Endocrinol Metab 2005; 90:5991–5997.
  37. Parkes DG, Pittner R, Jodka C, Smith P, Young A. Insulinotropic actions of exendin-4 and glucagon-like peptide-1 in vivo and in vitro. Metabolism 2001; 50:583–589.
  38. Linnebjerg H, Park S, Kothare PA, et al. Effect of exenatide on gastric emptying and relationship to postprandial glycemia in type 2 diabetes. Regul Pept 2008; 151:123–129.
  39. Cervera A, Wajcberg E, Sriwijitkamol A, et al. Mechanism of action of exenatide to reduce postprandial hyperglycemia in type 2 diabetes. Am J Physiol Endocrinol Metab 2008; 294:E846–E852.
  40. Ionut V, Zheng D, Stefanovski D, Bergman RN. Exenatide can reduce glucose independent of islet hormones or gastric emptying. Am J Physiol Endocrinol Metab 2008; 295:E269–E277.
  41. Kim D, MacConell L, Zhuang D, et al. Effects of once-weekly dosing of a long-acting release formulation of exenatide on glucose control and body weight in subjects with type 2 diabetes. Diabetes Care 2007; 30:1487–1493.
  42. Drucker DJ, Buse JB, Taylor K, et al. Exenatide once weekly versus twice daily for the treatment of type 2 diabetes: a randomised, open-label, non-inferiority study. Lancet 2008; 372:1240–1250.
  43. Knudsen LB, Nielsen PF, Huusfeldt PO, et al. Potent derivatives of glucagon-like peptide-1 with pharmacokinetic properties suitable for once daily administration. J Med Chem 2000; 43:1664–1669.
  44. Nauck M, Frid A, Hermansen K, et al; for the LEAD-2 Study Group. Efficacy and safety comparison of liraglutide, glimepiride, and placebo, all in combination with metformin, in type 2 diabetes: the LEAD (liraglutide effect and action in diabetes)-2 study. Diabetes Care 2009; 32:84–90.
  45. Committee for Medicinal Products for Human Use: Summary of Positive Opinion for Victoza. European Medicines Agency Web site. http://www.emea.europa.eu/pdfs/human/opinion/Victoza_14168909en.pdf. Published April 23, 2009. Accessed September 21, 2009.
  46. Garber A, Henry R, Ratner R, et al. Liraglutide versus glimepiride monotherapy for type 2 diabetes (LEAD-3 Mono): a randomised, 52-week, phase III, double-blind, parallel-treatment trial. Lancet 2009; 373:473–481.
  47. Marre M, Shaw J, Brändle M, et al. Liraglutide, a once-daily human GLP-1 analogue, added to a sulphonylurea over 26 weeks produces greater improvements in glycaemic and weight control compared with adding rosiglitazone or placebo in subjects with type 2 diabetes (LEAD-1 SU). Diabet Med 2009; 26:268–278.
  48. Aschner P, Kipnes MS, Lunceford JK, et al; for the Sitagliptin 021 Study Group. Effect of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy on glycemic control in patients with type 2 diabetes. Diabetes Care 2006; 29:2632–2637.
  49. Charbonnel B, Karasik A, Liu J, Wu M, Meininger G; for the Sitagliptin Study 020 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin added to ongoing metformin therapy in patients with type 2 diabetes inadequately controlled with metformin alone. Diabetes Care 2006; 29:2638–2643.
  50. Raz I, Hanefeld M, Xu L, et al. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy in patients with type 2 diabetes mellitus. Diabetologia 2006; 49:2564–2571.
  51. Rosenstock J, Brazg R, Andryuk PJ, Lu K, Stein P; for the Sitagliptin Study 019 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin added to ongoing pioglitazone therapy in patients with type 2 diabetes: a 24-week, multicenter, randomized, double-blind, placebo-controlled, parallel-group study. Clin Ther 2006; 28:1556–1568.
  52. DeFronzo RA, Okerson T, Viswanathan P, Guan X, Holcombe JH, MacConell L. Effects of exenatide versus sitagliptin on postprandial glucose, insulin and glucagon secretion, gastric emptying, and caloric intake: a randomized, cross-over study. Curr Med Res Opin 2008; 24:2943–2952.
  53. Herman GA, Stevens C, Van Dyck K, et al. Pharmacokinetics and pharmacodynamics of sitagliptin, an inhibitor of dipeptidyl peptidase IV, in healthy subjects: results from two randomized, double-blind, placebo-controlled studies with single oral doses. Clin Pharmacol Ther 2005; 78:675–688.
  54. Baggio LL, Drucker DJ, Maida A, Lamont BJ. ADA 2008: incretin-based therapeutics. MedscapeCME Web site. http://www.medscape.com/viewprogram/15786. Accessed September 18, 2009.
  55. US Department of Health and Human Services. FDA approves new drug treatment for type 2 diabetes. US Food and Drug Administration Web site. http://www.fda.gov/newsevents/newsroom/pressannouncements/ucm174780.htm. Published July 31, 2009. Accessed September 18, 2009.
  56. Covington P, Christopher R, Davenport M, et al. Pharmacokinetic, pharmacodynamic, and tolerability profiles of the dipeptidyl peptidase-4 inhibitor alogliptin: a randomized, double-blind, placebo-controlled, multiple-dose study in adult patients with type 2 diabetes. Clin Ther 2008; 30:499–512.
  57. DeFronzo RA, Fleck PR, Wilson CA, Mekki Q; on behalf of the Alogliptin Study 010 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor alogliptin in patients with type 2 diabetes and inadequate glycemic control: a randomized, double-blind, placebo-controlled study. Diabetes Care 2008; 31:2315–2317.
  58. Triplitt CL, McGill JB, Porte D Jr, Conner CS. The changing landscape of type 2 diabetes: the role of incretin-based therapies in managed care outcomes. J Manag Care Pharm 2007; 13(9 suppl C):S2–S16.
  59. Garber AJ, Spann SJ. An overview of incretin clinical trials. J Fam Pract 2008; 57(9 suppl):S10–S18.
  60. Henry RR. Evolving concepts of type 2 diabetes management with oral medications: new approaches to an old disease. Curr Med Res Opin 2008; 24:2189–2202.
  61. Centers for Disease Control and Prevention. National diabetes fact sheet: general information and national estimates on diabetes in the United States, 2007. Centers for Disease Control and Prevention Web site. http://www.cdc.gov/diabetes/pubs/pdf/ndfs_2007.pdf. Published 2008. Accessed September 21, 2009.
  62. Ong KL, Cheung BM, Wong LY, Wt NM, Tan KC, Lam KS. Prevalence, treatment, and control of diagnosed diabetes in the U.S. National Health and Nutrition Examination Survey 1999–2004. Ann Epidemiol 2008; 18:222–229.
  63. Sanders CL, Yesupriya AJ, Curtin LR. Analysis of population structure and stratification in NHANES III self-reported race/ethnicities. Centers for Disease Control and Prevention Web site. http://www.cdc.gov/genomics/events/file/print/10year/08_pop_struct_ab.pdf. Accessed September 21, 2009.
  64. Koro CE, Bowlin SJ, Bourgeois N, Fedder DO. Glycemic control from 1988 to 2000 among US adults diagnosed with type 2 diabetes: a preliminary report. Diabetes Care 2004; 27:17–20.
  65. Haffner SM, Lehto S, Rönnemaa T, Pyörälä K, Laakso M. Mortality from coronary heart disease in subjects with type 2 diabetes and in nondiabetic subjects with and without prior myocardial infarction. N Engl J Med 1998; 339:229–234.
  66. Lloyd-Jones D, Adams R, Carnethon M, et al. Heart disease and stroke statistics—2009 update: a report from the American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Circulation 2009; 119:480–486.
  67. Stonehouse AH, Holcombe JH, Kendall DM. GLP-1 analogues, DPP-IV inhibitors and the metabolic syndrome. In: Fonseca V, ed. Therapeutic Strategies in Metabolic Syndrome. Oxford, UK: Atlas Medical Publishing Ltd; 2008: 137–157.
  68. Purnell JQ, Weyer C. Weight effect of current and experimental drugs for diabetes mellitus: from promotion to alleviation of obesity. Treat Endocrinol 2003; 2:33–47.
  69. Klonoff DC, Buse JB, Nielsen LL, et al. Exenatide effects on diabetes, obesity, cardiovascular risk factors and hepatic biomarkers in patients with type 2 diabetes treated for at least 3 years. Curr Med Res Opin 2008; 24:275–286.
  70. Barnett AH, Burger J, Johns D, et al. Tolerability and efficacy of exenatide and titrated insulin glargine in adult patients with type 2 diabetes previously uncontrolled with metformin or a sulfonylurea: a multinational, randomized, open-label, two-period, crossover noninferiority trial. Clin Ther 2007; 29:2333–2348.
  71. Glass LC, Qu Y, Lenox S, et al. Effects of exenatide versus insulin analogues on weight change in subjects with type 2 diabetes: a pooled post-hoc analysis. Curr Med Res Opin 2008; 24:639–644.
  72. Heine RJ, Van Gaal LF, Johns D, Mihm MJ, Widel MH, Brodows RG; for the GWAA Study Group. Exenatide versus insulin glargine in patients with suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med 2005; 143:559–569.
  73. Nauck MA, Duran S, Kim D, et al. A comparison of twice-daily exenatide and biphasic insulin aspart in patients with type 2 diabetes who were suboptimally controlled with sulfonylurea and metformin: a non-inferiority study. Diabetologia 2007; 50:259–267.
  74. Ban K, Noyan-Ashraf MH, Hoefer J, Bolz SS, Drucker DJ, Husain M. Cardioprotective and vasodilatory actions of glucagon-like peptide 1 receptor are mediated through both glucagon-like peptide 1 receptor-dependent and -independent pathways. Circulation 2008; 117:2340–2350.
  75. Nikolaidis LA, Mankad S, Sokos GG, et al. Effects of glucagon-like peptide-1 in patients with acute myocardial infarction and left ventricular dysfunction after successful reperfusion. Circulation 2004; 109:962–965.
References
  1. Boyle PJ, Freeman JS. Application of incretin mimetics and dipeptidyl peptidase IV inhibitors in managing type 2 diabetes mellitus. J Am Osteopath Assoc 2007; 107(suppl 3):S10–S16.
  2. Freeman JS. The pathophysiologic role of incretins. J Am Osteopath Assoc 2007; 107(suppl 3):S6–S9.
  3. Drucker DJ, Nauck MA. The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 2006; 368:1696–1705.
  4. Nauck MA, Baller B, Meier JJ. Gastric inhibitory polypeptide and glucagon-like peptide-1 in the pathogenesis of type 2 diabetes. Diabetes 2004; 53(suppl 3):S190–S196.
  5. Stonehouse A, Okerson T, Kendall D, Maggs D. Emerging incretin based therapies for type 2 diabetes: incretin mimetics and DPP-4 inhibitors. Curr Diabetes Rev 2008; 4:101–109.
  6. Huda MS, Wilding JP, Pinkney JH. Gut peptides and the regulation of appetite. Obes Rev 2006; 7:163–182.
  7. Elrick H, Stimmler L, Hlad CJ Jr, Arai Y. Plasma insulin response to oral and intravenous glucose administration. J Clin Endocrinol Metab 1964; 24:1076–1082.
  8. Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP. Gastroenterology 2007; 132:2131–2157.
  9. Brown JC, Dryburgh JR, Ross SA, Dupré J. Identification and actions of gastric inhibitory polypeptide. Recent Prog Horm Res 1975; 31:487–532.
  10. Kreymann B, Williams G, Ghatei MA, Bloom SR. Glucagon-like peptide-1 7-36: a physiological incretin in man. Lancet 1987; 2:1300–1304.
  11. Nauck MA, Homberger E, Siegel EG, et al. Incretin effects of increasing glucose loads in man calculated from venous insulin and C-peptide responses. J Clin Endocrinol Metab 1986; 63:492–498.
  12. Farilla L, Bulotta A, Hirshberg B, et al. Glucagon-like peptide 1 inhibits cell apoptosis and improves glucose responsiveness of freshly isolated human islets. Endocrinology 2003; 144:5149–5158.
  13. Van Gaal LF, Gutkin SW, Nauck MA. Exploiting the antidiabetic properties of incretins to treat type 2 diabetes mellitus: glucagon-like peptide 1 receptor agonists or insulin for patients with inadequate glycemic control? Eur J Endocrinol 2008; 158:773–784.
  14. Deacon CF, Nauck MA, Toft-Nielsen M, Pridal L, Willms B, Holst JJ. Both subcutaneously and intravenously administered glucagon-like peptide I are rapidly degraded from the NH2-terminus in type II diabetic patients and in healthy subjects. Diabetes 1995; 44:1126–1131.
  15. Dai H, Gustavson SM, Preston GM, Eskra JD, Calle R, Hirshberg B. Non-linear increase in GLP-1 levels in response to DPP-IV inhibition in healthy adult subjects. Diabetes Obes Metab 2008; 10:506–513.
  16. Nauck MA, Stöckmann F, Ebert R, Creutzfeldt W. Reduced incretin effect in type 2 (non-insulin-dependent) diabetes. Diabetologia 1986; 29:46–52.
  17. Phillips WT, Schwartz JG, McMahan CA. Rapid gastric emptying of an oral glucose solution in type 2 diabetic patients. J Nucl Med 1992; 33:1496–1500.
  18. Toft-Nielsen MB, Damholt MB, Madsbad S, et al. Determinants of the impaired secretion of glucagon-like peptide-1 in type 2 diabetic patients. J Clin Endocrinol Metab 2001; 86:3717–3723.
  19. Nauck MA, Heimesaat MM, Orskov C, Holst JJ, Ebert R, Creutzfeldt W. Preserved incretin activity of glucagon-like peptide 1 [7-36 amide] but not of synthetic human gastric inhibitory polypeptide in patients with type-2 diabetes mellitus. J Clin Invest 1993; 91:301–307.
  20. Lynn FC, Thompson SA, Pospisilik JA, et al. A novel pathway for regulation of glucose-dependent insulinotropic polypeptide (GIP) receptor expression in beta cells. FASEB J 2003; 17:91–93.
  21. Vaag AA, Holst JJ, Vølund A, Beck-Nielsen HB. Gut incretin hormones in identical twins discordant for non-insulin-dependent diabetes mellitus (NIDDM)—evidence for decreased glucagon-like peptide-1 secretion during oral glucose ingestion in NIDDM twins. Eur J Endocrinol 1996; 135:425–432.
  22. Meier JJ, Gallwitz B, Salmen S, et al. Normalization of glucose concentrations and deceleration of gastric emptying after solid meals during intravenous glucagon-like peptide 1 in patients with type 2 diabetes. J Clin Endocrinol Metab 2003; 88:2719–2725.
  23. Nauck MA, Kleine N, Orskov C, Holst JJ, Willms B, Creutzfeldt W. Normalization of fasting hyperglycaemia by exogenous glucagon-like peptide 1 (7-36 amide) in type 2 (non-insulin-dependent) diabetic patients. Diabetologia 1993; 36:741–744.
  24. Knop FK, Vilsbøll T, Højberg PV, et al. Reduced incretin effect in type 2 diabetes: cause or consequence of the diabetic state? Diabetes 2007; 56:1951–1959.
  25. Lee YS, Shin S, Shigihara T, et al. Glucagon-like peptide-1 gene therapy in obese diabetic mice results in long-term cure of diabetes by improving insulin sensitivity and reducing hepatic gluconeogenesis. Diabetes 2007; 56:1671–1679.
  26. Muscelli E, Mari A, Casolaro A, et al. Separate impact of obesity and glucose tolerance on the incretin effect in normal subjects and type 2 diabetic patients. Diabetes 2008; 57:1340–1348.
  27. Nathan DM, Schreiber E, Fogel H, Mojsov S, Habener JF. Insulinotropic action of glucagonlike peptide-I-(7-37) in diabetic and nondiabetic subjects. Diabetes Care 1992; 15:270–276.
  28. Kolterman OG, Buse JB, Fineman MS, et al. Synthetic exendin-4 (exenatide) significantly reduces postprandial and fasting plasma glucose in subjects with type 2 diabetes. J Clin Endocrinol Metab 2003; 88:3082–3089.
  29. Zander M, Madsbad S, Madsen JL, Holst JJ. Effect of 6-week course of glucagon-like peptide 1 on glycaemic control, insulin sensitivity, and beta-cell function in type 2 diabetes: a parallel-group study. Lancet 2002; 359:824–830.
  30. Kolterman OG, Kim DD, Shen L, et al. Pharmacokinetics, pharmacodynamics, and safety of exenatide in patients with type 2 diabetes mellitus. Am J Health Syst Pharm 2005; 62:173–181.
  31. Buse JB, Henry RR, Han J, et al. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in sulfonylurea-treated patients with type 2 diabetes. Diabetes Care 2004; 27:2628–2635.
  32. Byetta [package insert]. San Diego, CA: Amylin Pharmaceuticals, Inc.; 2009.
  33. DeFronzo RA, Ratner RE, Han J, Kim DD, Fineman MS, Baron AD. Effects of exenatide (exendin-4) on glycemic control and weight over 30 weeks in metformin-treated patients with type 2 diabetes. Diabetes Care 2005; 28:1092–1100.
  34. Kendall DM, Riddle MC, Rosenstock J, et al. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in patients with type 2 diabetes treated with metformin and a sulfonylurea. Diabetes Care 2005; 28:1083–1091.
  35. Zinman B, Hoogwerf BJ, Durán García S, et al. The effect of adding exenatide to a thiazolidinedione in suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med 2007; 146:477–485.
  36. Fehse F, Trautmann M, Holst JJ, et al. Exenatide augments first- and second-phase insulin secretion in response to intravenous glucose in subjects with type 2 diabetes. J Clin Endocrinol Metab 2005; 90:5991–5997.
  37. Parkes DG, Pittner R, Jodka C, Smith P, Young A. Insulinotropic actions of exendin-4 and glucagon-like peptide-1 in vivo and in vitro. Metabolism 2001; 50:583–589.
  38. Linnebjerg H, Park S, Kothare PA, et al. Effect of exenatide on gastric emptying and relationship to postprandial glycemia in type 2 diabetes. Regul Pept 2008; 151:123–129.
  39. Cervera A, Wajcberg E, Sriwijitkamol A, et al. Mechanism of action of exenatide to reduce postprandial hyperglycemia in type 2 diabetes. Am J Physiol Endocrinol Metab 2008; 294:E846–E852.
  40. Ionut V, Zheng D, Stefanovski D, Bergman RN. Exenatide can reduce glucose independent of islet hormones or gastric emptying. Am J Physiol Endocrinol Metab 2008; 295:E269–E277.
  41. Kim D, MacConell L, Zhuang D, et al. Effects of once-weekly dosing of a long-acting release formulation of exenatide on glucose control and body weight in subjects with type 2 diabetes. Diabetes Care 2007; 30:1487–1493.
  42. Drucker DJ, Buse JB, Taylor K, et al. Exenatide once weekly versus twice daily for the treatment of type 2 diabetes: a randomised, open-label, non-inferiority study. Lancet 2008; 372:1240–1250.
  43. Knudsen LB, Nielsen PF, Huusfeldt PO, et al. Potent derivatives of glucagon-like peptide-1 with pharmacokinetic properties suitable for once daily administration. J Med Chem 2000; 43:1664–1669.
  44. Nauck M, Frid A, Hermansen K, et al; for the LEAD-2 Study Group. Efficacy and safety comparison of liraglutide, glimepiride, and placebo, all in combination with metformin, in type 2 diabetes: the LEAD (liraglutide effect and action in diabetes)-2 study. Diabetes Care 2009; 32:84–90.
  45. Committee for Medicinal Products for Human Use: Summary of Positive Opinion for Victoza. European Medicines Agency Web site. http://www.emea.europa.eu/pdfs/human/opinion/Victoza_14168909en.pdf. Published April 23, 2009. Accessed September 21, 2009.
  46. Garber A, Henry R, Ratner R, et al. Liraglutide versus glimepiride monotherapy for type 2 diabetes (LEAD-3 Mono): a randomised, 52-week, phase III, double-blind, parallel-treatment trial. Lancet 2009; 373:473–481.
  47. Marre M, Shaw J, Brändle M, et al. Liraglutide, a once-daily human GLP-1 analogue, added to a sulphonylurea over 26 weeks produces greater improvements in glycaemic and weight control compared with adding rosiglitazone or placebo in subjects with type 2 diabetes (LEAD-1 SU). Diabet Med 2009; 26:268–278.
  48. Aschner P, Kipnes MS, Lunceford JK, et al; for the Sitagliptin 021 Study Group. Effect of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy on glycemic control in patients with type 2 diabetes. Diabetes Care 2006; 29:2632–2637.
  49. Charbonnel B, Karasik A, Liu J, Wu M, Meininger G; for the Sitagliptin Study 020 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin added to ongoing metformin therapy in patients with type 2 diabetes inadequately controlled with metformin alone. Diabetes Care 2006; 29:2638–2643.
  50. Raz I, Hanefeld M, Xu L, et al. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy in patients with type 2 diabetes mellitus. Diabetologia 2006; 49:2564–2571.
  51. Rosenstock J, Brazg R, Andryuk PJ, Lu K, Stein P; for the Sitagliptin Study 019 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin added to ongoing pioglitazone therapy in patients with type 2 diabetes: a 24-week, multicenter, randomized, double-blind, placebo-controlled, parallel-group study. Clin Ther 2006; 28:1556–1568.
  52. DeFronzo RA, Okerson T, Viswanathan P, Guan X, Holcombe JH, MacConell L. Effects of exenatide versus sitagliptin on postprandial glucose, insulin and glucagon secretion, gastric emptying, and caloric intake: a randomized, cross-over study. Curr Med Res Opin 2008; 24:2943–2952.
  53. Herman GA, Stevens C, Van Dyck K, et al. Pharmacokinetics and pharmacodynamics of sitagliptin, an inhibitor of dipeptidyl peptidase IV, in healthy subjects: results from two randomized, double-blind, placebo-controlled studies with single oral doses. Clin Pharmacol Ther 2005; 78:675–688.
  54. Baggio LL, Drucker DJ, Maida A, Lamont BJ. ADA 2008: incretin-based therapeutics. MedscapeCME Web site. http://www.medscape.com/viewprogram/15786. Accessed September 18, 2009.
  55. US Department of Health and Human Services. FDA approves new drug treatment for type 2 diabetes. US Food and Drug Administration Web site. http://www.fda.gov/newsevents/newsroom/pressannouncements/ucm174780.htm. Published July 31, 2009. Accessed September 18, 2009.
  56. Covington P, Christopher R, Davenport M, et al. Pharmacokinetic, pharmacodynamic, and tolerability profiles of the dipeptidyl peptidase-4 inhibitor alogliptin: a randomized, double-blind, placebo-controlled, multiple-dose study in adult patients with type 2 diabetes. Clin Ther 2008; 30:499–512.
  57. DeFronzo RA, Fleck PR, Wilson CA, Mekki Q; on behalf of the Alogliptin Study 010 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor alogliptin in patients with type 2 diabetes and inadequate glycemic control: a randomized, double-blind, placebo-controlled study. Diabetes Care 2008; 31:2315–2317.
  58. Triplitt CL, McGill JB, Porte D Jr, Conner CS. The changing landscape of type 2 diabetes: the role of incretin-based therapies in managed care outcomes. J Manag Care Pharm 2007; 13(9 suppl C):S2–S16.
  59. Garber AJ, Spann SJ. An overview of incretin clinical trials. J Fam Pract 2008; 57(9 suppl):S10–S18.
  60. Henry RR. Evolving concepts of type 2 diabetes management with oral medications: new approaches to an old disease. Curr Med Res Opin 2008; 24:2189–2202.
  61. Centers for Disease Control and Prevention. National diabetes fact sheet: general information and national estimates on diabetes in the United States, 2007. Centers for Disease Control and Prevention Web site. http://www.cdc.gov/diabetes/pubs/pdf/ndfs_2007.pdf. Published 2008. Accessed September 21, 2009.
  62. Ong KL, Cheung BM, Wong LY, Wt NM, Tan KC, Lam KS. Prevalence, treatment, and control of diagnosed diabetes in the U.S. National Health and Nutrition Examination Survey 1999–2004. Ann Epidemiol 2008; 18:222–229.
  63. Sanders CL, Yesupriya AJ, Curtin LR. Analysis of population structure and stratification in NHANES III self-reported race/ethnicities. Centers for Disease Control and Prevention Web site. http://www.cdc.gov/genomics/events/file/print/10year/08_pop_struct_ab.pdf. Accessed September 21, 2009.
  64. Koro CE, Bowlin SJ, Bourgeois N, Fedder DO. Glycemic control from 1988 to 2000 among US adults diagnosed with type 2 diabetes: a preliminary report. Diabetes Care 2004; 27:17–20.
  65. Haffner SM, Lehto S, Rönnemaa T, Pyörälä K, Laakso M. Mortality from coronary heart disease in subjects with type 2 diabetes and in nondiabetic subjects with and without prior myocardial infarction. N Engl J Med 1998; 339:229–234.
  66. Lloyd-Jones D, Adams R, Carnethon M, et al. Heart disease and stroke statistics—2009 update: a report from the American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Circulation 2009; 119:480–486.
  67. Stonehouse AH, Holcombe JH, Kendall DM. GLP-1 analogues, DPP-IV inhibitors and the metabolic syndrome. In: Fonseca V, ed. Therapeutic Strategies in Metabolic Syndrome. Oxford, UK: Atlas Medical Publishing Ltd; 2008: 137–157.
  68. Purnell JQ, Weyer C. Weight effect of current and experimental drugs for diabetes mellitus: from promotion to alleviation of obesity. Treat Endocrinol 2003; 2:33–47.
  69. Klonoff DC, Buse JB, Nielsen LL, et al. Exenatide effects on diabetes, obesity, cardiovascular risk factors and hepatic biomarkers in patients with type 2 diabetes treated for at least 3 years. Curr Med Res Opin 2008; 24:275–286.
  70. Barnett AH, Burger J, Johns D, et al. Tolerability and efficacy of exenatide and titrated insulin glargine in adult patients with type 2 diabetes previously uncontrolled with metformin or a sulfonylurea: a multinational, randomized, open-label, two-period, crossover noninferiority trial. Clin Ther 2007; 29:2333–2348.
  71. Glass LC, Qu Y, Lenox S, et al. Effects of exenatide versus insulin analogues on weight change in subjects with type 2 diabetes: a pooled post-hoc analysis. Curr Med Res Opin 2008; 24:639–644.
  72. Heine RJ, Van Gaal LF, Johns D, Mihm MJ, Widel MH, Brodows RG; for the GWAA Study Group. Exenatide versus insulin glargine in patients with suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med 2005; 143:559–569.
  73. Nauck MA, Duran S, Kim D, et al. A comparison of twice-daily exenatide and biphasic insulin aspart in patients with type 2 diabetes who were suboptimally controlled with sulfonylurea and metformin: a non-inferiority study. Diabetologia 2007; 50:259–267.
  74. Ban K, Noyan-Ashraf MH, Hoefer J, Bolz SS, Drucker DJ, Husain M. Cardioprotective and vasodilatory actions of glucagon-like peptide 1 receptor are mediated through both glucagon-like peptide 1 receptor-dependent and -independent pathways. Circulation 2008; 117:2340–2350.
  75. Nikolaidis LA, Mankad S, Sokos GG, et al. Effects of glucagon-like peptide-1 in patients with acute myocardial infarction and left ventricular dysfunction after successful reperfusion. Circulation 2004; 109:962–965.
Page Number
S12-S19
Page Number
S12-S19
Publications
Publications
Article Type
Display Headline
Role of the incretin pathway in the pathogenesis of type 2 diabetes mellitus
Display Headline
Role of the incretin pathway in the pathogenesis of type 2 diabetes mellitus
Citation Override
Cleveland Clinic Journal of Medicine 2009 December;76(suppl 5):S12-S19
Inside the Article

KEY POINTS

  • The incretin effect may be responsible for up to 70% of insulin secretion following oral glucose ingestion; reduction of the incretin effect contributes to T2DM pathophysiology.
  • It is unknown whether incretin defects are a cause or consequence of T2DM.
  • Incretin therapies effectively lower glucose with concomitant favorable effects on body weight. GLP-1 receptor agonists reduce weight, while DPP-4 inhibitors are weight neutral.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Fri, 04/13/2018 - 08:30
Un-Gate On Date
Fri, 04/13/2018 - 08:30
Use ProPublica
CFC Schedule Remove Status
Fri, 04/13/2018 - 08:30
Article PDF Media

Patient and treatment perspectives: Revisiting the link between type 2 diabetes, weight gain, and cardiovascular risk

Article Type
Changed
Tue, 10/02/2018 - 11:45
Display Headline
Patient and treatment perspectives: Revisiting the link between type 2 diabetes, weight gain, and cardiovascular risk

Type 2 diabetes mellitus (T2DM), excess weight, and obesity are increasing in prevalence at alarming rates.1–3 Concurrent with the increased prevalence is increased risk of morbidity and mortality. A healthy diet and exercise in conjunction with antidiabetes medications can help lower glucose concentration in patients with T2DM. Because these patients are at increased risk of cardiovascular (CV) morbidity and mortality, however, treatment strategies should address the CV risk factors, including blood pressure (BP), lipids, and body weight, as well as glycemic aspects of the disease.

To help clinicians manage the complex issues in treating patients with T2DM, this article presents an overview of patient and treatment perspectives relevant to overweight/obesity and CV disease (CVD). It includes an examination of the latest guidelines and algorithms for the management of T2DM, which continue to be updated and modified.

T2DM, WEIGHT GAIN OR OBESITY, AND CV RISK: A CHALLENGING TRIAD

Despite therapeutic advances in the diagnosis and treatment of diabetes and CVD over the last decade, the estimated number of persons in the United States older than 35 years with self-reported diabetes (with T2DM accounting for 90% to 95% of diagnosed cases) and CVD has increased from 4.2 million in 1997 to 5.7 million in 2005.3,4 The CV risk for patients with T2DM who have not had a CV event such as a myocardial infarction (MI) is similar to that of individuals without diabetes who have had a prior MI.5 Patients with T2DM have nearly double the mortality of those without the disease.6 Adding to their risk, about 80% of patients with T2DM are overweight or obese, conditions associated with worsened insulin resistance and increased CV risk and disease burden.7,8 Even a modest weight gain (5 kg) may increase the risk of coronary heart disease (CHD) by 30%, while associated changes in lipids and BP can increase the risk by another 20%.9

It is as important to control CV risk factors as it is to control glycemia in patients with T2DM, and both are difficult to achieve. Data from a recent nationwide Norwegian survey showed that only 13% of patients with T2DM achieved study-defined target levels; ie, glycosylated hemoglobin (HbA1c) less than 7.5%, BP less than 140/85 mm Hg, and total cholesterol/high-density lipoprotein (HDL-C) ratio less than 4.0.10

BENEFITS OF MANAGING GLYCEMIA, WEIGHT REDUCTION, AND CV RISK FACTORS

Several large studies, many ongoing, are generating data on the relationships among glycemia, weight reduction, and CV risk. It is well established that individuals with T2DM need aggressive risk factor reduction (glucose control, blood pressure management, and treatment of dyslipidemia) to optimize outcomes. However, characterization of the benefits of various components of risk factor reduction, particularly over many years, is only now occurring.

Results from the United Kingdom Prospective Diabetes Studies (UKPDS) showed the benefits and risks of pharmacologic glycemic control—essentially monotherapy with insulin or a sulfonylurea—compared with conventional dietary therapy in reducing diabetic complications in patients with newly diagnosed T2DM. In UKPDS 33, both insulin and sulfonylureas (intensive treatment) reduced the risk of microvascular end points (retinopathy, nephropathy) in patients whose median HbA1c was lowered to 7.0% at 10 years of follow-up, compared with patients who reached an HbA1c of 7.9%. However, intensive glycemic control did not translate into a statistically significant reduction in macrovascular complications, including MI, stroke, CVD, and death. Additionally, patients assigned to insulin had greater weight gain (+4.0 kg) than did patients assigned to receive the sulfonylurea chlorpropamide (+2.6 kg) or glyburide (+1.7 kg) (P < .01).11

The UKPDS showed that intensive treatment with metformin reduced the risk of T2DM-related end points compared with conventional treatment (primarily diet alone) in overweight patients.12 Although there were fewer patients in the metformin-treated subset (n = 342) than in the conventional treatment cohort, a secondary analysis showed that metformin was associated with less weight gain and fewer hypoglycemic episodes than either insulin or sulfonylurea therapy.12 Since HbA1c levels in the treatment groups were equal, the additional benefits seen with metformin in overweight patients with T2DM were not based solely on glycemic control.

The Action to Control Cardiovascular Risk in Diabetes (ACCORD) trial involved 10,000 individuals with T2DM. The primary outcome measure was a composite of CV events. The intensively treated group was controlled to a target HbA1c of less than 6.0%, with most patients receiving insulin. The trial was terminated early because an increased risk of sudden death was observed.13 A similar study, Action in Diabetes and Vascular Disease: Preterax and Diamicron Modified Release Controlled Evaluation (ADVANCE), evaluated more than 11,000 patients with T2DM, starting with a sulfonylurea-based regimen. In this study, there was no reduction in macrovascular events, but there was a reduction in nephropathy in the intensively treated group.14 In both studies, hypoglycemia and weight gain were more frequent in intensively treated patients; and in ACCORD, there were more episodes of severe hypoglycemia in the intensive-treatment group.13,14

The Veterans Affairs Diabetes Trial (VADT) evaluated the effect of intensive glucose control on CVD in 1,791 patients (mean age, 60 years) with poorly controlled T2DM (average duration, 11.5 years). The primary end points included MI, stroke, new or worsening congestive heart failure (CHF), limb amputation, and invasive intervention for coronary or peripheral arterial disease. The hazard ratio for these end points in the intensive-treatment group was 0.88 (95% confidence interval [CI], 0.74 to 1.05).15,16 Specifically, the following beneficial effects were achieved:

  • HbA1c reduced by –1.0% to –2.5% in absolute units,
  • systolic BP (SBP) reduced by –4 to –7 mm Hg,
  • diastolic BP (DBP) reduced by –7 to –8 mm Hg,
  • low-density lipoprotein cholesterol (LDL-C) reduced by –27 to –28 mg/dL,
  • triglycerides reduced by –44 to –50 mg/dL, and
  • HDL-C increased by 4 to 5 mg/dL.

Despite these benefits, body weight increased approximately 9 to 18 lb (4 to 8 kg) during therapy.15

Since overweight and obesity are independent risk factors for CHD and CVD in patients with T2DM,17 weight management is an integral component in treatment. In the Action for Health in Diabetes (Look AHEAD) trial, an intensive exercise and weight-loss program resulted in clinically significant (P < .001) weight loss at 1 year in patients who had T2DM and a body mass index (BMI) greater than 25 kg2/m (> 27 kg2/m if receiving insulin).18 When compared with patients who received less structured, infrequent support and minimal education about diabetes, participants in the intensive program showed more weight loss, improved glucose control, decreased CV events, and reduced medicine use. The Look AHEAD trial is currently evaluating whether these improvements will continue to result in lower CV risk.

 

 

PATIENT ADHERENCE AND SATISFACTION

It is often challenging for patients with T2DM to adhere to their treatment regimens. The Diabetes Attitude, Wishes, and Needs (DAWN) study examined psychosocial barriers to self-care in patients with diabetes and found that while 78% of patients with T2DM adhered to their medications, only 39% achieved complete success in at least two-thirds of their self-care domains.19 A multicenter, randomized, clinical trial examined the correlates of treatment satisfaction, including body weight, on patients’ appraisal of treatment satisfaction with injectable insulin. The 14.5% of patients who experienced a reduction in BMI reported systematic improvement in treatment satisfaction.20 Similarly, a cross-sectionally designed study (n = 99) that analyzed the interrelation of adherence, BMI, and depression in adults with T2DM found that patients with higher BMI and poor adherence also had depression, which was mediated by lower self-efficacy perceptions and increased diabetes symptoms.21 The results from these studies show a clear relationship between adherence with treatment regimens and achievement of HbA1c goals.22

RECENT DEVELOPMENTS IN T2DM MANAGEMENT: STRATEGIES TO REDUCE CV RISK

Because excess weight and obesity are prominent features of T2DM, it is important to use an antidiabetes agent that does not induce unnecessary weight gain (particularly central weight gain, which is thought to be most atherogenic).23 Metformin, considered the first-line agent for treatment of T2DM, is generally weight neutral with a low level of hypoglycemia.24,25 Sulfonylureas, insulin, and thiazolidinediones (TZDs) are all associated with weight gain, although newer-analogue insulins may cause less weight gain than older agents. TZDs, especially pioglitazone, are associated with improvements in long-term beta-cell function and CV risk factors despite weight gain.26,27

The newer antidiabetes agents belong to the dipeptidyl peptidase–4 (DPP-4) inhibitor and the glucagon-like peptide–1 (GLP-1) receptor agonist therapeutic classes and have been shown to be either weight neutral (DPP-4 inhibitors) or to cause weight loss (GLP-1 receptor agonists).28

Figure 1. Actions of glucagon-like peptide–1 (GLP-1) in peripheral tissues. Most of the effects of GLP-1 are mediated by direct interaction with GLP-1 receptors on specific tissues. However, the actions of GLP-1 in liver, fat, and muscle most likely occur through indirect mechanisms.
Figure 1 illustrates the physiologic role of GLP-1,29 which induces glucose-dependent insulin secretion after food intake by binding to specific receptors on pancreatic beta cells, suppresses postprandial glucagon from pancreatic alpha cells, reduces postprandial plasma glucose (PPG) concentrations by delaying gastric emptying, and diminishes appetite.28 The diminished secretion of GLP-1 in T2DM30,31 has led to the development of two different treatment approaches.28 Since GLP-1 is rapidly degraded by DPP-4, GLP-1 receptor agonists have been developed to resist DPP-4 inactivation while exhibiting many of the actions of endogenous incretin hormones.28,29 DPP-4 inhibitors function as incretin enhancers by protecting endogenous GLP-1 and glucose-dependent insulinotropic peptide, another incretin, from enzymatic breakdown.31–33 Unlike the GLP-1 receptor agonists, which are administered subcutaneously (SC), DPP-4 inhibitors are administered orally.

Obesity and the incretin effect

Figure 2. Adipokine expression and secretion by adipose tissue in insulin-resistant, obese subjects.
A study in healthy subjects and patients with T2DM demonstrated that glucose tolerance and obesity independently impair the incretin effect, resulting in impaired insulin secretion and glucagon suppression.34 Obesity is considered a subclinical inflammatory condition that releases chemokines, leading to insulin resistance. Figure 2 illustrates the interaction between obesity, inflammation, and insulin resistance.35

Two recent studies showed that surgically induced weight loss enhances the physiologic “incretin effect.” In one study, obese individuals with T2DM whose weight loss was secondary to bariatric surgery combined with caloric restriction showed improved insulin sensitivity, improved carbohydrate metabolism, and elevated levels of adiponectin and GLP-1, all of which may reduce the incidence of T2DM.36 In the other study, bariatric surgery in morbidly obese individuals with T2DM improved insulin secretion and ameliorated insulin resistance.37

DPP-4 inhibitors

DPP-4 inhibitors such as sitagliptin and saxagliptin inhibit the enzymatic activity of DPP-4 and increase endogenous concentrations of GLP-1.28 Sitagliptin has been compared with placebo as monotherapy and has been studied in combination with other therapies.

In an 18-week study, sitagliptin monotherapy, 100 and 200 mg QD, significantly reduced HbA1c compared with placebo (placebo-subtracted HbA1c reduction, –0.60% and –0.48%, respectively) in patients with T2DM. Sitagliptin also significantly decreased fasting plasma glucose (FPG) concentration relative to placebo.38 Twelve weeks of sitagliptin monotherapy at dosages of 5, 12.5, 25, and 50 mg BID led to significant (P < .001) reductions in HbA1c compared with placebo. Sitagliptin also produced significant reductions in FPG and mean daily glucose concentrations across the doses studied.39 Similar results were reported in other 12-week studies: 50 mg BID and 100 mg QD sitagliptin monotherapy significantly (P < .05) reduced HbA1c –0.39% to –0.56% and FPG concentration –11.0 to –17.2 mg/dL compared with placebo40; sitagliptin 100 mg QD compared with placebo produced a least-squares mean change from baseline HbA1c of –0.65% versus 0.41% (P < .001) and FPG of –22.5 versus 9.4 mg/dL (P < .001).41

Sitagliptin also has been studied in combination with other therapies. After 24 weeks, sitagliptin combined with pioglitazone significantly reduced HbA1c by –0.70% and FPG by –17.7 mg/dL (P < .001 for both) compared with placebo.42 In another 24-week study, 100 mg sitagliptin QD significantly improved glycemic control and beta-cell function (P < .05 for both) in patients with T2DM who had inadequate glycemic control with glimepiride or glimepiride plus metformin.43

In addition to significantly reducing HbA1c, sitagliptin 100 and 200 mg QD produced only small differences in body weight relative to placebo: least-squares mean change from baseline for sitagliptin 100 mg was –0.7 kg (95% CI, –1.3 to –0.1) and for 200 mg was –0.6 kg (95% CI, –1.0 to –0.2); for placebo it was –0.2 kg (95% CI, –0.7 to 0.2).38 These findings were consistent with those from another 24-week monotherapy study where sitagliptin produced weight loss of up to –0.2 kg44 and a 30-week study of sitagliptin added to ongoing metformin therapy. In the latter study, both sitagliptin and placebo resulted in weight reductions of –0.5 kg.45

The effects of sitagliptin on lipids and BP have been reported in clinical studies in patients with and without T2DM. In one study of patients with T2DM, the addition of sitagliptin to metformin increased total cholesterol (+8.1 mg/dL), LDL-C (+9.2 mg/dL), and HDL-C (+1.8 mg/dL) but lowered triglyceride (–14.5 mg/dL) after 18 weeks of treatment (24-week data).46 Data from a small (n = 19) study in nondiabetic patients with mild to moderate hypertension showed that sitagliptin produced small reductions (–2 to –3 mm Hg) in 24-hour ambulatory BP measurements.47

Another DPP-4 inhibitor, saxagliptin, with efficacy similar to that described for sitagliptin, was recently approved by the US Food and Drug Administration (FDA) for treatment of T2DM.48

 

 

GLP-1 receptor agonists

Many of the GLP-1 receptor agonists developed or under development have glucoregulatory effects similar to GLP-1 but are resistant to degradation by DPP-4.28 Exenatide, an exendin-4 receptor agonist, has compared favorably with sitagliptin and with insulin analogues. Long-acting (once-weekly and once-daily) GLP-1 receptor agonists are under development.

In a 2-week, head-to-head study in metformin-treated patients with T2DM, exenatide had a greater effect than sitagliptin in lowering PPG and was more potent in increasing insulin secretion and reducing postprandial glucagon secretion. In contrast to sitagliptin, exenatide slowed gastric emptying and reduced caloric intake.49

In two studies of patients treated with exenatide, on a background of either metformin alone or metformin plus a sulfonylurea, patients who received metformin lost more weight (–1.6 to –2.8 kg; P ≤ .01) and experienced more significant decreases from baseline HbA1c (–0.4% to –0.8%; P < .002) at 30 weeks than did patients who received placebo.50,51 In a 16-week trial of exenatide in patients previously treated with a TZD with or without metformin, exenatide reduced HbA1c –0.98%, fasting blood glucose –1.69 mmol/L, and body weight –1.51 kg.52

When compared with insulin analogues, exenatide has been associated with weight loss (~ –3 kg) while the insulin analogues were associated with weight gain (~ +3 kg).53 After 26 weeks, body weight decreased –2.3 kg with exenatide and increased +1.8 kg with insulin glargine.54 Similar results were found in a crossover noninferiority trial, where the least-squares mean difference in weight change was significantly (P < .001) different (2.2 kg) between the treatments.55 When exenatide was compared with insulin aspart in an open-label, noninferiority trial, there was a between-group difference in weight of –5.4 kg after 52 weeks.32

Exenatide has also demonstrated these benefits in open-label extension studies. After 2 years, mean HbA1c reductions of –1.1% from baseline were sustained (P < .05), and weight loss of –4.7 kg was maintained (P < .001).56 After 82 weeks, similar HbA1c decreases (–1.1%) and weight loss (–4.4 kg) were exhibited.57 Even after 3 years, these benefits were maintained in patients who remained on the drug (HbA1c reduction from baseline, –1.0%; weight loss, –5.3 kg [P < .0001 for both]).58

Long-acting formulations of GLP-1 receptor agonists are in clinical development; two of these are once-weekly exenatide and once-daily liraglutide. Exenatide once weekly has the advantage of less frequent dosing and has elicited greater reductions in HbA1c than exenatide BID. After 15 weeks of once-weekly administration, the 0.8-mg formulation reduced HbA1c –1.4% and the 2-mg formulation reduced it –1.7% (P < .0001 for both compared with placebo). Body weight was lowered –3.8 kg (P < .05 compared with placebo) with the 2-mg formulation.59 Compared with exenatide BID, exenatide 2 mg once weekly showed greater reductions in HbA1c (–1.9% vs –1.5%; P = .0023) after 30 weeks of therapy.60 In a 1-year noncomparative trial, treatment with exenatide once weekly improved HbA1c (–2.0%) and weight (–4.1 kg), as well as BP and lipid profiles compared with baseline.61

Liraglutide, a once-daily human analogue GLP-1 receptor agonist, is under review by the FDA.28 In a 26-week study of patients with T2DM, liraglutide was associated with reductions in HbA1c (mean, –1.04%; P = 0.067 compared with insulin) and body weight (mean, –2.5 kg; P < .001 compared with insulin) at dosages of 0.6 to 1.8 mg/day SC. Liraglutide produced a decline in SBP from 0.6 to 3 mm Hg but was not associated with a decrease in DBP.62 In a 52-week study comparing liraglutide with glimepiride monotherapy, liraglutide 1.2 mg was associated with an HbA1c reduction of –0.84% (P = .0014) and the 1.8-mg dose with a reduction of –1.14% (P < .0001) compared with –0.51% for glimepiride. SBP decreased –0.7 mm Hg with glimepiride compared with –2.1 mm Hg for liraglutide 1.2 mg (P = .2912) and –3.6 mm Hg for liraglutide 1.8 mg (P < .0118). Mean DBP fell slightly but not significantly in all treatment groups.63 No effects on lipid parameters were reported in these two liraglutide studies.

The Liraglutide Effect and Action in Diabetes (LEAD-6) trial was undertaken to compare exenatide (10 mg BID SC) and liraglutide (1.8 mg/day SC) as add-on therapy to metformin, a sulfonylurea, or a combination of both in 464 patients with T2DM. After 26 weeks of treatment, liraglutide was associated with a significant reduction in HbA1c of –1.12%, compared with –0.79% with exenatide (P < .0001). Patients treated with liraglutide lost –3.2 kg while those on exenatide lost –2.9 kg. Among patients previously treated with metformin alone, there was a 1-kg difference in favor of liraglutide (P = NS).64

Safety profile

All of the drugs discussed have potential adverse effects. Metformin continues to have a black box warning for lactic acidosis.65 Sulfonylureas and insulin can cause hypoglycemia. TZDs can cause fluid retention and, in rare cases, CHF (for which these drugs also carry a black box warning).66,67 TZDs also increase the risk of distal fracture.66,67 The most common side effects of exenatide are gastrointestinal, but there have been reported cases of pancreatitis, some of which have been fatal.68,69 It has been difficult to prove whether exenatide increases the risk of pancreatitis, as patients with T2DM are already at an increased (three- to fourfold) risk for this condition compared with persons who do not have T2DM.69 Exenatide should not be used in patients with severe renal impairment or end-stage renal disease; it should be used with caution in patients who have undergone renal transplantation and in patients with moderate renal impairment.

The prescribing information for sitagliptin includes pancreatitis among the adverse reactions identified during the drug’s postapproval use.70 As with exenatide, it is not fully known whether a true association exists between the agent and pancreatitis. However, since pancreatitis can occur in this patient population, it is recommended that abdominal pain be fully evaluated to rule out pancreatitis. Continued postmarketing surveillance is important for all of these agents.

THE ROLE OF GUIDELINES

The American Association of Clinical Endocrinologists (AACE),26 the American Diabetes Association (ADA),71 and the ADA in conjunction with the European Association for the Study of Diabetes (EASD)24 have recently revised their recommendations for the management of patients with diabetes. The guidelines are unanimous in setting a glycemic goal (HbA1c < 7.0% for the ADA, HbA1c ≤ 6.5% for the AACE) and advocating individualized care for a treatment goal of HbA1c lower than 6.0% in patients who stand to benefit from near euglycemia without inducing severe hypoglycemia.24,26,71

CVD is the major cause of morbidity and mortality associated with T2DM and is a source of increasing concern.5 Accordingly, special consideration should be given to patients with coexisting CV risk factors, including hypertension and dyslipidemia. The ADA and the EASD advocate lifestyle modification to decrease body weight and the concurrent initiation of metformin as first-line therapy.24 If that strategy is insufficient, then two tiers of treatment guide the choice of next steps24:

  • Tier 1, in addition to metformin, includes the sulfonylureas and insulin. Although these are excellent glucose-lowering drugs, they are associated with weight gain, hypoglycemia, and no improvement in BP or lipid levels. They are relatively low in cost and have been used for many years. Their main drawback is evidence that despite their use, beta-cell failure continues unabated over time.
  • Tier 2 treatments include pioglitazone and the GLP-1 receptor agonist exenatide. Consideration may be given to the use of pioglitazone or exenatide when hypoglycemia is of concern, with exenatide being preferred when weight loss is a major objective and HbA1c is close to target (< 8.0%).24 Additionally, both the TZDs and exenatide probably help slow the rate of beta-cell failure, particularly if they are used early in the course of the disease.72,73 The AACE recommends different pharmacologic approaches based on HbA1c at diagnosis.26

The American Heart Association and the ADA have issued a joint scientific statement on the primary prevention of CVD in patients with diabetes.74 They advocate lifestyle management of body weight, nutrition, and physical activity.74 In addition, they stress the need for attention to BP, lipid levels, and smoking status, and the use of antiplatelet agents in patients at increased CV risk (> 40 years of age and a family history of CVD, hypertension, smoking, dyslipidemia, or albuminuria).

CONCLUSION

T2DM, weight gain/obesity, and CV risk present a continuing challenge to patients and clinicians. Anti­diabetes agents have varying degrees of evidence to support their effects on HbA1c, body weight, BP, and lipid levels. A better understanding of the pathophysiology of T2DM has led to the development of newer antidiabetes agents that target the fundamental defects of the disease. Evidence continues to accumulate for the improved benefits of glycemic control and weight loss in T2DM with GLP-1 receptor agonists such as exenatide currently having robust data in terms of beneficial effects on weight and CV risk factors. As clinicians continue to incorporate this knowledge into their practice patterns, patient adherence and clinical outcomes are expected to improve. Newer agents, such as incretin-based therapies, address T2DM as well as other factors that increase cardiometabolic risk through their effects not only on glycemic control but on body weight, BP, and lipids.

References
  1. Centers for Disease Control and Prevention (CDC). Prevalence of overweight and obesity among adults with diagnosed diabetes—United States, 1988–1994 and 1999–2002. MMWR Morb Mortal Wkly Rep 2004; 53:1066–1068.
  2. Prevalence of overweight and obesity among adults: United States 2003–2004. Centers for Disease Contral and Prevention Web site. http://www.cdc.gov/nchs/products/pubs/pubd/hestats/overweight/overwght_adult_03.htm. Published: April 2006. Accessed September 23, 2009.
  3. National Institute of Diabetes and Digestive and Kidney Diseases. National Diabetes Statistics, 2007 fact sheet. National Institutes of Health Web site. http://www.diabetes.niddk.nih.gov/dm/pubs/statistics/index.htm. Published 2008. Accessed September 16, 2009.
  4. Centers for Disease Control and Prevention (CDC). Prevalence of self-reported cardiovascular disease among persons aged >35 years with diabetes—United States, 1997–2005. MMWR Morb Mortal Wkly Rep 2007; 56:1129–1132.
  5. Haffner SM, Lehto S, Rönnemaa T, Pyörälä K, Laakso M. Mortality from coronary heart disease in subjects with type 2 diabetes and in nondiabetic subjects with and without prior myocardial infarction. N Engl J Med 1998; 339:229–234.
  6. Mulnier HE, Seaman HE, Raleigh VS, Soedamah-Muthu SS, Colhoun HM, Lawrenson RA. Mortality in people with type 2 diabetes in the UK. Diabet Med 2006; 23:516–521.
  7. Van Gaal LF, Gutkin SW, Nauck MA. Exploiting the antidiabetic properties of incretins to treat type 2 diabetes mellitus: glucagon-like peptide 1 receptor agonists or insulin for patients with inadequate glycemic control? Eur J Endocrinol 2008; 158:773–784.
  8. Bonora E, Targher G, Formentini G, et al. The metabolic syndrome is an independent predictor of cardiovascular disease in type 2 diabetic subjects: prospective data from the Verona Diabetes Complications Study. Diabet Med 2004; 21:52–58.
  9. Anderson JW, Kendall CW, Jenkins DJ. Importance of weight management in type 2 diabetes: review with meta-analysis of clinical studies. J Am Coll Nutr 2003; 22:331–339.
  10. Jenssen TG, Tonstad S, Claudi T, Midthejell K, Cooper J. The gap between guidelines and practice in the treatment of type 2 diabetes: a nationwide survey in Norway. Diabetes Res Clin Pract 2008; 80:314–320.
  11. UK Prospective Diabetes Study (UKPDS) Group. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). Lancet 1998; 352:837–853.
  12. UK Prospective Diabetes Study (UKPDS) Group. Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34). Lancet 1998; 352:854–865.
  13. Action to Control Cardiovascular Risk in Diabetes Study Group. Effects of intensive glucose lowering in type 2 diabetes. N Engl J Med 2008; 358:2545–2559.
  14. ADVANCE Collaborative Group. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N Engl J Med 2008; 358:2560–2572.
  15. Duckworth W, Abraira C, Moritz T, et al; for the VADT Investigators. Glucose control and vascular complications in veterans with type 2 diabetes. N Engl J Med 2009; 360:129–139.
  16. Skyler JS, Bergenstal R, Bonow RO, et al. Intensive glycemic control and the prevention of cardiovascular events: implications of the ACCORD, ADVANCE, and VA Diabetes Trials: a position statement of the American Diabetes Association and a scientific statement of the American College of Cardiology Foundation and the American Heart Association. Diabetes Care 2009; 32:187–192.
  17. Eeg-Olofsson K, Cederholm J, Nilsson PM, et al. Risk of cardiovascular disease and mortality in overweight and obese patients with type 2 diabetes: an observational study in 13,087 patients. Diabetologia 2009; 52:65–73.
  18. Look AHEAD Research Group. Reduction in weight and cardiovascular disease risk factors in individuals with type 2 diabetes: one-year results of the Look AHEAD trial. Diabetes Care 2007; 30:1374–1383.
  19. Peyrot M, Rubin RR, Lauritzen T, Snoek FJ, Matthews DR, Skovlund SE. Psychosocial problems and barriers to improved diabetes management: results of the Cross-National Diabetes Attitudes, Wishes and Needs (DAWN) Study. Diabet Med 2005; 22:1379–1385.
  20. Brod M, Cobden D, Lammert M, Bushnell D, Raskin P. Examining correlates of treatment satisfaction for injectable insulin in type 2 diabetes: lessons learned from a clinical trial comparing biphasic and basal analogues. Health Qual Life Outcomes 2007; 5:8.
  21. Sacco WP, Wells KJ, Friedman A, Matthew R, Perez S, Vaughan CA. Adherence, body mass index, and depression in adults with type 2 diabetes: the mediational role of diabetes symptoms and self-efficacy. Health Psychol 2007; 26:693–700.
  22. Ruelas V, Roybal GM, Lu Y, Goldman D, Peters A. Clinical and behavioral correlates of achieving and maintaining glycemic targets in an underserved population with type 2 diabetes. Diabetes Care 2009; 32:54–56.
  23. Nieves DJ, Cnop M, Retzlaff B, et al. The atherogenic lipoprotein profile associated with obesity and insulin resistance is largely attributable to intra-abdominal fat. Diabetes 2003; 52:172–179.
  24. Nathan DM, Buse JB, Davidson MB, et al. Medical management of hyperglycemia in type 2 diabetes: a consensus algorithm for the initiation and adjustment of therapy: a consensus statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care 2009; 32:193–203.
  25. Alexander GC, Sehgal NL, Moloney RM, Stafford RS. National trends in treatment of type 2 diabetes mellitus, 1994–2007. Arch Intern Med 2008; 168:2088–2094.
  26. AACE Diabetes Mellitus Clinical Practice Guidelines Task Force. American Association of Clinical Endocrinologists medical guidelines for clinical practice for the management of diabetes mellitus. Endocr Pract 2007; 13(suppl 1):S4–S68.
  27. Hermansen K, Mortensen LS. Bodyweight changes associated with antihyperglycaemic agents in type 2 diabetes mellitus. Drug Saf 2007; 30:1127–1142.
  28. Stonehouse A, Okerson T, Kendall D, Maggs D. Emerging incretin based therapies for type 2 diabetes: incretin mimetics and DPP-4 inhibitors. Curr Diabetes Rev 2008; 4:101–109.
  29. Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP. Gastroenterology 2007; 132:2131–2157.
  30. Nauck MA, Baller B, Meier JJ. Gastric inhibitory polypeptide and glucagon-like peptide-1 in the pathogenesis of type 2 diabetes. Diabetes 2004; 53(suppl 3):S190–S196.
  31. Toft-Nielsen MB, Madsbad S, Holst JJ. Determinants of the effectiveness of glucagon-like peptide-1 in type 2 diabetes. J Clin Endocrinol Metab 2001; 86:3853–3860.
  32. Nauck MA, Duran S, Kim D, et al. A comparison of twice-daily exenatide and biphasic insulin aspart in patients with type 2 diabetes who were suboptimally controlled with sulfonylurea and metformin: a non-inferiority study. Diabetologia 2007; 50:259–267.
  33. Nauck MA, Meininger G, Sheng D, Terranella L, Stein PP; for the Sitagliptin Study 024 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor, sitagliptin, compared with the sulfonylurea, glipizide, in patients with type 2 diabetes inadequately controlled on metformin alone: a randomized, double-blind, non-inferiority trial. Diabetes Obes Metab 2007; 9:194–205.
  34. Muscelli E, Mari A, Casolaro A, et al. Separate impact of obesity and glucose tolerance on the incretin effect in normal subjects and type 2 diabetic patients. Diabetes 2008; 57:1340–1348.
  35. Bastard JP, Maachi M, Lagathu C, et al. Recent advances in the relationship between obesity, inflammation, and insulin resistance. Eur Cytokine Netw 2006; 17:4–12.
  36. de Carvalho CP, Marin DM, de Souza AL, et al. GLP-1 and adiponectin: effect of weight loss after dietary restriction and gastric bypass in morbidly obese patients with normal and abnormal glucose metabolism. Obes Surg 2009; 19:313–320.
  37. Mingrone G. Role of the incretin system in the remission of type 2 diabetes following bariatric surgery. Nutr Metab Cardiovasc Dis 2008; 18:574–579.
  38. Raz I, Hanefeld M, Xu L, Caria C, Williams-Herman D, Khatami H; for the Sitagliptin Study 023 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy in patients with type 2 diabetes mellitus. Diabetologia 2006; 49:2564–2571.
  39. Scott R, Wu M, Sanchez M, Stein P. Efficacy and tolerability of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy over 12 weeks in patients with type 2 diabetes. Int J Clin Pract 2007; 61:171–180.
  40. Hanefeld M, Herman G, Wu M, Mickel C, Sanchez M, Stein PP; for the Sitagliptin Study 014 Investigators. Once-daily sitagliptin, a dipeptidyl peptidase-4 inhibitor, for the treatment of patients with type 2 diabetes. Curr Med Res Opin 2007; 23:1329–1339.
  41. Nonaka K, Kakikawa T, Sato A, et al. Efficacy and safety of sitagliptin monotherapy in Japanese patients with type 2 diabetes. Diabetes Res Clin Pract 2008; 79:291–298.
  42. Rosenstock J, Brazg R, Andryuk PJ, Lu K, Stein P; for the Sitagliptin Study 019 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin added to ongoing pioglitazone therapy in patients with type 2 diabetes: a 24-week, multicenter, randomized, double-blind, placebo-controlled, parallel-group study. Clin Ther 2006; 28:1556–1568.
  43. Hermansen K, Kipnes M, Luo E, Fanurik D, Khatami H, Stein P; for the Sitagliptin Study 035 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor, sitagliptin, in patients with type 2 diabetes mellitus inadequately controlled on glimepiride alone or on glimepiride and metformin. Diabetes Obes Metab 2007; 9:733–745.
  44. Aschner P, Kipnes MS, Lunceford JK, Sanchez M, Mickel C, Williams-Herman DE; for the Sitagliptin Study 021 Group. Effect of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy on glycemic control in patients with type 2 diabetes. Diabetes Care 2006; 29:2632–2637.
  45. Raz I, Chen Y, Wu M, et al. Efficacy and safety of sitagliptin added to ongoing metformin therapy in patients with type 2 diabetes. Curr Med Res Opin 2008; 24:537–550.
  46. Scott R, Loeys T, Davies MJ, Engel SS; for the Sitagliptin Study 801 Group. Efficacy and safety of sitagliptin when added to ongoing metformin therapy in patients with type 2 diabetes. Diabetes Obes Metab 2008; 10:959–969.
  47. Mistry GC, Maes AL, Lasseter KC, et al. Effect of sitagliptin, a dipeptidyl peptidase-4 inhibitor, on blood pressure in nondiabetic patients with mild to moderate hypertension. J Clin Pharmacol 2008; 48:592–598.
  48. US Department of Health and Human Services. FDA approves new drug treatment for type 2 diabetes. US Food and Drug Administration Web site. http://www.fda.gov/newsevents/newsroom/pressannouncements/ucm174780.htm. Published July 31, 2009. Accessed September 18, 2009.
  49. DeFronzo RA, Okerson T, Viswanathan P, Guan X, Holcombe JH, MacConell L. Effects of exenatide versus sitagliptin on postprandial glucose, insulin and glucagon secretion, gastric emptying, and caloric intake: a randomized, cross-over study. Curr Med Res Opin 2008; 24:2943–2952.
  50. DeFronzo RA, Ratner RE, Han J, Kim DD, Fineman MS, Baron AD. Effects of exenatide (exendin-4) on glycemic control and weight over 30 weeks in metformin-treated patients with type 2 diabetes. Diabetes Care 2005; 28:1092–1100.
  51. Kendall DM, Riddle MC, Rosenstock J, et al. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in patients with type 2 diabetes treated with metformin and a sulfonylurea. Diabetes Care 2005; 28:1083–1091.
  52. Zinman B, Hoogwerf BJ, Durán García S, et al. The effect of adding exenatide to a thiazolidinedione in suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med 2007; 146:477–485.
  53. Glass LC, Qu Y, Lenox S, et al. Effects of exenatide versus insulin analogues on weight change in subjects with type 2 diabetes: a pooled post-hoc analysis. Curr Med Res Opin 2008; 24:639–644.
  54. Heine RJ, Van Gaal LF, Johns D, Mihm MJ, Widel MH, Brodows RG; for the GWAA Study Group. Exenatide versus insulin glargine in patients with suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med 2005; 143:559–569.
  55. Barnett AH, Burger J, Johns D, et al. Tolerability and efficacy of exenatide and titrated insulin glargine in adult patients with type 2 diabetes previously uncontrolled with metformin or a sulfonylurea: a multinational, randomized, open-label, two-period, crossover noninferiority trial. Clin Ther 2007; 29:2333–2348.
  56. Buse JB, Klonoff DC, Nielsen LL, et al. Metabolic effects of two years of exenatide treatment on diabetes, obesity, and hepatic biomarkers in patients with type 2 diabetes: an interim analysis of data from the open-label, uncontrolled extension of three double-blind, placebo-controlled trials. Clin Ther 2007; 29:139–153.
  57. Blonde L, Klein EJ, Han J, et al. Interim analysis of the effects of exenatide treatment on A1C, weight and cardiovascular risk factors over 82 weeks in 314 overweight patients with type 2 diabetes. Diabetes Obes Metab 2006; 8:436–447.
  58. Klonoff DC, Buse JB, Nielsen LL, et al. Exenatide effects on diabetes, obesity, cardiovascular risk factors and hepatic biomarkers in patients with type 2 diabetes treated for at least 3 years. Curr Med Res Opin 2008; 24:275–286.
  59. Kim D, MacConell L, Zhuang D, et al. Effects of once-weekly dosing of a long-acting release formulation of exenatide on glucose control and body weight in subjects with type 2 diabetes. Diabetes Care 2007; 30:1487–1493.
  60. Drucker DJ, Buse JB, Taylor K, et al; for the DURATION-1 Study Group. Exenatide once weekly versus twice daily for the treatment of type 2 diabetes: a randomised, open-label, non-inferiority study. Lancet 2008; 372:1240–1250.
  61. Bergenstal RM, Kim T, Trautmann M, Zhuang D, Okerson T, Taylor K. Exenatide once weekly elicited improvements in blood pressure and lipid profile over 52 weeks in patients with type 2 diabetes. Circulation 2008; 118:S1086. Abstract 1239.
  62. Nauck M, Frid A, Hermansen K, et al; for the LEAD-2 Study Group. Efficacy and safety comparison of liraglutide, glimepiride, and placebo, all in combination with metformin, in type 2 diabetes: the LEAD (Liraglutide Effect and Action in Diabetes)-2 study. Diabetes Care 2009; 32:84–90.
  63. Garber A, Henry R. Ratner R, et al; for the LEAD-3 (Mono) Study Group. Liraglutide versus glimepiride monotherapy for type 2 diabetes (LEAD-3 Mono): a randomised, 52-week, phase III, double-blind, parallel-treatment trial. Lancet 2009; 373:473–481.
  64. Buse JB, Rosenstock J, Sesti G, et al; for the LEAD-6 Study Group. Liraglutide once a day versus exenatide twice a day for type 2 diabetes: a 26-week randomised, parallel-group, multinational, open-label trial (LEAD-6). Lancet 2009; 374:39–47.
  65. Fortamet [package insert]. Ft. Lauderdale, FL: Watson Laboratories; 2008.
  66. Actos. Physicians’ Desk Reference. 63rd edition. Montvale, NJ: Physicians’ Desk Reference Inc; 2008; 3077–3082.
  67. Avandia. Physicians’ Desk Reference. 63rd edition. Montvale, NJ: Physicians’ Desk Reference Inc; 2009; 1351–1359.
  68. Byetta [package insert]. San Diego, CA: Amylin Pharmaceuticals, Inc.; 2009.
  69. Idris I. News and views: FDA reviews incidences of acute pancreatitis in patients taking Byetta. Diabetes Obes Metab 2008; 10:96–98.
  70. Januvia [package insert]. Whitehouse Station, NJ: Merck & Co., Inc.; 2006, 2007.
  71. American Diabetes Association. Standards of medical care in diabetes—2009. Diabetes Care 2009; 32(suppl 1):S13–S61.
  72. Bunck MC, Diamant M, Cornér A, et al. One-year treatment with exenatide improves beta-cell function, compared with insulin glargine, in metformin-treated type 2 diabetic patients: a randomized, controlled trial. Diabetes Care 2009; 32:762–768.
  73. Gastaldelli A, Ferrannini E, Miyazaki Y, Matsuda M, Mari A, DeFronzo RA. Thiazolidinediones improve beta-cell function in type 2 diabetic patients. Am J Physiol Endocrinol Metab 2007; 292:E871–E883.
  74. Buse JB, Ginsberg HN, Bakris GL, et al. Primary prevention of cardiovascular diseases in people with diabetes mellitus: a scientific statement from the American Heart Association and the American Diabetes Association. Diabetes Care 2007; 30:162–172.
Article PDF
Author and Disclosure Information

Anne L. Peters, MD, CDE
Director, USC Clinical Diabetes Programs, and Professor of Clinical Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA

Correspondence: Anne L. Peters, MD, CDE, Director, USC Clinical Diabetes Programs, 150 N. Robertson Blvd., Suite 210, Beverly Hills, CA 90211; [email protected]

Dr. Peters reported that she has received consulting/advisory fees from Abbott Laboratories, Amylin Pharmaceuticals, Inc., Eli Lilly and Company, Medtronic MiniMed, Inc., Novo Nordisk, and Takeda Pharmaceutical Company Limited; and honoraria from Amylin Pharmaceuticals, Inc., Novo Nordisk, and Takeda Pharmaceutical Company. Dr. Peters reported that she did not receive an honorarium for writing this article.

Dr. Peters reported that she wrote this article and received no assistance with content development from unnamed contributors. She reported that BlueSpark Healthcare Communications, a medical communications company, assisted with preliminary literature searches, reference verification, proofing for grammar and style, table and figure rendering based on author instructions, copyright permission requests, and identification of topical overlap with other manuscripts in this supplement.

Publications
Page Number
S20-S27
Author and Disclosure Information

Anne L. Peters, MD, CDE
Director, USC Clinical Diabetes Programs, and Professor of Clinical Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA

Correspondence: Anne L. Peters, MD, CDE, Director, USC Clinical Diabetes Programs, 150 N. Robertson Blvd., Suite 210, Beverly Hills, CA 90211; [email protected]

Dr. Peters reported that she has received consulting/advisory fees from Abbott Laboratories, Amylin Pharmaceuticals, Inc., Eli Lilly and Company, Medtronic MiniMed, Inc., Novo Nordisk, and Takeda Pharmaceutical Company Limited; and honoraria from Amylin Pharmaceuticals, Inc., Novo Nordisk, and Takeda Pharmaceutical Company. Dr. Peters reported that she did not receive an honorarium for writing this article.

Dr. Peters reported that she wrote this article and received no assistance with content development from unnamed contributors. She reported that BlueSpark Healthcare Communications, a medical communications company, assisted with preliminary literature searches, reference verification, proofing for grammar and style, table and figure rendering based on author instructions, copyright permission requests, and identification of topical overlap with other manuscripts in this supplement.

Author and Disclosure Information

Anne L. Peters, MD, CDE
Director, USC Clinical Diabetes Programs, and Professor of Clinical Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA

Correspondence: Anne L. Peters, MD, CDE, Director, USC Clinical Diabetes Programs, 150 N. Robertson Blvd., Suite 210, Beverly Hills, CA 90211; [email protected]

Dr. Peters reported that she has received consulting/advisory fees from Abbott Laboratories, Amylin Pharmaceuticals, Inc., Eli Lilly and Company, Medtronic MiniMed, Inc., Novo Nordisk, and Takeda Pharmaceutical Company Limited; and honoraria from Amylin Pharmaceuticals, Inc., Novo Nordisk, and Takeda Pharmaceutical Company. Dr. Peters reported that she did not receive an honorarium for writing this article.

Dr. Peters reported that she wrote this article and received no assistance with content development from unnamed contributors. She reported that BlueSpark Healthcare Communications, a medical communications company, assisted with preliminary literature searches, reference verification, proofing for grammar and style, table and figure rendering based on author instructions, copyright permission requests, and identification of topical overlap with other manuscripts in this supplement.

Article PDF
Article PDF
Related Articles

Type 2 diabetes mellitus (T2DM), excess weight, and obesity are increasing in prevalence at alarming rates.1–3 Concurrent with the increased prevalence is increased risk of morbidity and mortality. A healthy diet and exercise in conjunction with antidiabetes medications can help lower glucose concentration in patients with T2DM. Because these patients are at increased risk of cardiovascular (CV) morbidity and mortality, however, treatment strategies should address the CV risk factors, including blood pressure (BP), lipids, and body weight, as well as glycemic aspects of the disease.

To help clinicians manage the complex issues in treating patients with T2DM, this article presents an overview of patient and treatment perspectives relevant to overweight/obesity and CV disease (CVD). It includes an examination of the latest guidelines and algorithms for the management of T2DM, which continue to be updated and modified.

T2DM, WEIGHT GAIN OR OBESITY, AND CV RISK: A CHALLENGING TRIAD

Despite therapeutic advances in the diagnosis and treatment of diabetes and CVD over the last decade, the estimated number of persons in the United States older than 35 years with self-reported diabetes (with T2DM accounting for 90% to 95% of diagnosed cases) and CVD has increased from 4.2 million in 1997 to 5.7 million in 2005.3,4 The CV risk for patients with T2DM who have not had a CV event such as a myocardial infarction (MI) is similar to that of individuals without diabetes who have had a prior MI.5 Patients with T2DM have nearly double the mortality of those without the disease.6 Adding to their risk, about 80% of patients with T2DM are overweight or obese, conditions associated with worsened insulin resistance and increased CV risk and disease burden.7,8 Even a modest weight gain (5 kg) may increase the risk of coronary heart disease (CHD) by 30%, while associated changes in lipids and BP can increase the risk by another 20%.9

It is as important to control CV risk factors as it is to control glycemia in patients with T2DM, and both are difficult to achieve. Data from a recent nationwide Norwegian survey showed that only 13% of patients with T2DM achieved study-defined target levels; ie, glycosylated hemoglobin (HbA1c) less than 7.5%, BP less than 140/85 mm Hg, and total cholesterol/high-density lipoprotein (HDL-C) ratio less than 4.0.10

BENEFITS OF MANAGING GLYCEMIA, WEIGHT REDUCTION, AND CV RISK FACTORS

Several large studies, many ongoing, are generating data on the relationships among glycemia, weight reduction, and CV risk. It is well established that individuals with T2DM need aggressive risk factor reduction (glucose control, blood pressure management, and treatment of dyslipidemia) to optimize outcomes. However, characterization of the benefits of various components of risk factor reduction, particularly over many years, is only now occurring.

Results from the United Kingdom Prospective Diabetes Studies (UKPDS) showed the benefits and risks of pharmacologic glycemic control—essentially monotherapy with insulin or a sulfonylurea—compared with conventional dietary therapy in reducing diabetic complications in patients with newly diagnosed T2DM. In UKPDS 33, both insulin and sulfonylureas (intensive treatment) reduced the risk of microvascular end points (retinopathy, nephropathy) in patients whose median HbA1c was lowered to 7.0% at 10 years of follow-up, compared with patients who reached an HbA1c of 7.9%. However, intensive glycemic control did not translate into a statistically significant reduction in macrovascular complications, including MI, stroke, CVD, and death. Additionally, patients assigned to insulin had greater weight gain (+4.0 kg) than did patients assigned to receive the sulfonylurea chlorpropamide (+2.6 kg) or glyburide (+1.7 kg) (P < .01).11

The UKPDS showed that intensive treatment with metformin reduced the risk of T2DM-related end points compared with conventional treatment (primarily diet alone) in overweight patients.12 Although there were fewer patients in the metformin-treated subset (n = 342) than in the conventional treatment cohort, a secondary analysis showed that metformin was associated with less weight gain and fewer hypoglycemic episodes than either insulin or sulfonylurea therapy.12 Since HbA1c levels in the treatment groups were equal, the additional benefits seen with metformin in overweight patients with T2DM were not based solely on glycemic control.

The Action to Control Cardiovascular Risk in Diabetes (ACCORD) trial involved 10,000 individuals with T2DM. The primary outcome measure was a composite of CV events. The intensively treated group was controlled to a target HbA1c of less than 6.0%, with most patients receiving insulin. The trial was terminated early because an increased risk of sudden death was observed.13 A similar study, Action in Diabetes and Vascular Disease: Preterax and Diamicron Modified Release Controlled Evaluation (ADVANCE), evaluated more than 11,000 patients with T2DM, starting with a sulfonylurea-based regimen. In this study, there was no reduction in macrovascular events, but there was a reduction in nephropathy in the intensively treated group.14 In both studies, hypoglycemia and weight gain were more frequent in intensively treated patients; and in ACCORD, there were more episodes of severe hypoglycemia in the intensive-treatment group.13,14

The Veterans Affairs Diabetes Trial (VADT) evaluated the effect of intensive glucose control on CVD in 1,791 patients (mean age, 60 years) with poorly controlled T2DM (average duration, 11.5 years). The primary end points included MI, stroke, new or worsening congestive heart failure (CHF), limb amputation, and invasive intervention for coronary or peripheral arterial disease. The hazard ratio for these end points in the intensive-treatment group was 0.88 (95% confidence interval [CI], 0.74 to 1.05).15,16 Specifically, the following beneficial effects were achieved:

  • HbA1c reduced by –1.0% to –2.5% in absolute units,
  • systolic BP (SBP) reduced by –4 to –7 mm Hg,
  • diastolic BP (DBP) reduced by –7 to –8 mm Hg,
  • low-density lipoprotein cholesterol (LDL-C) reduced by –27 to –28 mg/dL,
  • triglycerides reduced by –44 to –50 mg/dL, and
  • HDL-C increased by 4 to 5 mg/dL.

Despite these benefits, body weight increased approximately 9 to 18 lb (4 to 8 kg) during therapy.15

Since overweight and obesity are independent risk factors for CHD and CVD in patients with T2DM,17 weight management is an integral component in treatment. In the Action for Health in Diabetes (Look AHEAD) trial, an intensive exercise and weight-loss program resulted in clinically significant (P < .001) weight loss at 1 year in patients who had T2DM and a body mass index (BMI) greater than 25 kg2/m (> 27 kg2/m if receiving insulin).18 When compared with patients who received less structured, infrequent support and minimal education about diabetes, participants in the intensive program showed more weight loss, improved glucose control, decreased CV events, and reduced medicine use. The Look AHEAD trial is currently evaluating whether these improvements will continue to result in lower CV risk.

 

 

PATIENT ADHERENCE AND SATISFACTION

It is often challenging for patients with T2DM to adhere to their treatment regimens. The Diabetes Attitude, Wishes, and Needs (DAWN) study examined psychosocial barriers to self-care in patients with diabetes and found that while 78% of patients with T2DM adhered to their medications, only 39% achieved complete success in at least two-thirds of their self-care domains.19 A multicenter, randomized, clinical trial examined the correlates of treatment satisfaction, including body weight, on patients’ appraisal of treatment satisfaction with injectable insulin. The 14.5% of patients who experienced a reduction in BMI reported systematic improvement in treatment satisfaction.20 Similarly, a cross-sectionally designed study (n = 99) that analyzed the interrelation of adherence, BMI, and depression in adults with T2DM found that patients with higher BMI and poor adherence also had depression, which was mediated by lower self-efficacy perceptions and increased diabetes symptoms.21 The results from these studies show a clear relationship between adherence with treatment regimens and achievement of HbA1c goals.22

RECENT DEVELOPMENTS IN T2DM MANAGEMENT: STRATEGIES TO REDUCE CV RISK

Because excess weight and obesity are prominent features of T2DM, it is important to use an antidiabetes agent that does not induce unnecessary weight gain (particularly central weight gain, which is thought to be most atherogenic).23 Metformin, considered the first-line agent for treatment of T2DM, is generally weight neutral with a low level of hypoglycemia.24,25 Sulfonylureas, insulin, and thiazolidinediones (TZDs) are all associated with weight gain, although newer-analogue insulins may cause less weight gain than older agents. TZDs, especially pioglitazone, are associated with improvements in long-term beta-cell function and CV risk factors despite weight gain.26,27

The newer antidiabetes agents belong to the dipeptidyl peptidase–4 (DPP-4) inhibitor and the glucagon-like peptide–1 (GLP-1) receptor agonist therapeutic classes and have been shown to be either weight neutral (DPP-4 inhibitors) or to cause weight loss (GLP-1 receptor agonists).28

Figure 1. Actions of glucagon-like peptide–1 (GLP-1) in peripheral tissues. Most of the effects of GLP-1 are mediated by direct interaction with GLP-1 receptors on specific tissues. However, the actions of GLP-1 in liver, fat, and muscle most likely occur through indirect mechanisms.
Figure 1 illustrates the physiologic role of GLP-1,29 which induces glucose-dependent insulin secretion after food intake by binding to specific receptors on pancreatic beta cells, suppresses postprandial glucagon from pancreatic alpha cells, reduces postprandial plasma glucose (PPG) concentrations by delaying gastric emptying, and diminishes appetite.28 The diminished secretion of GLP-1 in T2DM30,31 has led to the development of two different treatment approaches.28 Since GLP-1 is rapidly degraded by DPP-4, GLP-1 receptor agonists have been developed to resist DPP-4 inactivation while exhibiting many of the actions of endogenous incretin hormones.28,29 DPP-4 inhibitors function as incretin enhancers by protecting endogenous GLP-1 and glucose-dependent insulinotropic peptide, another incretin, from enzymatic breakdown.31–33 Unlike the GLP-1 receptor agonists, which are administered subcutaneously (SC), DPP-4 inhibitors are administered orally.

Obesity and the incretin effect

Figure 2. Adipokine expression and secretion by adipose tissue in insulin-resistant, obese subjects.
A study in healthy subjects and patients with T2DM demonstrated that glucose tolerance and obesity independently impair the incretin effect, resulting in impaired insulin secretion and glucagon suppression.34 Obesity is considered a subclinical inflammatory condition that releases chemokines, leading to insulin resistance. Figure 2 illustrates the interaction between obesity, inflammation, and insulin resistance.35

Two recent studies showed that surgically induced weight loss enhances the physiologic “incretin effect.” In one study, obese individuals with T2DM whose weight loss was secondary to bariatric surgery combined with caloric restriction showed improved insulin sensitivity, improved carbohydrate metabolism, and elevated levels of adiponectin and GLP-1, all of which may reduce the incidence of T2DM.36 In the other study, bariatric surgery in morbidly obese individuals with T2DM improved insulin secretion and ameliorated insulin resistance.37

DPP-4 inhibitors

DPP-4 inhibitors such as sitagliptin and saxagliptin inhibit the enzymatic activity of DPP-4 and increase endogenous concentrations of GLP-1.28 Sitagliptin has been compared with placebo as monotherapy and has been studied in combination with other therapies.

In an 18-week study, sitagliptin monotherapy, 100 and 200 mg QD, significantly reduced HbA1c compared with placebo (placebo-subtracted HbA1c reduction, –0.60% and –0.48%, respectively) in patients with T2DM. Sitagliptin also significantly decreased fasting plasma glucose (FPG) concentration relative to placebo.38 Twelve weeks of sitagliptin monotherapy at dosages of 5, 12.5, 25, and 50 mg BID led to significant (P < .001) reductions in HbA1c compared with placebo. Sitagliptin also produced significant reductions in FPG and mean daily glucose concentrations across the doses studied.39 Similar results were reported in other 12-week studies: 50 mg BID and 100 mg QD sitagliptin monotherapy significantly (P < .05) reduced HbA1c –0.39% to –0.56% and FPG concentration –11.0 to –17.2 mg/dL compared with placebo40; sitagliptin 100 mg QD compared with placebo produced a least-squares mean change from baseline HbA1c of –0.65% versus 0.41% (P < .001) and FPG of –22.5 versus 9.4 mg/dL (P < .001).41

Sitagliptin also has been studied in combination with other therapies. After 24 weeks, sitagliptin combined with pioglitazone significantly reduced HbA1c by –0.70% and FPG by –17.7 mg/dL (P < .001 for both) compared with placebo.42 In another 24-week study, 100 mg sitagliptin QD significantly improved glycemic control and beta-cell function (P < .05 for both) in patients with T2DM who had inadequate glycemic control with glimepiride or glimepiride plus metformin.43

In addition to significantly reducing HbA1c, sitagliptin 100 and 200 mg QD produced only small differences in body weight relative to placebo: least-squares mean change from baseline for sitagliptin 100 mg was –0.7 kg (95% CI, –1.3 to –0.1) and for 200 mg was –0.6 kg (95% CI, –1.0 to –0.2); for placebo it was –0.2 kg (95% CI, –0.7 to 0.2).38 These findings were consistent with those from another 24-week monotherapy study where sitagliptin produced weight loss of up to –0.2 kg44 and a 30-week study of sitagliptin added to ongoing metformin therapy. In the latter study, both sitagliptin and placebo resulted in weight reductions of –0.5 kg.45

The effects of sitagliptin on lipids and BP have been reported in clinical studies in patients with and without T2DM. In one study of patients with T2DM, the addition of sitagliptin to metformin increased total cholesterol (+8.1 mg/dL), LDL-C (+9.2 mg/dL), and HDL-C (+1.8 mg/dL) but lowered triglyceride (–14.5 mg/dL) after 18 weeks of treatment (24-week data).46 Data from a small (n = 19) study in nondiabetic patients with mild to moderate hypertension showed that sitagliptin produced small reductions (–2 to –3 mm Hg) in 24-hour ambulatory BP measurements.47

Another DPP-4 inhibitor, saxagliptin, with efficacy similar to that described for sitagliptin, was recently approved by the US Food and Drug Administration (FDA) for treatment of T2DM.48

 

 

GLP-1 receptor agonists

Many of the GLP-1 receptor agonists developed or under development have glucoregulatory effects similar to GLP-1 but are resistant to degradation by DPP-4.28 Exenatide, an exendin-4 receptor agonist, has compared favorably with sitagliptin and with insulin analogues. Long-acting (once-weekly and once-daily) GLP-1 receptor agonists are under development.

In a 2-week, head-to-head study in metformin-treated patients with T2DM, exenatide had a greater effect than sitagliptin in lowering PPG and was more potent in increasing insulin secretion and reducing postprandial glucagon secretion. In contrast to sitagliptin, exenatide slowed gastric emptying and reduced caloric intake.49

In two studies of patients treated with exenatide, on a background of either metformin alone or metformin plus a sulfonylurea, patients who received metformin lost more weight (–1.6 to –2.8 kg; P ≤ .01) and experienced more significant decreases from baseline HbA1c (–0.4% to –0.8%; P < .002) at 30 weeks than did patients who received placebo.50,51 In a 16-week trial of exenatide in patients previously treated with a TZD with or without metformin, exenatide reduced HbA1c –0.98%, fasting blood glucose –1.69 mmol/L, and body weight –1.51 kg.52

When compared with insulin analogues, exenatide has been associated with weight loss (~ –3 kg) while the insulin analogues were associated with weight gain (~ +3 kg).53 After 26 weeks, body weight decreased –2.3 kg with exenatide and increased +1.8 kg with insulin glargine.54 Similar results were found in a crossover noninferiority trial, where the least-squares mean difference in weight change was significantly (P < .001) different (2.2 kg) between the treatments.55 When exenatide was compared with insulin aspart in an open-label, noninferiority trial, there was a between-group difference in weight of –5.4 kg after 52 weeks.32

Exenatide has also demonstrated these benefits in open-label extension studies. After 2 years, mean HbA1c reductions of –1.1% from baseline were sustained (P < .05), and weight loss of –4.7 kg was maintained (P < .001).56 After 82 weeks, similar HbA1c decreases (–1.1%) and weight loss (–4.4 kg) were exhibited.57 Even after 3 years, these benefits were maintained in patients who remained on the drug (HbA1c reduction from baseline, –1.0%; weight loss, –5.3 kg [P < .0001 for both]).58

Long-acting formulations of GLP-1 receptor agonists are in clinical development; two of these are once-weekly exenatide and once-daily liraglutide. Exenatide once weekly has the advantage of less frequent dosing and has elicited greater reductions in HbA1c than exenatide BID. After 15 weeks of once-weekly administration, the 0.8-mg formulation reduced HbA1c –1.4% and the 2-mg formulation reduced it –1.7% (P < .0001 for both compared with placebo). Body weight was lowered –3.8 kg (P < .05 compared with placebo) with the 2-mg formulation.59 Compared with exenatide BID, exenatide 2 mg once weekly showed greater reductions in HbA1c (–1.9% vs –1.5%; P = .0023) after 30 weeks of therapy.60 In a 1-year noncomparative trial, treatment with exenatide once weekly improved HbA1c (–2.0%) and weight (–4.1 kg), as well as BP and lipid profiles compared with baseline.61

Liraglutide, a once-daily human analogue GLP-1 receptor agonist, is under review by the FDA.28 In a 26-week study of patients with T2DM, liraglutide was associated with reductions in HbA1c (mean, –1.04%; P = 0.067 compared with insulin) and body weight (mean, –2.5 kg; P < .001 compared with insulin) at dosages of 0.6 to 1.8 mg/day SC. Liraglutide produced a decline in SBP from 0.6 to 3 mm Hg but was not associated with a decrease in DBP.62 In a 52-week study comparing liraglutide with glimepiride monotherapy, liraglutide 1.2 mg was associated with an HbA1c reduction of –0.84% (P = .0014) and the 1.8-mg dose with a reduction of –1.14% (P < .0001) compared with –0.51% for glimepiride. SBP decreased –0.7 mm Hg with glimepiride compared with –2.1 mm Hg for liraglutide 1.2 mg (P = .2912) and –3.6 mm Hg for liraglutide 1.8 mg (P < .0118). Mean DBP fell slightly but not significantly in all treatment groups.63 No effects on lipid parameters were reported in these two liraglutide studies.

The Liraglutide Effect and Action in Diabetes (LEAD-6) trial was undertaken to compare exenatide (10 mg BID SC) and liraglutide (1.8 mg/day SC) as add-on therapy to metformin, a sulfonylurea, or a combination of both in 464 patients with T2DM. After 26 weeks of treatment, liraglutide was associated with a significant reduction in HbA1c of –1.12%, compared with –0.79% with exenatide (P < .0001). Patients treated with liraglutide lost –3.2 kg while those on exenatide lost –2.9 kg. Among patients previously treated with metformin alone, there was a 1-kg difference in favor of liraglutide (P = NS).64

Safety profile

All of the drugs discussed have potential adverse effects. Metformin continues to have a black box warning for lactic acidosis.65 Sulfonylureas and insulin can cause hypoglycemia. TZDs can cause fluid retention and, in rare cases, CHF (for which these drugs also carry a black box warning).66,67 TZDs also increase the risk of distal fracture.66,67 The most common side effects of exenatide are gastrointestinal, but there have been reported cases of pancreatitis, some of which have been fatal.68,69 It has been difficult to prove whether exenatide increases the risk of pancreatitis, as patients with T2DM are already at an increased (three- to fourfold) risk for this condition compared with persons who do not have T2DM.69 Exenatide should not be used in patients with severe renal impairment or end-stage renal disease; it should be used with caution in patients who have undergone renal transplantation and in patients with moderate renal impairment.

The prescribing information for sitagliptin includes pancreatitis among the adverse reactions identified during the drug’s postapproval use.70 As with exenatide, it is not fully known whether a true association exists between the agent and pancreatitis. However, since pancreatitis can occur in this patient population, it is recommended that abdominal pain be fully evaluated to rule out pancreatitis. Continued postmarketing surveillance is important for all of these agents.

THE ROLE OF GUIDELINES

The American Association of Clinical Endocrinologists (AACE),26 the American Diabetes Association (ADA),71 and the ADA in conjunction with the European Association for the Study of Diabetes (EASD)24 have recently revised their recommendations for the management of patients with diabetes. The guidelines are unanimous in setting a glycemic goal (HbA1c < 7.0% for the ADA, HbA1c ≤ 6.5% for the AACE) and advocating individualized care for a treatment goal of HbA1c lower than 6.0% in patients who stand to benefit from near euglycemia without inducing severe hypoglycemia.24,26,71

CVD is the major cause of morbidity and mortality associated with T2DM and is a source of increasing concern.5 Accordingly, special consideration should be given to patients with coexisting CV risk factors, including hypertension and dyslipidemia. The ADA and the EASD advocate lifestyle modification to decrease body weight and the concurrent initiation of metformin as first-line therapy.24 If that strategy is insufficient, then two tiers of treatment guide the choice of next steps24:

  • Tier 1, in addition to metformin, includes the sulfonylureas and insulin. Although these are excellent glucose-lowering drugs, they are associated with weight gain, hypoglycemia, and no improvement in BP or lipid levels. They are relatively low in cost and have been used for many years. Their main drawback is evidence that despite their use, beta-cell failure continues unabated over time.
  • Tier 2 treatments include pioglitazone and the GLP-1 receptor agonist exenatide. Consideration may be given to the use of pioglitazone or exenatide when hypoglycemia is of concern, with exenatide being preferred when weight loss is a major objective and HbA1c is close to target (< 8.0%).24 Additionally, both the TZDs and exenatide probably help slow the rate of beta-cell failure, particularly if they are used early in the course of the disease.72,73 The AACE recommends different pharmacologic approaches based on HbA1c at diagnosis.26

The American Heart Association and the ADA have issued a joint scientific statement on the primary prevention of CVD in patients with diabetes.74 They advocate lifestyle management of body weight, nutrition, and physical activity.74 In addition, they stress the need for attention to BP, lipid levels, and smoking status, and the use of antiplatelet agents in patients at increased CV risk (> 40 years of age and a family history of CVD, hypertension, smoking, dyslipidemia, or albuminuria).

CONCLUSION

T2DM, weight gain/obesity, and CV risk present a continuing challenge to patients and clinicians. Anti­diabetes agents have varying degrees of evidence to support their effects on HbA1c, body weight, BP, and lipid levels. A better understanding of the pathophysiology of T2DM has led to the development of newer antidiabetes agents that target the fundamental defects of the disease. Evidence continues to accumulate for the improved benefits of glycemic control and weight loss in T2DM with GLP-1 receptor agonists such as exenatide currently having robust data in terms of beneficial effects on weight and CV risk factors. As clinicians continue to incorporate this knowledge into their practice patterns, patient adherence and clinical outcomes are expected to improve. Newer agents, such as incretin-based therapies, address T2DM as well as other factors that increase cardiometabolic risk through their effects not only on glycemic control but on body weight, BP, and lipids.

Type 2 diabetes mellitus (T2DM), excess weight, and obesity are increasing in prevalence at alarming rates.1–3 Concurrent with the increased prevalence is increased risk of morbidity and mortality. A healthy diet and exercise in conjunction with antidiabetes medications can help lower glucose concentration in patients with T2DM. Because these patients are at increased risk of cardiovascular (CV) morbidity and mortality, however, treatment strategies should address the CV risk factors, including blood pressure (BP), lipids, and body weight, as well as glycemic aspects of the disease.

To help clinicians manage the complex issues in treating patients with T2DM, this article presents an overview of patient and treatment perspectives relevant to overweight/obesity and CV disease (CVD). It includes an examination of the latest guidelines and algorithms for the management of T2DM, which continue to be updated and modified.

T2DM, WEIGHT GAIN OR OBESITY, AND CV RISK: A CHALLENGING TRIAD

Despite therapeutic advances in the diagnosis and treatment of diabetes and CVD over the last decade, the estimated number of persons in the United States older than 35 years with self-reported diabetes (with T2DM accounting for 90% to 95% of diagnosed cases) and CVD has increased from 4.2 million in 1997 to 5.7 million in 2005.3,4 The CV risk for patients with T2DM who have not had a CV event such as a myocardial infarction (MI) is similar to that of individuals without diabetes who have had a prior MI.5 Patients with T2DM have nearly double the mortality of those without the disease.6 Adding to their risk, about 80% of patients with T2DM are overweight or obese, conditions associated with worsened insulin resistance and increased CV risk and disease burden.7,8 Even a modest weight gain (5 kg) may increase the risk of coronary heart disease (CHD) by 30%, while associated changes in lipids and BP can increase the risk by another 20%.9

It is as important to control CV risk factors as it is to control glycemia in patients with T2DM, and both are difficult to achieve. Data from a recent nationwide Norwegian survey showed that only 13% of patients with T2DM achieved study-defined target levels; ie, glycosylated hemoglobin (HbA1c) less than 7.5%, BP less than 140/85 mm Hg, and total cholesterol/high-density lipoprotein (HDL-C) ratio less than 4.0.10

BENEFITS OF MANAGING GLYCEMIA, WEIGHT REDUCTION, AND CV RISK FACTORS

Several large studies, many ongoing, are generating data on the relationships among glycemia, weight reduction, and CV risk. It is well established that individuals with T2DM need aggressive risk factor reduction (glucose control, blood pressure management, and treatment of dyslipidemia) to optimize outcomes. However, characterization of the benefits of various components of risk factor reduction, particularly over many years, is only now occurring.

Results from the United Kingdom Prospective Diabetes Studies (UKPDS) showed the benefits and risks of pharmacologic glycemic control—essentially monotherapy with insulin or a sulfonylurea—compared with conventional dietary therapy in reducing diabetic complications in patients with newly diagnosed T2DM. In UKPDS 33, both insulin and sulfonylureas (intensive treatment) reduced the risk of microvascular end points (retinopathy, nephropathy) in patients whose median HbA1c was lowered to 7.0% at 10 years of follow-up, compared with patients who reached an HbA1c of 7.9%. However, intensive glycemic control did not translate into a statistically significant reduction in macrovascular complications, including MI, stroke, CVD, and death. Additionally, patients assigned to insulin had greater weight gain (+4.0 kg) than did patients assigned to receive the sulfonylurea chlorpropamide (+2.6 kg) or glyburide (+1.7 kg) (P < .01).11

The UKPDS showed that intensive treatment with metformin reduced the risk of T2DM-related end points compared with conventional treatment (primarily diet alone) in overweight patients.12 Although there were fewer patients in the metformin-treated subset (n = 342) than in the conventional treatment cohort, a secondary analysis showed that metformin was associated with less weight gain and fewer hypoglycemic episodes than either insulin or sulfonylurea therapy.12 Since HbA1c levels in the treatment groups were equal, the additional benefits seen with metformin in overweight patients with T2DM were not based solely on glycemic control.

The Action to Control Cardiovascular Risk in Diabetes (ACCORD) trial involved 10,000 individuals with T2DM. The primary outcome measure was a composite of CV events. The intensively treated group was controlled to a target HbA1c of less than 6.0%, with most patients receiving insulin. The trial was terminated early because an increased risk of sudden death was observed.13 A similar study, Action in Diabetes and Vascular Disease: Preterax and Diamicron Modified Release Controlled Evaluation (ADVANCE), evaluated more than 11,000 patients with T2DM, starting with a sulfonylurea-based regimen. In this study, there was no reduction in macrovascular events, but there was a reduction in nephropathy in the intensively treated group.14 In both studies, hypoglycemia and weight gain were more frequent in intensively treated patients; and in ACCORD, there were more episodes of severe hypoglycemia in the intensive-treatment group.13,14

The Veterans Affairs Diabetes Trial (VADT) evaluated the effect of intensive glucose control on CVD in 1,791 patients (mean age, 60 years) with poorly controlled T2DM (average duration, 11.5 years). The primary end points included MI, stroke, new or worsening congestive heart failure (CHF), limb amputation, and invasive intervention for coronary or peripheral arterial disease. The hazard ratio for these end points in the intensive-treatment group was 0.88 (95% confidence interval [CI], 0.74 to 1.05).15,16 Specifically, the following beneficial effects were achieved:

  • HbA1c reduced by –1.0% to –2.5% in absolute units,
  • systolic BP (SBP) reduced by –4 to –7 mm Hg,
  • diastolic BP (DBP) reduced by –7 to –8 mm Hg,
  • low-density lipoprotein cholesterol (LDL-C) reduced by –27 to –28 mg/dL,
  • triglycerides reduced by –44 to –50 mg/dL, and
  • HDL-C increased by 4 to 5 mg/dL.

Despite these benefits, body weight increased approximately 9 to 18 lb (4 to 8 kg) during therapy.15

Since overweight and obesity are independent risk factors for CHD and CVD in patients with T2DM,17 weight management is an integral component in treatment. In the Action for Health in Diabetes (Look AHEAD) trial, an intensive exercise and weight-loss program resulted in clinically significant (P < .001) weight loss at 1 year in patients who had T2DM and a body mass index (BMI) greater than 25 kg2/m (> 27 kg2/m if receiving insulin).18 When compared with patients who received less structured, infrequent support and minimal education about diabetes, participants in the intensive program showed more weight loss, improved glucose control, decreased CV events, and reduced medicine use. The Look AHEAD trial is currently evaluating whether these improvements will continue to result in lower CV risk.

 

 

PATIENT ADHERENCE AND SATISFACTION

It is often challenging for patients with T2DM to adhere to their treatment regimens. The Diabetes Attitude, Wishes, and Needs (DAWN) study examined psychosocial barriers to self-care in patients with diabetes and found that while 78% of patients with T2DM adhered to their medications, only 39% achieved complete success in at least two-thirds of their self-care domains.19 A multicenter, randomized, clinical trial examined the correlates of treatment satisfaction, including body weight, on patients’ appraisal of treatment satisfaction with injectable insulin. The 14.5% of patients who experienced a reduction in BMI reported systematic improvement in treatment satisfaction.20 Similarly, a cross-sectionally designed study (n = 99) that analyzed the interrelation of adherence, BMI, and depression in adults with T2DM found that patients with higher BMI and poor adherence also had depression, which was mediated by lower self-efficacy perceptions and increased diabetes symptoms.21 The results from these studies show a clear relationship between adherence with treatment regimens and achievement of HbA1c goals.22

RECENT DEVELOPMENTS IN T2DM MANAGEMENT: STRATEGIES TO REDUCE CV RISK

Because excess weight and obesity are prominent features of T2DM, it is important to use an antidiabetes agent that does not induce unnecessary weight gain (particularly central weight gain, which is thought to be most atherogenic).23 Metformin, considered the first-line agent for treatment of T2DM, is generally weight neutral with a low level of hypoglycemia.24,25 Sulfonylureas, insulin, and thiazolidinediones (TZDs) are all associated with weight gain, although newer-analogue insulins may cause less weight gain than older agents. TZDs, especially pioglitazone, are associated with improvements in long-term beta-cell function and CV risk factors despite weight gain.26,27

The newer antidiabetes agents belong to the dipeptidyl peptidase–4 (DPP-4) inhibitor and the glucagon-like peptide–1 (GLP-1) receptor agonist therapeutic classes and have been shown to be either weight neutral (DPP-4 inhibitors) or to cause weight loss (GLP-1 receptor agonists).28

Figure 1. Actions of glucagon-like peptide–1 (GLP-1) in peripheral tissues. Most of the effects of GLP-1 are mediated by direct interaction with GLP-1 receptors on specific tissues. However, the actions of GLP-1 in liver, fat, and muscle most likely occur through indirect mechanisms.
Figure 1 illustrates the physiologic role of GLP-1,29 which induces glucose-dependent insulin secretion after food intake by binding to specific receptors on pancreatic beta cells, suppresses postprandial glucagon from pancreatic alpha cells, reduces postprandial plasma glucose (PPG) concentrations by delaying gastric emptying, and diminishes appetite.28 The diminished secretion of GLP-1 in T2DM30,31 has led to the development of two different treatment approaches.28 Since GLP-1 is rapidly degraded by DPP-4, GLP-1 receptor agonists have been developed to resist DPP-4 inactivation while exhibiting many of the actions of endogenous incretin hormones.28,29 DPP-4 inhibitors function as incretin enhancers by protecting endogenous GLP-1 and glucose-dependent insulinotropic peptide, another incretin, from enzymatic breakdown.31–33 Unlike the GLP-1 receptor agonists, which are administered subcutaneously (SC), DPP-4 inhibitors are administered orally.

Obesity and the incretin effect

Figure 2. Adipokine expression and secretion by adipose tissue in insulin-resistant, obese subjects.
A study in healthy subjects and patients with T2DM demonstrated that glucose tolerance and obesity independently impair the incretin effect, resulting in impaired insulin secretion and glucagon suppression.34 Obesity is considered a subclinical inflammatory condition that releases chemokines, leading to insulin resistance. Figure 2 illustrates the interaction between obesity, inflammation, and insulin resistance.35

Two recent studies showed that surgically induced weight loss enhances the physiologic “incretin effect.” In one study, obese individuals with T2DM whose weight loss was secondary to bariatric surgery combined with caloric restriction showed improved insulin sensitivity, improved carbohydrate metabolism, and elevated levels of adiponectin and GLP-1, all of which may reduce the incidence of T2DM.36 In the other study, bariatric surgery in morbidly obese individuals with T2DM improved insulin secretion and ameliorated insulin resistance.37

DPP-4 inhibitors

DPP-4 inhibitors such as sitagliptin and saxagliptin inhibit the enzymatic activity of DPP-4 and increase endogenous concentrations of GLP-1.28 Sitagliptin has been compared with placebo as monotherapy and has been studied in combination with other therapies.

In an 18-week study, sitagliptin monotherapy, 100 and 200 mg QD, significantly reduced HbA1c compared with placebo (placebo-subtracted HbA1c reduction, –0.60% and –0.48%, respectively) in patients with T2DM. Sitagliptin also significantly decreased fasting plasma glucose (FPG) concentration relative to placebo.38 Twelve weeks of sitagliptin monotherapy at dosages of 5, 12.5, 25, and 50 mg BID led to significant (P < .001) reductions in HbA1c compared with placebo. Sitagliptin also produced significant reductions in FPG and mean daily glucose concentrations across the doses studied.39 Similar results were reported in other 12-week studies: 50 mg BID and 100 mg QD sitagliptin monotherapy significantly (P < .05) reduced HbA1c –0.39% to –0.56% and FPG concentration –11.0 to –17.2 mg/dL compared with placebo40; sitagliptin 100 mg QD compared with placebo produced a least-squares mean change from baseline HbA1c of –0.65% versus 0.41% (P < .001) and FPG of –22.5 versus 9.4 mg/dL (P < .001).41

Sitagliptin also has been studied in combination with other therapies. After 24 weeks, sitagliptin combined with pioglitazone significantly reduced HbA1c by –0.70% and FPG by –17.7 mg/dL (P < .001 for both) compared with placebo.42 In another 24-week study, 100 mg sitagliptin QD significantly improved glycemic control and beta-cell function (P < .05 for both) in patients with T2DM who had inadequate glycemic control with glimepiride or glimepiride plus metformin.43

In addition to significantly reducing HbA1c, sitagliptin 100 and 200 mg QD produced only small differences in body weight relative to placebo: least-squares mean change from baseline for sitagliptin 100 mg was –0.7 kg (95% CI, –1.3 to –0.1) and for 200 mg was –0.6 kg (95% CI, –1.0 to –0.2); for placebo it was –0.2 kg (95% CI, –0.7 to 0.2).38 These findings were consistent with those from another 24-week monotherapy study where sitagliptin produced weight loss of up to –0.2 kg44 and a 30-week study of sitagliptin added to ongoing metformin therapy. In the latter study, both sitagliptin and placebo resulted in weight reductions of –0.5 kg.45

The effects of sitagliptin on lipids and BP have been reported in clinical studies in patients with and without T2DM. In one study of patients with T2DM, the addition of sitagliptin to metformin increased total cholesterol (+8.1 mg/dL), LDL-C (+9.2 mg/dL), and HDL-C (+1.8 mg/dL) but lowered triglyceride (–14.5 mg/dL) after 18 weeks of treatment (24-week data).46 Data from a small (n = 19) study in nondiabetic patients with mild to moderate hypertension showed that sitagliptin produced small reductions (–2 to –3 mm Hg) in 24-hour ambulatory BP measurements.47

Another DPP-4 inhibitor, saxagliptin, with efficacy similar to that described for sitagliptin, was recently approved by the US Food and Drug Administration (FDA) for treatment of T2DM.48

 

 

GLP-1 receptor agonists

Many of the GLP-1 receptor agonists developed or under development have glucoregulatory effects similar to GLP-1 but are resistant to degradation by DPP-4.28 Exenatide, an exendin-4 receptor agonist, has compared favorably with sitagliptin and with insulin analogues. Long-acting (once-weekly and once-daily) GLP-1 receptor agonists are under development.

In a 2-week, head-to-head study in metformin-treated patients with T2DM, exenatide had a greater effect than sitagliptin in lowering PPG and was more potent in increasing insulin secretion and reducing postprandial glucagon secretion. In contrast to sitagliptin, exenatide slowed gastric emptying and reduced caloric intake.49

In two studies of patients treated with exenatide, on a background of either metformin alone or metformin plus a sulfonylurea, patients who received metformin lost more weight (–1.6 to –2.8 kg; P ≤ .01) and experienced more significant decreases from baseline HbA1c (–0.4% to –0.8%; P < .002) at 30 weeks than did patients who received placebo.50,51 In a 16-week trial of exenatide in patients previously treated with a TZD with or without metformin, exenatide reduced HbA1c –0.98%, fasting blood glucose –1.69 mmol/L, and body weight –1.51 kg.52

When compared with insulin analogues, exenatide has been associated with weight loss (~ –3 kg) while the insulin analogues were associated with weight gain (~ +3 kg).53 After 26 weeks, body weight decreased –2.3 kg with exenatide and increased +1.8 kg with insulin glargine.54 Similar results were found in a crossover noninferiority trial, where the least-squares mean difference in weight change was significantly (P < .001) different (2.2 kg) between the treatments.55 When exenatide was compared with insulin aspart in an open-label, noninferiority trial, there was a between-group difference in weight of –5.4 kg after 52 weeks.32

Exenatide has also demonstrated these benefits in open-label extension studies. After 2 years, mean HbA1c reductions of –1.1% from baseline were sustained (P < .05), and weight loss of –4.7 kg was maintained (P < .001).56 After 82 weeks, similar HbA1c decreases (–1.1%) and weight loss (–4.4 kg) were exhibited.57 Even after 3 years, these benefits were maintained in patients who remained on the drug (HbA1c reduction from baseline, –1.0%; weight loss, –5.3 kg [P < .0001 for both]).58

Long-acting formulations of GLP-1 receptor agonists are in clinical development; two of these are once-weekly exenatide and once-daily liraglutide. Exenatide once weekly has the advantage of less frequent dosing and has elicited greater reductions in HbA1c than exenatide BID. After 15 weeks of once-weekly administration, the 0.8-mg formulation reduced HbA1c –1.4% and the 2-mg formulation reduced it –1.7% (P < .0001 for both compared with placebo). Body weight was lowered –3.8 kg (P < .05 compared with placebo) with the 2-mg formulation.59 Compared with exenatide BID, exenatide 2 mg once weekly showed greater reductions in HbA1c (–1.9% vs –1.5%; P = .0023) after 30 weeks of therapy.60 In a 1-year noncomparative trial, treatment with exenatide once weekly improved HbA1c (–2.0%) and weight (–4.1 kg), as well as BP and lipid profiles compared with baseline.61

Liraglutide, a once-daily human analogue GLP-1 receptor agonist, is under review by the FDA.28 In a 26-week study of patients with T2DM, liraglutide was associated with reductions in HbA1c (mean, –1.04%; P = 0.067 compared with insulin) and body weight (mean, –2.5 kg; P < .001 compared with insulin) at dosages of 0.6 to 1.8 mg/day SC. Liraglutide produced a decline in SBP from 0.6 to 3 mm Hg but was not associated with a decrease in DBP.62 In a 52-week study comparing liraglutide with glimepiride monotherapy, liraglutide 1.2 mg was associated with an HbA1c reduction of –0.84% (P = .0014) and the 1.8-mg dose with a reduction of –1.14% (P < .0001) compared with –0.51% for glimepiride. SBP decreased –0.7 mm Hg with glimepiride compared with –2.1 mm Hg for liraglutide 1.2 mg (P = .2912) and –3.6 mm Hg for liraglutide 1.8 mg (P < .0118). Mean DBP fell slightly but not significantly in all treatment groups.63 No effects on lipid parameters were reported in these two liraglutide studies.

The Liraglutide Effect and Action in Diabetes (LEAD-6) trial was undertaken to compare exenatide (10 mg BID SC) and liraglutide (1.8 mg/day SC) as add-on therapy to metformin, a sulfonylurea, or a combination of both in 464 patients with T2DM. After 26 weeks of treatment, liraglutide was associated with a significant reduction in HbA1c of –1.12%, compared with –0.79% with exenatide (P < .0001). Patients treated with liraglutide lost –3.2 kg while those on exenatide lost –2.9 kg. Among patients previously treated with metformin alone, there was a 1-kg difference in favor of liraglutide (P = NS).64

Safety profile

All of the drugs discussed have potential adverse effects. Metformin continues to have a black box warning for lactic acidosis.65 Sulfonylureas and insulin can cause hypoglycemia. TZDs can cause fluid retention and, in rare cases, CHF (for which these drugs also carry a black box warning).66,67 TZDs also increase the risk of distal fracture.66,67 The most common side effects of exenatide are gastrointestinal, but there have been reported cases of pancreatitis, some of which have been fatal.68,69 It has been difficult to prove whether exenatide increases the risk of pancreatitis, as patients with T2DM are already at an increased (three- to fourfold) risk for this condition compared with persons who do not have T2DM.69 Exenatide should not be used in patients with severe renal impairment or end-stage renal disease; it should be used with caution in patients who have undergone renal transplantation and in patients with moderate renal impairment.

The prescribing information for sitagliptin includes pancreatitis among the adverse reactions identified during the drug’s postapproval use.70 As with exenatide, it is not fully known whether a true association exists between the agent and pancreatitis. However, since pancreatitis can occur in this patient population, it is recommended that abdominal pain be fully evaluated to rule out pancreatitis. Continued postmarketing surveillance is important for all of these agents.

THE ROLE OF GUIDELINES

The American Association of Clinical Endocrinologists (AACE),26 the American Diabetes Association (ADA),71 and the ADA in conjunction with the European Association for the Study of Diabetes (EASD)24 have recently revised their recommendations for the management of patients with diabetes. The guidelines are unanimous in setting a glycemic goal (HbA1c < 7.0% for the ADA, HbA1c ≤ 6.5% for the AACE) and advocating individualized care for a treatment goal of HbA1c lower than 6.0% in patients who stand to benefit from near euglycemia without inducing severe hypoglycemia.24,26,71

CVD is the major cause of morbidity and mortality associated with T2DM and is a source of increasing concern.5 Accordingly, special consideration should be given to patients with coexisting CV risk factors, including hypertension and dyslipidemia. The ADA and the EASD advocate lifestyle modification to decrease body weight and the concurrent initiation of metformin as first-line therapy.24 If that strategy is insufficient, then two tiers of treatment guide the choice of next steps24:

  • Tier 1, in addition to metformin, includes the sulfonylureas and insulin. Although these are excellent glucose-lowering drugs, they are associated with weight gain, hypoglycemia, and no improvement in BP or lipid levels. They are relatively low in cost and have been used for many years. Their main drawback is evidence that despite their use, beta-cell failure continues unabated over time.
  • Tier 2 treatments include pioglitazone and the GLP-1 receptor agonist exenatide. Consideration may be given to the use of pioglitazone or exenatide when hypoglycemia is of concern, with exenatide being preferred when weight loss is a major objective and HbA1c is close to target (< 8.0%).24 Additionally, both the TZDs and exenatide probably help slow the rate of beta-cell failure, particularly if they are used early in the course of the disease.72,73 The AACE recommends different pharmacologic approaches based on HbA1c at diagnosis.26

The American Heart Association and the ADA have issued a joint scientific statement on the primary prevention of CVD in patients with diabetes.74 They advocate lifestyle management of body weight, nutrition, and physical activity.74 In addition, they stress the need for attention to BP, lipid levels, and smoking status, and the use of antiplatelet agents in patients at increased CV risk (> 40 years of age and a family history of CVD, hypertension, smoking, dyslipidemia, or albuminuria).

CONCLUSION

T2DM, weight gain/obesity, and CV risk present a continuing challenge to patients and clinicians. Anti­diabetes agents have varying degrees of evidence to support their effects on HbA1c, body weight, BP, and lipid levels. A better understanding of the pathophysiology of T2DM has led to the development of newer antidiabetes agents that target the fundamental defects of the disease. Evidence continues to accumulate for the improved benefits of glycemic control and weight loss in T2DM with GLP-1 receptor agonists such as exenatide currently having robust data in terms of beneficial effects on weight and CV risk factors. As clinicians continue to incorporate this knowledge into their practice patterns, patient adherence and clinical outcomes are expected to improve. Newer agents, such as incretin-based therapies, address T2DM as well as other factors that increase cardiometabolic risk through their effects not only on glycemic control but on body weight, BP, and lipids.

References
  1. Centers for Disease Control and Prevention (CDC). Prevalence of overweight and obesity among adults with diagnosed diabetes—United States, 1988–1994 and 1999–2002. MMWR Morb Mortal Wkly Rep 2004; 53:1066–1068.
  2. Prevalence of overweight and obesity among adults: United States 2003–2004. Centers for Disease Contral and Prevention Web site. http://www.cdc.gov/nchs/products/pubs/pubd/hestats/overweight/overwght_adult_03.htm. Published: April 2006. Accessed September 23, 2009.
  3. National Institute of Diabetes and Digestive and Kidney Diseases. National Diabetes Statistics, 2007 fact sheet. National Institutes of Health Web site. http://www.diabetes.niddk.nih.gov/dm/pubs/statistics/index.htm. Published 2008. Accessed September 16, 2009.
  4. Centers for Disease Control and Prevention (CDC). Prevalence of self-reported cardiovascular disease among persons aged >35 years with diabetes—United States, 1997–2005. MMWR Morb Mortal Wkly Rep 2007; 56:1129–1132.
  5. Haffner SM, Lehto S, Rönnemaa T, Pyörälä K, Laakso M. Mortality from coronary heart disease in subjects with type 2 diabetes and in nondiabetic subjects with and without prior myocardial infarction. N Engl J Med 1998; 339:229–234.
  6. Mulnier HE, Seaman HE, Raleigh VS, Soedamah-Muthu SS, Colhoun HM, Lawrenson RA. Mortality in people with type 2 diabetes in the UK. Diabet Med 2006; 23:516–521.
  7. Van Gaal LF, Gutkin SW, Nauck MA. Exploiting the antidiabetic properties of incretins to treat type 2 diabetes mellitus: glucagon-like peptide 1 receptor agonists or insulin for patients with inadequate glycemic control? Eur J Endocrinol 2008; 158:773–784.
  8. Bonora E, Targher G, Formentini G, et al. The metabolic syndrome is an independent predictor of cardiovascular disease in type 2 diabetic subjects: prospective data from the Verona Diabetes Complications Study. Diabet Med 2004; 21:52–58.
  9. Anderson JW, Kendall CW, Jenkins DJ. Importance of weight management in type 2 diabetes: review with meta-analysis of clinical studies. J Am Coll Nutr 2003; 22:331–339.
  10. Jenssen TG, Tonstad S, Claudi T, Midthejell K, Cooper J. The gap between guidelines and practice in the treatment of type 2 diabetes: a nationwide survey in Norway. Diabetes Res Clin Pract 2008; 80:314–320.
  11. UK Prospective Diabetes Study (UKPDS) Group. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). Lancet 1998; 352:837–853.
  12. UK Prospective Diabetes Study (UKPDS) Group. Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34). Lancet 1998; 352:854–865.
  13. Action to Control Cardiovascular Risk in Diabetes Study Group. Effects of intensive glucose lowering in type 2 diabetes. N Engl J Med 2008; 358:2545–2559.
  14. ADVANCE Collaborative Group. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N Engl J Med 2008; 358:2560–2572.
  15. Duckworth W, Abraira C, Moritz T, et al; for the VADT Investigators. Glucose control and vascular complications in veterans with type 2 diabetes. N Engl J Med 2009; 360:129–139.
  16. Skyler JS, Bergenstal R, Bonow RO, et al. Intensive glycemic control and the prevention of cardiovascular events: implications of the ACCORD, ADVANCE, and VA Diabetes Trials: a position statement of the American Diabetes Association and a scientific statement of the American College of Cardiology Foundation and the American Heart Association. Diabetes Care 2009; 32:187–192.
  17. Eeg-Olofsson K, Cederholm J, Nilsson PM, et al. Risk of cardiovascular disease and mortality in overweight and obese patients with type 2 diabetes: an observational study in 13,087 patients. Diabetologia 2009; 52:65–73.
  18. Look AHEAD Research Group. Reduction in weight and cardiovascular disease risk factors in individuals with type 2 diabetes: one-year results of the Look AHEAD trial. Diabetes Care 2007; 30:1374–1383.
  19. Peyrot M, Rubin RR, Lauritzen T, Snoek FJ, Matthews DR, Skovlund SE. Psychosocial problems and barriers to improved diabetes management: results of the Cross-National Diabetes Attitudes, Wishes and Needs (DAWN) Study. Diabet Med 2005; 22:1379–1385.
  20. Brod M, Cobden D, Lammert M, Bushnell D, Raskin P. Examining correlates of treatment satisfaction for injectable insulin in type 2 diabetes: lessons learned from a clinical trial comparing biphasic and basal analogues. Health Qual Life Outcomes 2007; 5:8.
  21. Sacco WP, Wells KJ, Friedman A, Matthew R, Perez S, Vaughan CA. Adherence, body mass index, and depression in adults with type 2 diabetes: the mediational role of diabetes symptoms and self-efficacy. Health Psychol 2007; 26:693–700.
  22. Ruelas V, Roybal GM, Lu Y, Goldman D, Peters A. Clinical and behavioral correlates of achieving and maintaining glycemic targets in an underserved population with type 2 diabetes. Diabetes Care 2009; 32:54–56.
  23. Nieves DJ, Cnop M, Retzlaff B, et al. The atherogenic lipoprotein profile associated with obesity and insulin resistance is largely attributable to intra-abdominal fat. Diabetes 2003; 52:172–179.
  24. Nathan DM, Buse JB, Davidson MB, et al. Medical management of hyperglycemia in type 2 diabetes: a consensus algorithm for the initiation and adjustment of therapy: a consensus statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care 2009; 32:193–203.
  25. Alexander GC, Sehgal NL, Moloney RM, Stafford RS. National trends in treatment of type 2 diabetes mellitus, 1994–2007. Arch Intern Med 2008; 168:2088–2094.
  26. AACE Diabetes Mellitus Clinical Practice Guidelines Task Force. American Association of Clinical Endocrinologists medical guidelines for clinical practice for the management of diabetes mellitus. Endocr Pract 2007; 13(suppl 1):S4–S68.
  27. Hermansen K, Mortensen LS. Bodyweight changes associated with antihyperglycaemic agents in type 2 diabetes mellitus. Drug Saf 2007; 30:1127–1142.
  28. Stonehouse A, Okerson T, Kendall D, Maggs D. Emerging incretin based therapies for type 2 diabetes: incretin mimetics and DPP-4 inhibitors. Curr Diabetes Rev 2008; 4:101–109.
  29. Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP. Gastroenterology 2007; 132:2131–2157.
  30. Nauck MA, Baller B, Meier JJ. Gastric inhibitory polypeptide and glucagon-like peptide-1 in the pathogenesis of type 2 diabetes. Diabetes 2004; 53(suppl 3):S190–S196.
  31. Toft-Nielsen MB, Madsbad S, Holst JJ. Determinants of the effectiveness of glucagon-like peptide-1 in type 2 diabetes. J Clin Endocrinol Metab 2001; 86:3853–3860.
  32. Nauck MA, Duran S, Kim D, et al. A comparison of twice-daily exenatide and biphasic insulin aspart in patients with type 2 diabetes who were suboptimally controlled with sulfonylurea and metformin: a non-inferiority study. Diabetologia 2007; 50:259–267.
  33. Nauck MA, Meininger G, Sheng D, Terranella L, Stein PP; for the Sitagliptin Study 024 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor, sitagliptin, compared with the sulfonylurea, glipizide, in patients with type 2 diabetes inadequately controlled on metformin alone: a randomized, double-blind, non-inferiority trial. Diabetes Obes Metab 2007; 9:194–205.
  34. Muscelli E, Mari A, Casolaro A, et al. Separate impact of obesity and glucose tolerance on the incretin effect in normal subjects and type 2 diabetic patients. Diabetes 2008; 57:1340–1348.
  35. Bastard JP, Maachi M, Lagathu C, et al. Recent advances in the relationship between obesity, inflammation, and insulin resistance. Eur Cytokine Netw 2006; 17:4–12.
  36. de Carvalho CP, Marin DM, de Souza AL, et al. GLP-1 and adiponectin: effect of weight loss after dietary restriction and gastric bypass in morbidly obese patients with normal and abnormal glucose metabolism. Obes Surg 2009; 19:313–320.
  37. Mingrone G. Role of the incretin system in the remission of type 2 diabetes following bariatric surgery. Nutr Metab Cardiovasc Dis 2008; 18:574–579.
  38. Raz I, Hanefeld M, Xu L, Caria C, Williams-Herman D, Khatami H; for the Sitagliptin Study 023 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy in patients with type 2 diabetes mellitus. Diabetologia 2006; 49:2564–2571.
  39. Scott R, Wu M, Sanchez M, Stein P. Efficacy and tolerability of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy over 12 weeks in patients with type 2 diabetes. Int J Clin Pract 2007; 61:171–180.
  40. Hanefeld M, Herman G, Wu M, Mickel C, Sanchez M, Stein PP; for the Sitagliptin Study 014 Investigators. Once-daily sitagliptin, a dipeptidyl peptidase-4 inhibitor, for the treatment of patients with type 2 diabetes. Curr Med Res Opin 2007; 23:1329–1339.
  41. Nonaka K, Kakikawa T, Sato A, et al. Efficacy and safety of sitagliptin monotherapy in Japanese patients with type 2 diabetes. Diabetes Res Clin Pract 2008; 79:291–298.
  42. Rosenstock J, Brazg R, Andryuk PJ, Lu K, Stein P; for the Sitagliptin Study 019 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin added to ongoing pioglitazone therapy in patients with type 2 diabetes: a 24-week, multicenter, randomized, double-blind, placebo-controlled, parallel-group study. Clin Ther 2006; 28:1556–1568.
  43. Hermansen K, Kipnes M, Luo E, Fanurik D, Khatami H, Stein P; for the Sitagliptin Study 035 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor, sitagliptin, in patients with type 2 diabetes mellitus inadequately controlled on glimepiride alone or on glimepiride and metformin. Diabetes Obes Metab 2007; 9:733–745.
  44. Aschner P, Kipnes MS, Lunceford JK, Sanchez M, Mickel C, Williams-Herman DE; for the Sitagliptin Study 021 Group. Effect of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy on glycemic control in patients with type 2 diabetes. Diabetes Care 2006; 29:2632–2637.
  45. Raz I, Chen Y, Wu M, et al. Efficacy and safety of sitagliptin added to ongoing metformin therapy in patients with type 2 diabetes. Curr Med Res Opin 2008; 24:537–550.
  46. Scott R, Loeys T, Davies MJ, Engel SS; for the Sitagliptin Study 801 Group. Efficacy and safety of sitagliptin when added to ongoing metformin therapy in patients with type 2 diabetes. Diabetes Obes Metab 2008; 10:959–969.
  47. Mistry GC, Maes AL, Lasseter KC, et al. Effect of sitagliptin, a dipeptidyl peptidase-4 inhibitor, on blood pressure in nondiabetic patients with mild to moderate hypertension. J Clin Pharmacol 2008; 48:592–598.
  48. US Department of Health and Human Services. FDA approves new drug treatment for type 2 diabetes. US Food and Drug Administration Web site. http://www.fda.gov/newsevents/newsroom/pressannouncements/ucm174780.htm. Published July 31, 2009. Accessed September 18, 2009.
  49. DeFronzo RA, Okerson T, Viswanathan P, Guan X, Holcombe JH, MacConell L. Effects of exenatide versus sitagliptin on postprandial glucose, insulin and glucagon secretion, gastric emptying, and caloric intake: a randomized, cross-over study. Curr Med Res Opin 2008; 24:2943–2952.
  50. DeFronzo RA, Ratner RE, Han J, Kim DD, Fineman MS, Baron AD. Effects of exenatide (exendin-4) on glycemic control and weight over 30 weeks in metformin-treated patients with type 2 diabetes. Diabetes Care 2005; 28:1092–1100.
  51. Kendall DM, Riddle MC, Rosenstock J, et al. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in patients with type 2 diabetes treated with metformin and a sulfonylurea. Diabetes Care 2005; 28:1083–1091.
  52. Zinman B, Hoogwerf BJ, Durán García S, et al. The effect of adding exenatide to a thiazolidinedione in suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med 2007; 146:477–485.
  53. Glass LC, Qu Y, Lenox S, et al. Effects of exenatide versus insulin analogues on weight change in subjects with type 2 diabetes: a pooled post-hoc analysis. Curr Med Res Opin 2008; 24:639–644.
  54. Heine RJ, Van Gaal LF, Johns D, Mihm MJ, Widel MH, Brodows RG; for the GWAA Study Group. Exenatide versus insulin glargine in patients with suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med 2005; 143:559–569.
  55. Barnett AH, Burger J, Johns D, et al. Tolerability and efficacy of exenatide and titrated insulin glargine in adult patients with type 2 diabetes previously uncontrolled with metformin or a sulfonylurea: a multinational, randomized, open-label, two-period, crossover noninferiority trial. Clin Ther 2007; 29:2333–2348.
  56. Buse JB, Klonoff DC, Nielsen LL, et al. Metabolic effects of two years of exenatide treatment on diabetes, obesity, and hepatic biomarkers in patients with type 2 diabetes: an interim analysis of data from the open-label, uncontrolled extension of three double-blind, placebo-controlled trials. Clin Ther 2007; 29:139–153.
  57. Blonde L, Klein EJ, Han J, et al. Interim analysis of the effects of exenatide treatment on A1C, weight and cardiovascular risk factors over 82 weeks in 314 overweight patients with type 2 diabetes. Diabetes Obes Metab 2006; 8:436–447.
  58. Klonoff DC, Buse JB, Nielsen LL, et al. Exenatide effects on diabetes, obesity, cardiovascular risk factors and hepatic biomarkers in patients with type 2 diabetes treated for at least 3 years. Curr Med Res Opin 2008; 24:275–286.
  59. Kim D, MacConell L, Zhuang D, et al. Effects of once-weekly dosing of a long-acting release formulation of exenatide on glucose control and body weight in subjects with type 2 diabetes. Diabetes Care 2007; 30:1487–1493.
  60. Drucker DJ, Buse JB, Taylor K, et al; for the DURATION-1 Study Group. Exenatide once weekly versus twice daily for the treatment of type 2 diabetes: a randomised, open-label, non-inferiority study. Lancet 2008; 372:1240–1250.
  61. Bergenstal RM, Kim T, Trautmann M, Zhuang D, Okerson T, Taylor K. Exenatide once weekly elicited improvements in blood pressure and lipid profile over 52 weeks in patients with type 2 diabetes. Circulation 2008; 118:S1086. Abstract 1239.
  62. Nauck M, Frid A, Hermansen K, et al; for the LEAD-2 Study Group. Efficacy and safety comparison of liraglutide, glimepiride, and placebo, all in combination with metformin, in type 2 diabetes: the LEAD (Liraglutide Effect and Action in Diabetes)-2 study. Diabetes Care 2009; 32:84–90.
  63. Garber A, Henry R. Ratner R, et al; for the LEAD-3 (Mono) Study Group. Liraglutide versus glimepiride monotherapy for type 2 diabetes (LEAD-3 Mono): a randomised, 52-week, phase III, double-blind, parallel-treatment trial. Lancet 2009; 373:473–481.
  64. Buse JB, Rosenstock J, Sesti G, et al; for the LEAD-6 Study Group. Liraglutide once a day versus exenatide twice a day for type 2 diabetes: a 26-week randomised, parallel-group, multinational, open-label trial (LEAD-6). Lancet 2009; 374:39–47.
  65. Fortamet [package insert]. Ft. Lauderdale, FL: Watson Laboratories; 2008.
  66. Actos. Physicians’ Desk Reference. 63rd edition. Montvale, NJ: Physicians’ Desk Reference Inc; 2008; 3077–3082.
  67. Avandia. Physicians’ Desk Reference. 63rd edition. Montvale, NJ: Physicians’ Desk Reference Inc; 2009; 1351–1359.
  68. Byetta [package insert]. San Diego, CA: Amylin Pharmaceuticals, Inc.; 2009.
  69. Idris I. News and views: FDA reviews incidences of acute pancreatitis in patients taking Byetta. Diabetes Obes Metab 2008; 10:96–98.
  70. Januvia [package insert]. Whitehouse Station, NJ: Merck & Co., Inc.; 2006, 2007.
  71. American Diabetes Association. Standards of medical care in diabetes—2009. Diabetes Care 2009; 32(suppl 1):S13–S61.
  72. Bunck MC, Diamant M, Cornér A, et al. One-year treatment with exenatide improves beta-cell function, compared with insulin glargine, in metformin-treated type 2 diabetic patients: a randomized, controlled trial. Diabetes Care 2009; 32:762–768.
  73. Gastaldelli A, Ferrannini E, Miyazaki Y, Matsuda M, Mari A, DeFronzo RA. Thiazolidinediones improve beta-cell function in type 2 diabetic patients. Am J Physiol Endocrinol Metab 2007; 292:E871–E883.
  74. Buse JB, Ginsberg HN, Bakris GL, et al. Primary prevention of cardiovascular diseases in people with diabetes mellitus: a scientific statement from the American Heart Association and the American Diabetes Association. Diabetes Care 2007; 30:162–172.
References
  1. Centers for Disease Control and Prevention (CDC). Prevalence of overweight and obesity among adults with diagnosed diabetes—United States, 1988–1994 and 1999–2002. MMWR Morb Mortal Wkly Rep 2004; 53:1066–1068.
  2. Prevalence of overweight and obesity among adults: United States 2003–2004. Centers for Disease Contral and Prevention Web site. http://www.cdc.gov/nchs/products/pubs/pubd/hestats/overweight/overwght_adult_03.htm. Published: April 2006. Accessed September 23, 2009.
  3. National Institute of Diabetes and Digestive and Kidney Diseases. National Diabetes Statistics, 2007 fact sheet. National Institutes of Health Web site. http://www.diabetes.niddk.nih.gov/dm/pubs/statistics/index.htm. Published 2008. Accessed September 16, 2009.
  4. Centers for Disease Control and Prevention (CDC). Prevalence of self-reported cardiovascular disease among persons aged >35 years with diabetes—United States, 1997–2005. MMWR Morb Mortal Wkly Rep 2007; 56:1129–1132.
  5. Haffner SM, Lehto S, Rönnemaa T, Pyörälä K, Laakso M. Mortality from coronary heart disease in subjects with type 2 diabetes and in nondiabetic subjects with and without prior myocardial infarction. N Engl J Med 1998; 339:229–234.
  6. Mulnier HE, Seaman HE, Raleigh VS, Soedamah-Muthu SS, Colhoun HM, Lawrenson RA. Mortality in people with type 2 diabetes in the UK. Diabet Med 2006; 23:516–521.
  7. Van Gaal LF, Gutkin SW, Nauck MA. Exploiting the antidiabetic properties of incretins to treat type 2 diabetes mellitus: glucagon-like peptide 1 receptor agonists or insulin for patients with inadequate glycemic control? Eur J Endocrinol 2008; 158:773–784.
  8. Bonora E, Targher G, Formentini G, et al. The metabolic syndrome is an independent predictor of cardiovascular disease in type 2 diabetic subjects: prospective data from the Verona Diabetes Complications Study. Diabet Med 2004; 21:52–58.
  9. Anderson JW, Kendall CW, Jenkins DJ. Importance of weight management in type 2 diabetes: review with meta-analysis of clinical studies. J Am Coll Nutr 2003; 22:331–339.
  10. Jenssen TG, Tonstad S, Claudi T, Midthejell K, Cooper J. The gap between guidelines and practice in the treatment of type 2 diabetes: a nationwide survey in Norway. Diabetes Res Clin Pract 2008; 80:314–320.
  11. UK Prospective Diabetes Study (UKPDS) Group. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). Lancet 1998; 352:837–853.
  12. UK Prospective Diabetes Study (UKPDS) Group. Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34). Lancet 1998; 352:854–865.
  13. Action to Control Cardiovascular Risk in Diabetes Study Group. Effects of intensive glucose lowering in type 2 diabetes. N Engl J Med 2008; 358:2545–2559.
  14. ADVANCE Collaborative Group. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N Engl J Med 2008; 358:2560–2572.
  15. Duckworth W, Abraira C, Moritz T, et al; for the VADT Investigators. Glucose control and vascular complications in veterans with type 2 diabetes. N Engl J Med 2009; 360:129–139.
  16. Skyler JS, Bergenstal R, Bonow RO, et al. Intensive glycemic control and the prevention of cardiovascular events: implications of the ACCORD, ADVANCE, and VA Diabetes Trials: a position statement of the American Diabetes Association and a scientific statement of the American College of Cardiology Foundation and the American Heart Association. Diabetes Care 2009; 32:187–192.
  17. Eeg-Olofsson K, Cederholm J, Nilsson PM, et al. Risk of cardiovascular disease and mortality in overweight and obese patients with type 2 diabetes: an observational study in 13,087 patients. Diabetologia 2009; 52:65–73.
  18. Look AHEAD Research Group. Reduction in weight and cardiovascular disease risk factors in individuals with type 2 diabetes: one-year results of the Look AHEAD trial. Diabetes Care 2007; 30:1374–1383.
  19. Peyrot M, Rubin RR, Lauritzen T, Snoek FJ, Matthews DR, Skovlund SE. Psychosocial problems and barriers to improved diabetes management: results of the Cross-National Diabetes Attitudes, Wishes and Needs (DAWN) Study. Diabet Med 2005; 22:1379–1385.
  20. Brod M, Cobden D, Lammert M, Bushnell D, Raskin P. Examining correlates of treatment satisfaction for injectable insulin in type 2 diabetes: lessons learned from a clinical trial comparing biphasic and basal analogues. Health Qual Life Outcomes 2007; 5:8.
  21. Sacco WP, Wells KJ, Friedman A, Matthew R, Perez S, Vaughan CA. Adherence, body mass index, and depression in adults with type 2 diabetes: the mediational role of diabetes symptoms and self-efficacy. Health Psychol 2007; 26:693–700.
  22. Ruelas V, Roybal GM, Lu Y, Goldman D, Peters A. Clinical and behavioral correlates of achieving and maintaining glycemic targets in an underserved population with type 2 diabetes. Diabetes Care 2009; 32:54–56.
  23. Nieves DJ, Cnop M, Retzlaff B, et al. The atherogenic lipoprotein profile associated with obesity and insulin resistance is largely attributable to intra-abdominal fat. Diabetes 2003; 52:172–179.
  24. Nathan DM, Buse JB, Davidson MB, et al. Medical management of hyperglycemia in type 2 diabetes: a consensus algorithm for the initiation and adjustment of therapy: a consensus statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care 2009; 32:193–203.
  25. Alexander GC, Sehgal NL, Moloney RM, Stafford RS. National trends in treatment of type 2 diabetes mellitus, 1994–2007. Arch Intern Med 2008; 168:2088–2094.
  26. AACE Diabetes Mellitus Clinical Practice Guidelines Task Force. American Association of Clinical Endocrinologists medical guidelines for clinical practice for the management of diabetes mellitus. Endocr Pract 2007; 13(suppl 1):S4–S68.
  27. Hermansen K, Mortensen LS. Bodyweight changes associated with antihyperglycaemic agents in type 2 diabetes mellitus. Drug Saf 2007; 30:1127–1142.
  28. Stonehouse A, Okerson T, Kendall D, Maggs D. Emerging incretin based therapies for type 2 diabetes: incretin mimetics and DPP-4 inhibitors. Curr Diabetes Rev 2008; 4:101–109.
  29. Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP. Gastroenterology 2007; 132:2131–2157.
  30. Nauck MA, Baller B, Meier JJ. Gastric inhibitory polypeptide and glucagon-like peptide-1 in the pathogenesis of type 2 diabetes. Diabetes 2004; 53(suppl 3):S190–S196.
  31. Toft-Nielsen MB, Madsbad S, Holst JJ. Determinants of the effectiveness of glucagon-like peptide-1 in type 2 diabetes. J Clin Endocrinol Metab 2001; 86:3853–3860.
  32. Nauck MA, Duran S, Kim D, et al. A comparison of twice-daily exenatide and biphasic insulin aspart in patients with type 2 diabetes who were suboptimally controlled with sulfonylurea and metformin: a non-inferiority study. Diabetologia 2007; 50:259–267.
  33. Nauck MA, Meininger G, Sheng D, Terranella L, Stein PP; for the Sitagliptin Study 024 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor, sitagliptin, compared with the sulfonylurea, glipizide, in patients with type 2 diabetes inadequately controlled on metformin alone: a randomized, double-blind, non-inferiority trial. Diabetes Obes Metab 2007; 9:194–205.
  34. Muscelli E, Mari A, Casolaro A, et al. Separate impact of obesity and glucose tolerance on the incretin effect in normal subjects and type 2 diabetic patients. Diabetes 2008; 57:1340–1348.
  35. Bastard JP, Maachi M, Lagathu C, et al. Recent advances in the relationship between obesity, inflammation, and insulin resistance. Eur Cytokine Netw 2006; 17:4–12.
  36. de Carvalho CP, Marin DM, de Souza AL, et al. GLP-1 and adiponectin: effect of weight loss after dietary restriction and gastric bypass in morbidly obese patients with normal and abnormal glucose metabolism. Obes Surg 2009; 19:313–320.
  37. Mingrone G. Role of the incretin system in the remission of type 2 diabetes following bariatric surgery. Nutr Metab Cardiovasc Dis 2008; 18:574–579.
  38. Raz I, Hanefeld M, Xu L, Caria C, Williams-Herman D, Khatami H; for the Sitagliptin Study 023 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy in patients with type 2 diabetes mellitus. Diabetologia 2006; 49:2564–2571.
  39. Scott R, Wu M, Sanchez M, Stein P. Efficacy and tolerability of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy over 12 weeks in patients with type 2 diabetes. Int J Clin Pract 2007; 61:171–180.
  40. Hanefeld M, Herman G, Wu M, Mickel C, Sanchez M, Stein PP; for the Sitagliptin Study 014 Investigators. Once-daily sitagliptin, a dipeptidyl peptidase-4 inhibitor, for the treatment of patients with type 2 diabetes. Curr Med Res Opin 2007; 23:1329–1339.
  41. Nonaka K, Kakikawa T, Sato A, et al. Efficacy and safety of sitagliptin monotherapy in Japanese patients with type 2 diabetes. Diabetes Res Clin Pract 2008; 79:291–298.
  42. Rosenstock J, Brazg R, Andryuk PJ, Lu K, Stein P; for the Sitagliptin Study 019 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin added to ongoing pioglitazone therapy in patients with type 2 diabetes: a 24-week, multicenter, randomized, double-blind, placebo-controlled, parallel-group study. Clin Ther 2006; 28:1556–1568.
  43. Hermansen K, Kipnes M, Luo E, Fanurik D, Khatami H, Stein P; for the Sitagliptin Study 035 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor, sitagliptin, in patients with type 2 diabetes mellitus inadequately controlled on glimepiride alone or on glimepiride and metformin. Diabetes Obes Metab 2007; 9:733–745.
  44. Aschner P, Kipnes MS, Lunceford JK, Sanchez M, Mickel C, Williams-Herman DE; for the Sitagliptin Study 021 Group. Effect of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy on glycemic control in patients with type 2 diabetes. Diabetes Care 2006; 29:2632–2637.
  45. Raz I, Chen Y, Wu M, et al. Efficacy and safety of sitagliptin added to ongoing metformin therapy in patients with type 2 diabetes. Curr Med Res Opin 2008; 24:537–550.
  46. Scott R, Loeys T, Davies MJ, Engel SS; for the Sitagliptin Study 801 Group. Efficacy and safety of sitagliptin when added to ongoing metformin therapy in patients with type 2 diabetes. Diabetes Obes Metab 2008; 10:959–969.
  47. Mistry GC, Maes AL, Lasseter KC, et al. Effect of sitagliptin, a dipeptidyl peptidase-4 inhibitor, on blood pressure in nondiabetic patients with mild to moderate hypertension. J Clin Pharmacol 2008; 48:592–598.
  48. US Department of Health and Human Services. FDA approves new drug treatment for type 2 diabetes. US Food and Drug Administration Web site. http://www.fda.gov/newsevents/newsroom/pressannouncements/ucm174780.htm. Published July 31, 2009. Accessed September 18, 2009.
  49. DeFronzo RA, Okerson T, Viswanathan P, Guan X, Holcombe JH, MacConell L. Effects of exenatide versus sitagliptin on postprandial glucose, insulin and glucagon secretion, gastric emptying, and caloric intake: a randomized, cross-over study. Curr Med Res Opin 2008; 24:2943–2952.
  50. DeFronzo RA, Ratner RE, Han J, Kim DD, Fineman MS, Baron AD. Effects of exenatide (exendin-4) on glycemic control and weight over 30 weeks in metformin-treated patients with type 2 diabetes. Diabetes Care 2005; 28:1092–1100.
  51. Kendall DM, Riddle MC, Rosenstock J, et al. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in patients with type 2 diabetes treated with metformin and a sulfonylurea. Diabetes Care 2005; 28:1083–1091.
  52. Zinman B, Hoogwerf BJ, Durán García S, et al. The effect of adding exenatide to a thiazolidinedione in suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med 2007; 146:477–485.
  53. Glass LC, Qu Y, Lenox S, et al. Effects of exenatide versus insulin analogues on weight change in subjects with type 2 diabetes: a pooled post-hoc analysis. Curr Med Res Opin 2008; 24:639–644.
  54. Heine RJ, Van Gaal LF, Johns D, Mihm MJ, Widel MH, Brodows RG; for the GWAA Study Group. Exenatide versus insulin glargine in patients with suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med 2005; 143:559–569.
  55. Barnett AH, Burger J, Johns D, et al. Tolerability and efficacy of exenatide and titrated insulin glargine in adult patients with type 2 diabetes previously uncontrolled with metformin or a sulfonylurea: a multinational, randomized, open-label, two-period, crossover noninferiority trial. Clin Ther 2007; 29:2333–2348.
  56. Buse JB, Klonoff DC, Nielsen LL, et al. Metabolic effects of two years of exenatide treatment on diabetes, obesity, and hepatic biomarkers in patients with type 2 diabetes: an interim analysis of data from the open-label, uncontrolled extension of three double-blind, placebo-controlled trials. Clin Ther 2007; 29:139–153.
  57. Blonde L, Klein EJ, Han J, et al. Interim analysis of the effects of exenatide treatment on A1C, weight and cardiovascular risk factors over 82 weeks in 314 overweight patients with type 2 diabetes. Diabetes Obes Metab 2006; 8:436–447.
  58. Klonoff DC, Buse JB, Nielsen LL, et al. Exenatide effects on diabetes, obesity, cardiovascular risk factors and hepatic biomarkers in patients with type 2 diabetes treated for at least 3 years. Curr Med Res Opin 2008; 24:275–286.
  59. Kim D, MacConell L, Zhuang D, et al. Effects of once-weekly dosing of a long-acting release formulation of exenatide on glucose control and body weight in subjects with type 2 diabetes. Diabetes Care 2007; 30:1487–1493.
  60. Drucker DJ, Buse JB, Taylor K, et al; for the DURATION-1 Study Group. Exenatide once weekly versus twice daily for the treatment of type 2 diabetes: a randomised, open-label, non-inferiority study. Lancet 2008; 372:1240–1250.
  61. Bergenstal RM, Kim T, Trautmann M, Zhuang D, Okerson T, Taylor K. Exenatide once weekly elicited improvements in blood pressure and lipid profile over 52 weeks in patients with type 2 diabetes. Circulation 2008; 118:S1086. Abstract 1239.
  62. Nauck M, Frid A, Hermansen K, et al; for the LEAD-2 Study Group. Efficacy and safety comparison of liraglutide, glimepiride, and placebo, all in combination with metformin, in type 2 diabetes: the LEAD (Liraglutide Effect and Action in Diabetes)-2 study. Diabetes Care 2009; 32:84–90.
  63. Garber A, Henry R. Ratner R, et al; for the LEAD-3 (Mono) Study Group. Liraglutide versus glimepiride monotherapy for type 2 diabetes (LEAD-3 Mono): a randomised, 52-week, phase III, double-blind, parallel-treatment trial. Lancet 2009; 373:473–481.
  64. Buse JB, Rosenstock J, Sesti G, et al; for the LEAD-6 Study Group. Liraglutide once a day versus exenatide twice a day for type 2 diabetes: a 26-week randomised, parallel-group, multinational, open-label trial (LEAD-6). Lancet 2009; 374:39–47.
  65. Fortamet [package insert]. Ft. Lauderdale, FL: Watson Laboratories; 2008.
  66. Actos. Physicians’ Desk Reference. 63rd edition. Montvale, NJ: Physicians’ Desk Reference Inc; 2008; 3077–3082.
  67. Avandia. Physicians’ Desk Reference. 63rd edition. Montvale, NJ: Physicians’ Desk Reference Inc; 2009; 1351–1359.
  68. Byetta [package insert]. San Diego, CA: Amylin Pharmaceuticals, Inc.; 2009.
  69. Idris I. News and views: FDA reviews incidences of acute pancreatitis in patients taking Byetta. Diabetes Obes Metab 2008; 10:96–98.
  70. Januvia [package insert]. Whitehouse Station, NJ: Merck & Co., Inc.; 2006, 2007.
  71. American Diabetes Association. Standards of medical care in diabetes—2009. Diabetes Care 2009; 32(suppl 1):S13–S61.
  72. Bunck MC, Diamant M, Cornér A, et al. One-year treatment with exenatide improves beta-cell function, compared with insulin glargine, in metformin-treated type 2 diabetic patients: a randomized, controlled trial. Diabetes Care 2009; 32:762–768.
  73. Gastaldelli A, Ferrannini E, Miyazaki Y, Matsuda M, Mari A, DeFronzo RA. Thiazolidinediones improve beta-cell function in type 2 diabetic patients. Am J Physiol Endocrinol Metab 2007; 292:E871–E883.
  74. Buse JB, Ginsberg HN, Bakris GL, et al. Primary prevention of cardiovascular diseases in people with diabetes mellitus: a scientific statement from the American Heart Association and the American Diabetes Association. Diabetes Care 2007; 30:162–172.
Page Number
S20-S27
Page Number
S20-S27
Publications
Publications
Article Type
Display Headline
Patient and treatment perspectives: Revisiting the link between type 2 diabetes, weight gain, and cardiovascular risk
Display Headline
Patient and treatment perspectives: Revisiting the link between type 2 diabetes, weight gain, and cardiovascular risk
Citation Override
Cleveland Clinic Journal of Medicine 2009 December;76(suppl 5):S20-S27
Inside the Article

KEY POINTS

  • Control of cardiovascular risk factors is as important as glycemic control in patients with T2DM.
  • Intensive glucose control has shown mixed results in terms of correlation with improved cardiovascular risk factors.
  • Newer agents target the fundamental pathophysiologic defects of T2DM, with beneficial effects on weight and other cardiovascular risk factors.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Fri, 04/13/2018 - 09:15
Un-Gate On Date
Fri, 04/13/2018 - 09:15
Use ProPublica
CFC Schedule Remove Status
Fri, 04/13/2018 - 09:15
Article PDF Media

Advances in therapy for type 2 diabetes: GLP–1 receptor agonists and DPP–4 inhibitors

Article Type
Changed
Tue, 10/02/2018 - 11:45
Display Headline
Advances in therapy for type 2 diabetes: GLP–1 receptor agonists and DPP–4 inhibitors

The prevalence of type 2 diabetes mellitus (T2DM) is increasing exponentially worldwide. According to the Centers for Disease Control and Prevention, more than 23 million Americans had diabetes in 2007.1 Globally, the prevalence of diabetes, of which T2DM accounts for 90% to 95% of cases,1 is expected to increase from 171 million in 2000 to 366 million in 2030.2 The National Health and Nutrition Examination Survey (NHANES) showed that about 66% of Americans were overweight or obese between 2003–2004.3 Data from a Swedish National Diabetes Register study showed both overweight and obesity as independent risk factors for cardiovascular disease (CVD) in patients with T2DM.4

This article presents an overview of the evolving concepts of the pathophysiology of T2DM, with a focus on two new therapeutic classes: the glucagon-like peptide–1 (GLP-1) receptor agonists and the dipeptidyl peptidase–4 (DPP-4) inhibitors.

THE PATHOPHYSIOLOGY OF T2DM

The American Association of Clinical Endocrinologists (AACE) describes T2DM as “a progressive, complex metabolic disorder characterized by coexisting defects of multiple organ sites including insulin resistance in muscle and adipose tissue, a progressive decline in pancreatic insulin secretion, unrestrained hepatic glucose production, and other hormonal deficiencies.”5 Other defects include accelerated gastric emptying in patients with T2DM, especially those who are obese or who have the disease for a long duration.6,7

Hormonal deficiencies in T2DM are related to abnormalities in the secretion of the beta-cell hormone amylin, the alpha-cell hormone glucagon, and the incretin hormones GLP-1 and glucose-dependent insulinotropic polypeptide (GIP).8,9 In addition to the triumvirate of core defects associated with T2DM (involvement of the pancreatic beta cell, muscle, and liver), other mechanisms of disease onset have been advanced, including accelerated lipolysis, hyperglucagonemia, and incretin deficiency/resistance.9 Also, the rate of basal hepatic glucose production is markedly increased in patients with T2DM, which is closely correlated with elevations in fasting plasma glucagon concentration.9

The incretin effect—the intestinal augmentation of secretion of insulin—attributed to GLP-1 and GIP is reduced in patients with T2DM.10 The secretion of GIP may be normal or elevated in patients with T2DM while the secretion of GLP-1 is deficient; however, cellular responsiveness to GLP-1 is preserved while responsiveness to GIP is diminished.11

Both endogenous and exogenous GLP-1 and GIP are degraded in vivo and in vitro by the enzyme DPP-4,12
a ubiquitous, membrane-spanning, cell-surface aminopeptidase that preferentially cleaves peptides with a proline or alanine residue in the second amino-terminal position. DPP-4 is widely expressed (eg, in the liver, lungs, kidney, lymphocytes, epithelial cells, endothelial cells). The role of DPP-4 in the immune system stems from its exopeptidase activity and its interactions with various molecules, including cyto­kines and chemokines.13

INCRETIN-BASED THERAPIES: GLP-1 RECEPTOR AGONISTS AND DPP-4 INHIBITORS

Exenatide is a GLP-1 receptor agonist that is resistant to DPP-4 degradation. Based on preclinical studies, exenatide, which shares a 53% amino acid sequence identity with human GLP-1, is approximately 5,500 times more potent than endogenous GLP-1 in glucose lowering.14,15 Among the acute actions of exenatide is glucose-dependent insulinotropism, the end result of which may be a reduced risk of hypoglycemia.16 This contrasts with insulin secretagogues (eg, sulfonylureas), which increase insulin secretion regardless of glucose concentrations.

Exenatide received US Food and Drug Administration (FDA) approval in 2005 and is indicated for the treatment of patients with T2DM.13,17 Exenatide is administered BID as a subcutaneous (SC) injection in doses of 5 or 10 μg within 1 hour before the two major meals of the day, which should be eaten about 6 hours apart.18

Approved in 2006, sitagliptin was the first DPP-4 inhibitor indicated for adjunctive therapy to lifestyle modifications for the treatment of patients with T2DM.17 The recommended dosage of oral sitagliptin is 100 mg QD. A single-tablet formulation of the combination of sitagliptin and metformin was approved by the FDA in 2007.19 Another DPP-4 inhibitor, saxagliptin, was approved in July 2009 for treatment of patients with T2DM either as monotherapy or in combination with metformin, sulfonylurea, or a thiazolidinedione (TZD).20 The DPP-4 inhibitor vildagliptin is approved in the European Union and Latin America but not in the United States. Vildagliptin is available as a 50- or 100-mg daily dosage; it has been recommended for use at 50 mg QD in combination with a sulfonylurea or at 50 mg BID with either metformin or a TZD.18

GLP-1 RECEPTOR AGONISTS AND DPP-4 INHIBITORS IN DEVELOPMENT

Exenatide is currently being evaluated as a once-weekly formulation.21,22 Compared with the BID formulation, exenatide once weekly has been shown to produce significantly greater improvements in glycemic control, with similar reductions in body weight and no increased risk of hypoglycemia.21

Also undergoing regulatory review is the partly DPP-4–resistant acylated GLP-1 receptor agonist liraglutide.13 Liraglutide, a human analogue GLP-1 receptor agonist, has 97% linear amino acid sequence homology to human GLP-1.23,24 Based on its prolonged degradation time and resulting 10- to 14-hour half-life, liraglutide is anticipated to be dosed once daily.13,25,26

Other GLP-1 receptor agonists and DPP-4 inhibitors are in varying stages of development.27 Albiglutide is a long-acting GLP-1 receptor agonist that is generated by the genetic fusion of a DPP-4–resistant GLP-1 to human albumin. Based on pharmacokinetic studies, albiglutide has a half-life of 6 to 8 days. AVE0010, an exendin-4-based GLP-1 receptor agonist, was shown in a 28-day T2DM clinical trial to have an affinity four times greater than native GLP-1 for the human GLP-1 receptor.27 Taspoglutide (R1583), a human analogue GLP-1 receptor agonist, was evaluated in three randomized, placebo-controlled studies as a GLP-1 receptor agonist. Alogliptin, a DPP-4 inhibitor currently in development, has been shown to be safe and effective in studies as monotherapy and in combination with other antidiabetes agents.28–30

 

 

CLINICAL TRIALS: GLP-1 RECEPTOR AGONISTS AND DPP-4 INHIBITORS

This section summarizes clinical trials of GLP-1 receptor agonists and DPP-4 inhibitors. The summary is based on literature published from 2005 to 2009 relevant to phase 3 or 4 T2DM clinical trials with currently available agents, or agents with pending new drug applications.

Table 1 summarizes the data on the effects of the GLP-1 receptor agonists on glucose lowering based on glycosylated hemoglobin (HbA1c) mean changes from baseline, body weight, and hypoglycemia. Eleven studies were identified for exenatide, including three pivotal trials,31–33 three insulin-comparator studies,34–36 one long-term study,37 one monotherapy study (a use for which it is not currently indicated),38 one head-to-head study with a DPP-4 inhibitor,39 and two studies with exenatide once weekly (which is currently investigational).21,22 Five primary efficacy studies with liraglutide were also identified.23,25,26,40,41

Table 2 summarizes the corresponding data for the DPP-4 inhibitors. Ten studies with sitagliptin were identified, including four monotherapy studies,42–45 one head-to-head study with a GLP-1 receptor agonist,39 and five studies in which sitagliptin was used in combination or as add-on therapy.46–50 Five saxagliptin studies are reviewed, including two in which saxagliptin was used in combination with metformin and one in combination with glyburide.51–55 Six studies with vildagliptin were reviewed,56–61 but no trials specific to the single-tablet formulation of sitagliptin plus metformin were identified.

Effects on HbA1c and weight

GLP-1 receptor agonists reduced HbA1c. Based on the studies reviewed in Table 1, exenatide BID reduced baseline HbA1c by a maximum of –1.5% at 30 weeks.21,31,32 Exenatide has demonstrated sustained reductions in HbA1c of –0.8% for up to 3.5 years in an open-label extension trial.37 Even greater reductions in HbA1c (–1.4% at 15 weeks and –1.9% at 30 weeks) have been reported with the once-weekly formulation under clinical development.21,22 Liraglutide, another GLP-1 receptor agonist under development, has reported HbA1c reductions from baseline up to –1.67% at 14 weeks,40,41 up to –1.1% at 26 weeks,23,26 and up to –1.14% at 52 weeks.25 The reductions quoted generally refer to means, and individual patients may have greater or lesser responses. Also, baseline HbA1c is a significant determinant of the potential HbA1c reduction. Higher baseline values drop more significantly than do baseline values that are closer to normal.

Weight reduction with GLP-1 receptor agonists. In addition to effective glucose lowering, the GLP-1 receptor agonists, particularly exendin-4 agonists, produced beneficial effects on weight (Table 1). Exenatide BID elicited mean weight reductions up to –3.6 kg at 30 weeks21,31,32 and –5.3 kg at 3.5 years.37 Exenatide once weekly resulted in mean weight reductions of up to –3.8 kg at 15 weeks22 and –3.7 kg at 30 weeks.21 Effects on weight with liraglutide varied from a mean reduction of up to –2.99 kg to a slight gain of up to +0.13 kg at 14 weeks40,41 and with weight loss of up to –2.8 kg at 26 weeks23,26 and up to –2.5 kg at 52 weeks.25 In this review, only exenatide has been assessed in insulin-comparator studies, where it was shown to reduce weight compared with the insulin analogues, which led to weight gain.34–36

Hypoglycemia. Patients receiving exenatide experienced lower rates of hypoglycemia (up to 17%) than patients treated with either insulin glargine or insulin aspart (~25%).34,36 The rate of hypoglycemia with exenatide is comparable to that seen with metformin (up to 21%) in a systematic review of oral antidiabetes agents conducted by the Agency for Healthcare Research and Quality.62 No major hypoglycemic events were reported in the liraglutide studies reviewed. The incidence of hypoglycemia reported with DPP-4 inhibitors (Table 2) is also low (2% or less in most studies). The glucose-dependent mechanisms of the incretin-based therapies minimizes the risk of hypoglycemia.

DPP-4 inhibitors and sustained HbA1c reduction. The effects of the DPP-4 inhibitors on HbA1c and weight, either as monotherapy or in combination with other agents, were evaluated in studies ranging in duration from 12 to 52 weeks (Table 2). No studies were identified that compared the glycemic control effects of DPP-4 inhibitors and insulin analogues. Sitagliptin led to a mean reduction in HbA1c from baseline of up to –0.65% at 12 weeks,43,45 up to –0.48% at 18 weeks,44 up to –0.85% at 24 weeks,42,46,47,50 up to –1.0% at 30 weeks,49 and up to –0.67% at 52 weeks.48 Saxagliptin mean reductions in HbA1c ranged from –0.43% to –1.17%.51–54 Data from four 24-week T2DM studies56–60 showed vildagliptin reducing HbA1c up to –1.4% at 24 weeks, with the greatest reduction in a study that involved drug-naïve patients with a relatively short duration of disease (mean, 1.2 years).59 Reductions in HbA1c of –1.0% were sustained in a 52-week study61 and its 52-week extension.58

DPP-4 inhibitors: weight neutral. The DPP-4 inhibitors appear to have a weight-neutral effect (Table 2). The effects of sitagliptin on weight ranged from a loss of –1.5 kg48 at 52 weeks to a gain of +1.8 kg at 24 weeks.50 Weight changes with saxagliptin ranged from a mean reduction of –1.8 kg53 to a gain of +0.7 kg.51 Two vildagliptin studies showed varying effects on weight ranging from a loss of up to –1.8 kg from baseline56 to a gain of up to +1.3 kg57 relative to placebo, both at 24 weeks.

Potential for CV risk reduction

Potentially beneficial effects on CV risk factors, including blood pressure (ie, reduction) and lipid concentrations (ie, differential effects on low-density lipoprotein and high-density lipoprotein cholesterol), were identified in seven GLP-1 receptor studies—three with exenatide (two with exenatide BID,37,38 and one with the investigational exenatide once weekly21) and four with liraglutide.23,25,26,41 For the DPP-4 inhibitors, three studies were identified—two with sitagliptin45,50 and one with vildagliptin61—in which potentially beneficial effects on CV risk factors were demonstrated.The data have been encouraging, although the clinical implications have yet to be fully understood.

Head-to-head comparison

A recent study compared the effects of the GLP-1 receptor agonist exenatide and the DPP-4 inhibitor sitagliptin on postprandial glucose (PPG) concentrations, insulin and glucagon secretion, gastric intake, and caloric intake.39 Although limited by a short treatment duration (2 weeks), the study showed that the GLP-1 receptor agonist had a greater effect than the DPP-4 inhibitor in reducing PPG concentrations, a more potent effect in increasing insulin secretion and decreasing postprandial glucagon secretion, and a relatively greater effect in reducing caloric intake; and that it decreased the rate of gastric emptying (sitagliptin had no effect). These differences suggest that exenatide may provide a greater degree of GLP-1 receptor activation than the more physiologic concentrations of GLP-1 reached with DPP-4 inhibition.39 Results of a scintigraphic study showed that exenatide substantially slows the gastric emptying that is accelerated in patients with T2DM. This could be another beneficial mechanism in treating postprandial glycemia.63

Adverse effects

Exenatide has shown effects on hepatic injury markers (ie, improvement in alanine and aspartate aminotransferases) for up to 3.5 years of treatment.37 For the GLP-1 receptor agonist and DPP-4 inhibitor studies reviewed, the adverse events were generally mild and included nausea and vomiting, nasopharyngitis, and mild hypoglycemia.

 

 

Meta-analysis conclusions

The published clinical trial data presented in this review expand the body of evidence on the safety and efficacy of incretin-based therapy in patients with T2DM. These data include the results of a meta-analysis by Amori et al,17 which examined randomized controlled trials of 12 weeks’ or longer duration that compared incretin-based therapy with placebo or other diabetes medications and reported HbA1c changes in adults with T2DM. The meta-analysis showed that incretin-based therapies reduced HbA1c more than placebo (weighted mean difference, –0.97% [95% confidence interval (CI), –1.13% to –0.81%] for GLP-1 receptor agonists and –0.74% [95% CI, –0.85% to –0.62%] for DPP-4 inhibitors) and were noninferior to other antidiabetes agents. Treatment with a GLP-1 receptor agonist (ie, exenatide) caused weight loss (–1.4 kg and –4.8 kg vs placebo and insulin, respectively) while DPP-4 inhibitors (ie, sitagliptin, vildagliptin) were weight neutral.17

Beta-cell function

Evidence regarding the effects of incretin-based therapies, particularly the exendin-4 GLP-1 receptor agonists, on beta-cell function in patients with T2DM continues to accumulate. When assessing long-term (1 year) exenatide treatment in patients with T2DM, a trial (n = 69) comparing exenatide with the basal insulin analogue insulin glargine showed that exenatide and insulin glargine resulted in similar reductions in HbA1c (–0.8% vs –0.7%; P = .55).64 However, exenatide significantly reduced body weight while insulin glargine resulted in weight gain (–3.6 kg vs +1.0 kg; P < .0001). In terms of beta-cell function, arginine-stimulated C-peptide secretion during hyperglycemia increased 2.46-fold from baseline after 52 weeks of exenatide treatment compared with 1.31-fold with insulin glargine treatment (P < .0001).64

With respect to the direct beta-cell effects of liraglutide, a preclinical study reported that liraglutide improved glucose homeostasis in marginal mass islet transplantation in diabetic mice.65 In this study, liraglutide was shown, in a mouse model, to reduce the time to normoglycemia after islet cell transplantation (median time, 1 vs 72.5 days; P < .0001). The effects of liraglutide on beta-cell function also were assessed in 13 patients with T2DM. After 7 days of treatment, liraglutide improved beta-cell function, which was associated with improvement in glucose concentration.66 Liraglutide improved potentiation of insulin secretion during the first meal, owing in part to restoration of the potentiation peak (which is markedly blunted in T2DM), in a phenomenon similar to that observed with exenatide.67

Beneficial effects on beta-cell function have also been reported with DPP-4 inhibitors. In a model-based analysis of patients with T2DM, it was shown that sitagliptin improved basal, static, and dynamic responsiveness of pancreatic beta cells to glucose. The results were observed when sitagliptin was administered both as an add-on to metformin therapy and as monotherapy.68 A 52-week, double-blind, randomized, parallel-group study compared vildagliptin 50 mg/day and placebo in 306 patients with T2DM and mild hyperglycemia (HbA1c, 6.2% to 7.5%). Vildagliptin was shown to significantly increase fasting insulin secretory tone, glucose sensitivity, and rate sensitivity, all of which are aspects of beta-cell function.69

Summary

Based on the ability of incretin-based therapies to address various disease mechanisms, including beta-cell defects (ie, hyperglycemia), hormone-related abnormalities (ie, hyperglucagonemia, incretin deficiency/resistance), and accelerated gastric emptying (especially with GLP-1 receptor agonists); their favorable effects on weight (reduction with GLP-1 receptor agonists and neutral with DPP-4 inhibitors); their beneficial effects on CV risk factors; and their good safety profile (ie, hypoglycemia risk comparable with metformin), these agents could be considered therapeutic advances for the treatment of patients with T2DM.

INCRETIN-BASED THERAPIES IN GUIDELINES AND ALGORITHMS

The 2007 AACE medical guidelines for clinical practice for the management of diabetes recognized the place of the incretin-based therapies and included them among the pharmacologic options.5 Exenatide was specifically recommended for combination therapy with metformin, a sulfonylurea (secretagogue), a sulfonylurea plus metformin, or a TZD. Sitagliptin was recommended for use as monotherapy or in combination with metformin or a TZD.5

In 2009, the American Diabetes Association (ADA) and the European Association for the Study of Diabetes convened a consensus panel to produce an algorithm for the initiation and adjustment of therapy for patients with T2DM. In this algorithm, GLP-1 receptor agonists were considered appropriate in certain clinical scenarios (eg, when hypoglycemia was an issue or weight loss was a major consideration during treatment). However, the groups also noted a need for more data on long-term safety and the cost of treatment with incretin-based therapies.70

The AACE and the American College of Endocrinology recently developed “road maps” for managing patients with T2DM. In patients with T2DM who are naïve to therapy, DPP-4 inhibitors are among the recommended first options when the initial HbA1c is 6.0% to 7.0% and as a combination therapy component when HbA1c reaches 7.0% to 9.0%. In patients who have already received monotherapy for 2 to 3 months and whose HbA1c is 6.5% to 8.5%, treatment options include combination therapy with a DPP-4 inhibitor and metformin or a TZD. Another option includes the initiation of treatment with a GLP-1 receptor agonist in combination with a TZD, with metformin or a sulfonylurea, or with metformin and a sulfonylurea.71

The role of GLP-1 receptor agonist therapies and their incorporation into T2DM treatment algorithms was noted at the 2008 annual meeting of the ADA. In the Banting lecture, Ralph A. DeFronzo, MD, advocated the early use of triple-drug therapy with metformin, exenatide, and a TZD in the management of patients with T2DM.9

CONCLUSION

T2DM, which is linked to weight gain and obesity, is a complex disease that predisposes patients to and is associated with CVD. A better understanding and appreciation of the role of the incretin system in the pathogenesis of T2DM has led to the development of incretin-based therapies, such as the GLP-1 receptor agonists and DPP-4 inhibitors. As more experimental and clinical evidence becomes available, subtle nuances are emerging that distinguish the roles of these two therapeutic classes.

References
  1. 2007 National Diabetes Fact Sheet. Centers for Disease Control and Prevention Web site. http://www.cdc.gov/diabetes/pubs/estimates07.htm. Updated: July 23, 2008. Accessed September 25, 2009.
  2. Wild S, Roglic G, Green A, Sicree R, King H. Global prevalence of diabetes: estimates for the year 2000 and projections for 2030. Diabetes Care 2004; 27:1047–1053.
  3. Prevalence of overweight and obesity among adults: United States 2003–2004. Centers for Disease Control and Prevention Web site. http://www.cdc.gov/nchs/products/pubs/pubd/hestats/overweight/overwght_adult_03.htm. Published: April 2006. Accessed September 23, 2009.
  4. Eeg-Olofsson K, Cederholm J, Nilsson PM, et al. Risk of cardiovascular disease and mortality in overweight and obese patients with type 2 diabetes: an observational study in 13,087 patients. Diabetologia 2009; 52:65–73.
  5. AACE Diabetes Mellitus Clinical Practice Guidelines Task Force. American Association of Clinical Endocrinologists medical guidelines for clinical practice for the management of diabetes mellitus. Endocr Pract 2007; 13(suppl 1):S4–S68.
  6. Bertin E, Schneider N, Abdelli N, et al. Gastric emptying is accelerated in obese type 2 diabetic patients without autonomic neuropathy. Diabetes Metab 2001; 27:357–364.
  7. Weytjens C, Keymeulen B, Van Haleweyn C, Somers G, Bossuyt A. Rapid gastric emptying of a liquid meal in long-term type 2 diabetes mellitus. Diabet Med 1998; 15:1022–1027.
  8. Stonehouse AH, Holcombe JH, Kendall DM. GLP-1 analogues, DPP-IV inhibitors and the metabolic syndrome. In: Fonseca V, ed. Therapeutic Strategies in Metabolic Syndrome. Oxford, UK: Atlas Medical Publishing Ltd; 2008: 137–157.
  9. DeFronzo RA. From the triumvirate to the ominous octet: a new paradigm for the treatment of type 2 diabetes mellitus. Diabetes 2009; 58:773–795.
  10. Nauck M, Stöckmann F, Ebert R, Creutzfeldt W. Reduced incretin effect in type 2 (non-insulin-dependent) diabetes. Diabetologia 1986; 29:46–52.
  11. Nauck MA, Heimesaat MM, Orskov C, Holst JJ, Ebert R, Creutzfeldt W. Preserved incretin activity of glucagon-like peptide 1 [7-36 amide] but not of synthetic human gastric inhibitory polypeptide in patients with type-2 diabetes mellitus. J Clin Invest 1993; 91:301–307.
  12. Deacon CF, Johnsen AH, Holst JJ. Degradation of glucagon-like peptide-1 by human plasma in vitro yields an N-terminally truncated peptide that is a major endogenous metabolite in vivo. J Clin Endocrinol Metab 1995; 80:952–957.
  13. Drucker DJ, Nauck MA. The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 2006; 368:1696–1705.
  14. Eng J, Kleinman WA, Singh L, Singh G, Raufman J-P. Isolation and characterization of exendin-4, an exendin-3 analogue, from Heloderma suspectum venom: further evidence for an exendin receptor on dispersed acini from guinea pig pancreas. J Biol Chem 1992; 267:7402–7405.
  15. Young AA, Gedulin BR, Bhavsar S, et al. Glucose-lowering and insulin-sensitizing actions of exendin-4: studies in obese diabetic (ob/ob, db/db) mice, diabetic fatty Zucker rats, and diabetic rhesus monkeys (Macaca mulatta). Diabetes 1999; 48:1026–1034.
  16. Kolterman OG, Buse JB, Fineman MS, et al. Synthetic exendin-4 (exenatide) significantly reduces postprandial and fasting plasma glucose in subjects with type 2 diabetes. J Clin Endocrinol Metab 2003; 88:3082–3089.
  17. Amori RE, Lau J, Pittas AG. Efficacy and safety of incretin therapy in type 2 diabetes: systematic review and meta-analysis. JAMA 2007; 298:194–206.
  18. Davidson JA, Parente EB, Gross JL. Incretin mimetics and dipeptidyl peptidase-4 inhibitors: innovative treatment therapies for type 2 diabetes. Arq Bras Endocrinol Metabol 2008; 52:1039–1049.
  19. Alexander GC, Sehgal NL, Moloney RM, Stafford RS. National trends in treatment of type 2 diabetes mellitus, 1994–2007. Arch Intern Med 2008; 168:2088–2094.
  20. US Department of Health and Human Services. FDA approves new drug treatment for type 2 diabetes. US Food and Drug Administration Web site. http://www.fda.gov/newsevents/newsroom/pressannouncements/ucm174780.htm. Published July 31, 2009. Accessed September 18, 2009.
  21. Drucker DJ, Buse JB, Taylor K, et al; for the DURATION-1 Study Group. Exenatide once weekly versus twice daily for the treatment of type 2 diabetes: a randomised, open-label, non-inferiority study. Lancet 2008; 372:1240–1250.
  22. Kim D, MacConell L, Zhuang D, et al. Effects of once-weekly dosing of a long-acting release formulation of exenatide on glucose control and body weight in subjects with type 2 diabetes. Diabetes Care 2007; 30:1487–1493.
  23. Marre M, Shaw J, Brändle M, et al; for the LEAD-1 SU Study Group. Liraglutide, a once-daily human GLP-1 analogue, added to a sulphonylurea over 26 weeks produces greater improvements in glycaemic and weight control compared with adding rosiglitazone or placebo in subjects with type 2 diabetes (LEAD-1 SU). Diabetes Med 2009; 26:268–278.
  24. Knudsen LB, Nielsen PF, Huusfeldt PO, et al. Potent derivatives of glucagon-like peptide 1 with pharmacokinetic properties suitable for once-daily administration. J Med Chem 2000; 43:1664–1669.
  25. Garber A, Henry R, Ratner R, et al; for the LEAD-3 (Mono) Study Group. Liraglutide versus glimepiride monotherapy for type 2 diabetes (LEAD-3 Mono): a randomised, 52-week, phase III, double-blind, parallel-treatment trial. Lancet 2009; 373:473–481.
  26. Nauck M, Frid A, Hermansen K, et al; for the LEAD-2 Study Group. Efficacy and safety comparison of liraglutide, glimepiride, and placebo, all in combination with metformin, in type 2 diabetes: the LEAD (Liraglutide Effect and Action in Diabetes)-2 study. Diabetes Care 2009; 32:84–90.
  27. Baggio LL, Drucker DJ, Maida A, Lamont BJ. ADA 2008: incretin-based therapeutics. MedscapeCME Web site. http://www.medscape.com/viewprogram/15786. Accessed September 18, 2009.
  28. Fleck P, Christopher R, Covington P, Wilson C, Mekki Q. Efficacy and safety of alogliptin monotherapy over 12 weeks in patients with type 2 diabetes. Paper presented at: 68th Annual Meeting of the American Diabetes Association; June 6–10, 2008; San Francisco, CA. Abstract 479-P.
  29. DeFronzo RA, Burant CF, Fleck P, Wilson C, Mekki Q, Pratley RE. Effect of alogliptin combined with pioglitazone on glycemic control in metformin-treated patients with type 2 diabetes. Paper presented at: 69th Annual Meeting of the American Diabetes Association; June 5–9, 2009; New Orleans, LA. Abstract 2024-PO.
  30. Nauck M, Ellis G, Fleck P, Wilson C, Mekki Q. Efficacy and safety of alogliptin added to metformin therapy in patients with type 2 diabetes. Paper presented at: 68th Annual Meeting of the American Diabetes Association; June 6–10, 2008; San Francisco, CA. Abstract 477-P.
  31. DeFronzo RA, Ratner RE, Han J, Kim DD, Fineman MS, Baron AD. Effects of exenatide (exendin-4) on glycemic control and weight over 30 weeks in metformin-treated patients with type 2 diabetes. Diabetes Care 2005; 28:1092–1100.
  32. Kendall DM, Riddle MC, Rosenstock J, et al. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in patients with type 2 diabetes treated with metformin and a sulfonylurea. Diabetes Care 2005; 28:1083–1091.
  33. Zinman B, Hoogwerf BJ, Durán García S, et al. The effect of adding exenatide to a thiazolidinedione in suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med 2007; 146:477–485.
  34. Barnett AH, Burger J, Johns D, et al. Tolerability and efficacy of exenatide and titrated insulin glargine in adult patients with type 2 diabetes previously uncontrolled with metformin or a sulfonylurea: a multinational, randomized, open-label, two-period, crossover noninferiority trial. Clin Ther 2007; 29:2333–2348.
  35. Heine RJ, Van Gaal LF, Johns D, Mihm MJ, Widel MH, Brodows RG; for the GWAA Study Group. Exenatide versus insulin glargine in patients with suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med 2005; 143:559–569.
  36. Nauck MA, Duran S, Kim D, et al. A comparison of twice-daily exenatide and biphasic insulin aspart in patients with type 2 diabetes who were suboptimally controlled with sulfonylurea and metformin: a non-inferiority study. Diabetologia 2007; 50:259–267.
  37. Klonoff DC, Buse JB, Nielsen LL, et al. Exenatide effects on diabetes, obesity, cardiovascular risk factors and hepatic biomarkers in patients with type 2 diabetes treated for at least 3 years. Curr Med Res Opin 2008; 24:275–286.
  38. Moretto TJ, Milton DR, Ridge TD, et al. Efficacy and tolerability of exenatide monotherapy over 24 weeks in antidiabetic drug-naïve patients with type 2 diabetes: a randomized, double-blind, placebo-controlled, parallel-group study. Clin Ther 2008; 30:1448–1460.
  39. DeFronzo RA, Okerson T, Viswanathan P, Guan X, Holcombe JH, MacConell L. Effects of exenatide versus sitagliptin on postprandial glucose, insulin and glucagon secretion, gastric emptying, and caloric intake: a randomized, cross-over study. Curr Med Res Opin 2008; 24:2943–2952.
  40. Seino Y, Rasmussen MF, Zdravkovic M, Kaku K. Dose-dependent improvement in glycemia with once-daily liraglutide without hypoglycemia or weight gain: a double-blind, randomized, controlled trial in Japanese patients with type 2 diabetes. Diabetes Res Clin Pract 2008; 81:161–168.
  41. Vilsbøll T, Zdravkovic M, Le-Thi T, et al. Liraglutide, a long-acting human glucagon-like peptide-1 analog, given as monotherapy significantly improves glycemic control and lowers body weight without risk of hypoglycemia in patients with type 2 diabetes. Diabetes Care 2007; 30:1608–1610.
  42. Aschner P, Kipnes MS, Lunceford JK, Sanchez M, Mickel C, Williams-Herman DE; for the Sitagliptin Study 021 Group. Effect of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy on glycemic control in patients with type 2 diabetes. Diabetes Care 2006; 29:2632–2637.
  43. Nonaka K, Kakikawa T, Sato A, et al. Efficacy and safety of sitagliptin monotherapy in Japanese patients with type 2 diabetes. Diabetes Res Clin Pract 2008; 79:291–298.
  44. Raz I, Hanefeld M, Xu L, Caria C, Williams-Herman D, Khatami H; for the Sitagliptin Study 023 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy in patients with type 2 diabetes mellitus. Diabetologia 2006; 49:2564–2571.
  45. Scott R, Wu M, Sanchez M, Stein P. Efficacy and tolerability of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy over 12 weeks in patients with type 2 diabetes. Int J Clin Pract 2007; 61:171–180.
  46. Charbonnel B, Karasik A, Liu J, Wu M, Meininger G; for the Sitagliptin Study 020 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin added to ongoing metformin therapy in patients with type 2 diabetes inadequately controlled with metformin alone. Diabetes Care 2006; 29:2638–2643.
  47. Hermansen K, Kipnes M, Luo E, Fanurik D, Khatami H, Stein P; for the Sitagliptin Study 035 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor, sitagliptin, in patients with type 2 diabetes mellitus inadequately controlled on glimepiride alone or on glimepiride and metformin. Diabetes Obes Metab 2007; 9:733–745.
  48. Nauck MA, Meininger G, Sheng D, Terranella L, Stein PP; for the Sitagliptin Study 024 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor, sitagliptin, compared with the sulfonylurea, glipizide, in patients with type 2 diabetes inadequately controlled on metformin alone: a randomized, double-blind, non-inferiority trial. Diabetes Obes Metab 2007; 9:194–205.
  49. Raz I, Chen Y, Wu M, et al. Efficacy and safety of sitagliptin added to ongoing metformin therapy in patients with type 2 diabetes. Curr Med Res Opin 2008; 24:537–550.
  50. Rosenstock J, Brazg R, Andryuk PJ, Lu K, Stein P; for the Sitagliptin Study 019 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin added to ongoing pioglitazone therapy in patients with type 2 diabetes: a 24-week, multicenter, randomized, double-blind, placebo-controlled, parallel-group study. Clin Ther 2006; 28:1556–1568.
  51. Chacra AR, Tan GH, Apanovitch A, Ravichandran S, List J, Chen R; for the CV181-040 Investigators. Saxagliptin added to a submaximal dose of sulphonylurea improves glycaemic control compared with uptitration of sulphonylurea in patients with type 2 diabetes: a randomised controlled trial. Int J Clin Pract 2009; 63:1395–1406.
  52. DeFronzo RA, Hissa MN, Garber AJ, et al. The efficacy and safety of saxagliptin when added to metformin therapy in patients with inadequately controlled type 2 diabetes with metformin alone. Diabetes Care 2009; 32:1649–1655.
  53. Jadzinsky M, Pfützner A, Paz-Pacheco E, Xu Z, Allen E, Chen R; for the CV181-039 Investigators. Saxagliptin given in combination with metformin as initial therapy improves glycaemic control in patients with type 2 diabetes compared with either monotherapy: a randomized controlled trial. Diabetes Obes Metab 2009; 11:611–622.
  54. Rosenstock J, Aguilar-Salinas C, Klein E, Nepal S, List J, Chen R; for the CV181-011 Study Investigators. Effect of saxagliptin monotherapy in treatment-naïve patients with type 2 diabetes. Curr Med Res Opin 2009; 25:2401–2411.
  55. Rosenstock J, Sankoh S, List JF. Glucose-lowering activity of the dipeptidyl peptidase-4 inhibitor saxagliptin in drug-naïve patients with type 2 diabetes. Diabetes Obes Metab 2008; 10:376–386.
  56. Dejager S, Razac S, Foley JE, Schweizer A. Vildagliptin in drug-naïve patients with type 2 diabetes: a 24-week, double-blind, randomized, placebo-controlled, multiple-dose study. Horm Metab Res 2007; 39:218–223.
  57. Garber AJ, Schweizer A, Baron MA, Rochotte E, Dejager S. Vildagliptin in combination with pioglitazone improves glycaemic control in patients with type 2 diabetes failing thiazolidinedione monotherapy: a randomized, placebo-controlled study. Diabetes Obes Metab 2007; 9:166–174.
  58. Göke B, Hershon K, Kerr D, et al. Efficacy and safety of vildagliptin monotherapy during 2-year treatment of drug-naïve patients with type 2 diabetes: comparison with metformin. Horm Metab Res 2008; 40:892–895.
  59. Pan C, Yang W, Barona JP, et al. Comparison of vildagliptin and acarbose monotherapy in patients with type 2 diabetes: a 24-week, double-blind, randomized trial. Diabet Med 2008; 25:435–441.
  60. Pi-Sunyer FX, Schweizer A, Mills D, Dejager S. Efficacy and tolerability of vildagliptin monotherapy in drug-naïve patients with type 2 diabetes. Diabetes Res Clin Pract 2007; 76:132–138.
  61. Schweizer A, Couturier A, Foley JE, Dejager S. Comparison between vildagliptin and metformin to sustain reductions in HbA(1c) over 1 year in drug-naïve patients with type 2 diabetes. Diabetes Med 2007; 24:955–961.
  62. Bolen S, Feldman L, Vassy J, et al. Systematic review: comparative effectiveness and safety of oral medications for type 2 diabetes mellitus. Ann Intern Med 2007; 147:386–399.
  63. Linnebjerg H, Park S, Kothare PA, et al. Effect of exenatide on gastric emptying and relationship to postprandial glycemia in type 2 diabetes. Regul Pept 2008; 151:123–129.
  64. Bunck MC, Diamant M, Cornér A, et al. One-year treatment with exenatide improves beta-cell function, compared with insulin glargine, in metformin-treated type 2 diabetic patients: a randomized, controlled trial. Diabetes Care 2009; 32:762–768.
  65. Merani S, Truong W, Emamaullee JA, Toso C, Knudsen LB, Shapiro AM. Liraglutide, a long-acting human glucagon-like peptide 1 analog, improves glucose homeostasis in marginal mass islet transplantation in mice. Endocrinology 2008; 149:4322–4328.
  66. Mari A, Degn K, Brock B, Rungby J, Ferrannini E, Schmitz O. Effects of the long-acting human glucagon-like peptide-1 analog liraglutide on beta-cell function in normal living conditions. Diabetes Care 2007; 30:2032–2033.
  67. Mari A, Nielsen LL, Nanayakkara N, DeFronzo RA, Ferrannini E, Halseth A. Mathematical modeling shows exenatide improved beta-cell function in patients with type 2 diabetes treated with metformin or metformin and a sulfonylurea. Horm Metab Res 2006; 38:838–844.
  68. Xu L, Man CD, Charbonnel B, et al. Effect of sitagliptin, a dipeptidyl peptidase-4 inhibitor, on beta-cell function in patients with type 2 diabetes: a model-based approach. Diabetes Obes Metab 2008; 10:1212–1220.
  69. Mari A, Scherbaum WA, Nilsson PM, et al. Characterization of the influence of vildagliptin on model-assessed b-cell function in patients with type 2 diabetes and mild hyperglycemia. J Clin Endocrinol Metab 2008; 93:103–109.
  70. Nathan DM, Buse JB, Davidson MB, et al. Medical management of hyperglycemia in type 2 diabetes: a consensus algorithm for the initiation and adjustment of therapy: a consensus statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care 2009; 32:193–203.
  71. Jellinger PS, Davidson JA, Blonde L, et al; for the ACE/AACE Diabetes Road Map Task Force. Road maps to achieve glycemic control in type 2 diabetes mellitus: ACE/AACE Diabetes Road Map Task Force. Endocr Pract 2007; 13:260–268.
Article PDF
Author and Disclosure Information

Jaime A. Davidson, MD
Clinical Professor of Internal Medicine, Division of Endocrinology, University of Texas Southwestern Medical School, Dallas, TX

Correspondence: Jaime A. Davidson, MD, 7777 Forest Lane, Suite C204, Dallas, TX 75230; [email protected]

Dr. Davidson reported that he has received grant support from Bristol-Myers Squibb, Eli Lilly and Company, GlaxoSmithKline, MannKind Corporation, Novo Nordisk, Pfizer Inc., and Sanofi-Aventis; consulting/advisory fees from AstraZeneca, Boehringer Ingelheim GmbH, Bristol-Myers Squibb, Eli Lilly and Company, F. Hoffmann-La Roche Ltd., Generex Biotechnology Corporation, Johnson & Johnson, Novo Nordisk, and Takeda Pharmaceutical Company Limited; and speakers’ bureau fees from Eli Lilly and Company, Novo Nordisk, and Takeda Pharmaceutical Company Limited. He reported that he has stock ownership interest in Eli Lilly and Company, Generex Biotechnology Corporation, GlaxoSmithKline, and Pfizer, Inc., managed by Royal Alliance Associates, Inc. Dr. Davidson reported that he received no honorarium for writing this article.

Dr. Davidson reported that he wrote this article and received no assistance with content development from unnamed contributors. He reported that BlueSpark Healthcare Communications, a medical communications company, assisted with preliminary literature searches, reference verification, proofing for grammar and style, table and figure rendering based on author instructions, copyright permission requests, and identification of topical overlap with other manuscripts in this supplement.

Publications
Page Number
S28-S38
Author and Disclosure Information

Jaime A. Davidson, MD
Clinical Professor of Internal Medicine, Division of Endocrinology, University of Texas Southwestern Medical School, Dallas, TX

Correspondence: Jaime A. Davidson, MD, 7777 Forest Lane, Suite C204, Dallas, TX 75230; [email protected]

Dr. Davidson reported that he has received grant support from Bristol-Myers Squibb, Eli Lilly and Company, GlaxoSmithKline, MannKind Corporation, Novo Nordisk, Pfizer Inc., and Sanofi-Aventis; consulting/advisory fees from AstraZeneca, Boehringer Ingelheim GmbH, Bristol-Myers Squibb, Eli Lilly and Company, F. Hoffmann-La Roche Ltd., Generex Biotechnology Corporation, Johnson & Johnson, Novo Nordisk, and Takeda Pharmaceutical Company Limited; and speakers’ bureau fees from Eli Lilly and Company, Novo Nordisk, and Takeda Pharmaceutical Company Limited. He reported that he has stock ownership interest in Eli Lilly and Company, Generex Biotechnology Corporation, GlaxoSmithKline, and Pfizer, Inc., managed by Royal Alliance Associates, Inc. Dr. Davidson reported that he received no honorarium for writing this article.

Dr. Davidson reported that he wrote this article and received no assistance with content development from unnamed contributors. He reported that BlueSpark Healthcare Communications, a medical communications company, assisted with preliminary literature searches, reference verification, proofing for grammar and style, table and figure rendering based on author instructions, copyright permission requests, and identification of topical overlap with other manuscripts in this supplement.

Author and Disclosure Information

Jaime A. Davidson, MD
Clinical Professor of Internal Medicine, Division of Endocrinology, University of Texas Southwestern Medical School, Dallas, TX

Correspondence: Jaime A. Davidson, MD, 7777 Forest Lane, Suite C204, Dallas, TX 75230; [email protected]

Dr. Davidson reported that he has received grant support from Bristol-Myers Squibb, Eli Lilly and Company, GlaxoSmithKline, MannKind Corporation, Novo Nordisk, Pfizer Inc., and Sanofi-Aventis; consulting/advisory fees from AstraZeneca, Boehringer Ingelheim GmbH, Bristol-Myers Squibb, Eli Lilly and Company, F. Hoffmann-La Roche Ltd., Generex Biotechnology Corporation, Johnson & Johnson, Novo Nordisk, and Takeda Pharmaceutical Company Limited; and speakers’ bureau fees from Eli Lilly and Company, Novo Nordisk, and Takeda Pharmaceutical Company Limited. He reported that he has stock ownership interest in Eli Lilly and Company, Generex Biotechnology Corporation, GlaxoSmithKline, and Pfizer, Inc., managed by Royal Alliance Associates, Inc. Dr. Davidson reported that he received no honorarium for writing this article.

Dr. Davidson reported that he wrote this article and received no assistance with content development from unnamed contributors. He reported that BlueSpark Healthcare Communications, a medical communications company, assisted with preliminary literature searches, reference verification, proofing for grammar and style, table and figure rendering based on author instructions, copyright permission requests, and identification of topical overlap with other manuscripts in this supplement.

Article PDF
Article PDF
Related Articles

The prevalence of type 2 diabetes mellitus (T2DM) is increasing exponentially worldwide. According to the Centers for Disease Control and Prevention, more than 23 million Americans had diabetes in 2007.1 Globally, the prevalence of diabetes, of which T2DM accounts for 90% to 95% of cases,1 is expected to increase from 171 million in 2000 to 366 million in 2030.2 The National Health and Nutrition Examination Survey (NHANES) showed that about 66% of Americans were overweight or obese between 2003–2004.3 Data from a Swedish National Diabetes Register study showed both overweight and obesity as independent risk factors for cardiovascular disease (CVD) in patients with T2DM.4

This article presents an overview of the evolving concepts of the pathophysiology of T2DM, with a focus on two new therapeutic classes: the glucagon-like peptide–1 (GLP-1) receptor agonists and the dipeptidyl peptidase–4 (DPP-4) inhibitors.

THE PATHOPHYSIOLOGY OF T2DM

The American Association of Clinical Endocrinologists (AACE) describes T2DM as “a progressive, complex metabolic disorder characterized by coexisting defects of multiple organ sites including insulin resistance in muscle and adipose tissue, a progressive decline in pancreatic insulin secretion, unrestrained hepatic glucose production, and other hormonal deficiencies.”5 Other defects include accelerated gastric emptying in patients with T2DM, especially those who are obese or who have the disease for a long duration.6,7

Hormonal deficiencies in T2DM are related to abnormalities in the secretion of the beta-cell hormone amylin, the alpha-cell hormone glucagon, and the incretin hormones GLP-1 and glucose-dependent insulinotropic polypeptide (GIP).8,9 In addition to the triumvirate of core defects associated with T2DM (involvement of the pancreatic beta cell, muscle, and liver), other mechanisms of disease onset have been advanced, including accelerated lipolysis, hyperglucagonemia, and incretin deficiency/resistance.9 Also, the rate of basal hepatic glucose production is markedly increased in patients with T2DM, which is closely correlated with elevations in fasting plasma glucagon concentration.9

The incretin effect—the intestinal augmentation of secretion of insulin—attributed to GLP-1 and GIP is reduced in patients with T2DM.10 The secretion of GIP may be normal or elevated in patients with T2DM while the secretion of GLP-1 is deficient; however, cellular responsiveness to GLP-1 is preserved while responsiveness to GIP is diminished.11

Both endogenous and exogenous GLP-1 and GIP are degraded in vivo and in vitro by the enzyme DPP-4,12
a ubiquitous, membrane-spanning, cell-surface aminopeptidase that preferentially cleaves peptides with a proline or alanine residue in the second amino-terminal position. DPP-4 is widely expressed (eg, in the liver, lungs, kidney, lymphocytes, epithelial cells, endothelial cells). The role of DPP-4 in the immune system stems from its exopeptidase activity and its interactions with various molecules, including cyto­kines and chemokines.13

INCRETIN-BASED THERAPIES: GLP-1 RECEPTOR AGONISTS AND DPP-4 INHIBITORS

Exenatide is a GLP-1 receptor agonist that is resistant to DPP-4 degradation. Based on preclinical studies, exenatide, which shares a 53% amino acid sequence identity with human GLP-1, is approximately 5,500 times more potent than endogenous GLP-1 in glucose lowering.14,15 Among the acute actions of exenatide is glucose-dependent insulinotropism, the end result of which may be a reduced risk of hypoglycemia.16 This contrasts with insulin secretagogues (eg, sulfonylureas), which increase insulin secretion regardless of glucose concentrations.

Exenatide received US Food and Drug Administration (FDA) approval in 2005 and is indicated for the treatment of patients with T2DM.13,17 Exenatide is administered BID as a subcutaneous (SC) injection in doses of 5 or 10 μg within 1 hour before the two major meals of the day, which should be eaten about 6 hours apart.18

Approved in 2006, sitagliptin was the first DPP-4 inhibitor indicated for adjunctive therapy to lifestyle modifications for the treatment of patients with T2DM.17 The recommended dosage of oral sitagliptin is 100 mg QD. A single-tablet formulation of the combination of sitagliptin and metformin was approved by the FDA in 2007.19 Another DPP-4 inhibitor, saxagliptin, was approved in July 2009 for treatment of patients with T2DM either as monotherapy or in combination with metformin, sulfonylurea, or a thiazolidinedione (TZD).20 The DPP-4 inhibitor vildagliptin is approved in the European Union and Latin America but not in the United States. Vildagliptin is available as a 50- or 100-mg daily dosage; it has been recommended for use at 50 mg QD in combination with a sulfonylurea or at 50 mg BID with either metformin or a TZD.18

GLP-1 RECEPTOR AGONISTS AND DPP-4 INHIBITORS IN DEVELOPMENT

Exenatide is currently being evaluated as a once-weekly formulation.21,22 Compared with the BID formulation, exenatide once weekly has been shown to produce significantly greater improvements in glycemic control, with similar reductions in body weight and no increased risk of hypoglycemia.21

Also undergoing regulatory review is the partly DPP-4–resistant acylated GLP-1 receptor agonist liraglutide.13 Liraglutide, a human analogue GLP-1 receptor agonist, has 97% linear amino acid sequence homology to human GLP-1.23,24 Based on its prolonged degradation time and resulting 10- to 14-hour half-life, liraglutide is anticipated to be dosed once daily.13,25,26

Other GLP-1 receptor agonists and DPP-4 inhibitors are in varying stages of development.27 Albiglutide is a long-acting GLP-1 receptor agonist that is generated by the genetic fusion of a DPP-4–resistant GLP-1 to human albumin. Based on pharmacokinetic studies, albiglutide has a half-life of 6 to 8 days. AVE0010, an exendin-4-based GLP-1 receptor agonist, was shown in a 28-day T2DM clinical trial to have an affinity four times greater than native GLP-1 for the human GLP-1 receptor.27 Taspoglutide (R1583), a human analogue GLP-1 receptor agonist, was evaluated in three randomized, placebo-controlled studies as a GLP-1 receptor agonist. Alogliptin, a DPP-4 inhibitor currently in development, has been shown to be safe and effective in studies as monotherapy and in combination with other antidiabetes agents.28–30

 

 

CLINICAL TRIALS: GLP-1 RECEPTOR AGONISTS AND DPP-4 INHIBITORS

This section summarizes clinical trials of GLP-1 receptor agonists and DPP-4 inhibitors. The summary is based on literature published from 2005 to 2009 relevant to phase 3 or 4 T2DM clinical trials with currently available agents, or agents with pending new drug applications.

Table 1 summarizes the data on the effects of the GLP-1 receptor agonists on glucose lowering based on glycosylated hemoglobin (HbA1c) mean changes from baseline, body weight, and hypoglycemia. Eleven studies were identified for exenatide, including three pivotal trials,31–33 three insulin-comparator studies,34–36 one long-term study,37 one monotherapy study (a use for which it is not currently indicated),38 one head-to-head study with a DPP-4 inhibitor,39 and two studies with exenatide once weekly (which is currently investigational).21,22 Five primary efficacy studies with liraglutide were also identified.23,25,26,40,41

Table 2 summarizes the corresponding data for the DPP-4 inhibitors. Ten studies with sitagliptin were identified, including four monotherapy studies,42–45 one head-to-head study with a GLP-1 receptor agonist,39 and five studies in which sitagliptin was used in combination or as add-on therapy.46–50 Five saxagliptin studies are reviewed, including two in which saxagliptin was used in combination with metformin and one in combination with glyburide.51–55 Six studies with vildagliptin were reviewed,56–61 but no trials specific to the single-tablet formulation of sitagliptin plus metformin were identified.

Effects on HbA1c and weight

GLP-1 receptor agonists reduced HbA1c. Based on the studies reviewed in Table 1, exenatide BID reduced baseline HbA1c by a maximum of –1.5% at 30 weeks.21,31,32 Exenatide has demonstrated sustained reductions in HbA1c of –0.8% for up to 3.5 years in an open-label extension trial.37 Even greater reductions in HbA1c (–1.4% at 15 weeks and –1.9% at 30 weeks) have been reported with the once-weekly formulation under clinical development.21,22 Liraglutide, another GLP-1 receptor agonist under development, has reported HbA1c reductions from baseline up to –1.67% at 14 weeks,40,41 up to –1.1% at 26 weeks,23,26 and up to –1.14% at 52 weeks.25 The reductions quoted generally refer to means, and individual patients may have greater or lesser responses. Also, baseline HbA1c is a significant determinant of the potential HbA1c reduction. Higher baseline values drop more significantly than do baseline values that are closer to normal.

Weight reduction with GLP-1 receptor agonists. In addition to effective glucose lowering, the GLP-1 receptor agonists, particularly exendin-4 agonists, produced beneficial effects on weight (Table 1). Exenatide BID elicited mean weight reductions up to –3.6 kg at 30 weeks21,31,32 and –5.3 kg at 3.5 years.37 Exenatide once weekly resulted in mean weight reductions of up to –3.8 kg at 15 weeks22 and –3.7 kg at 30 weeks.21 Effects on weight with liraglutide varied from a mean reduction of up to –2.99 kg to a slight gain of up to +0.13 kg at 14 weeks40,41 and with weight loss of up to –2.8 kg at 26 weeks23,26 and up to –2.5 kg at 52 weeks.25 In this review, only exenatide has been assessed in insulin-comparator studies, where it was shown to reduce weight compared with the insulin analogues, which led to weight gain.34–36

Hypoglycemia. Patients receiving exenatide experienced lower rates of hypoglycemia (up to 17%) than patients treated with either insulin glargine or insulin aspart (~25%).34,36 The rate of hypoglycemia with exenatide is comparable to that seen with metformin (up to 21%) in a systematic review of oral antidiabetes agents conducted by the Agency for Healthcare Research and Quality.62 No major hypoglycemic events were reported in the liraglutide studies reviewed. The incidence of hypoglycemia reported with DPP-4 inhibitors (Table 2) is also low (2% or less in most studies). The glucose-dependent mechanisms of the incretin-based therapies minimizes the risk of hypoglycemia.

DPP-4 inhibitors and sustained HbA1c reduction. The effects of the DPP-4 inhibitors on HbA1c and weight, either as monotherapy or in combination with other agents, were evaluated in studies ranging in duration from 12 to 52 weeks (Table 2). No studies were identified that compared the glycemic control effects of DPP-4 inhibitors and insulin analogues. Sitagliptin led to a mean reduction in HbA1c from baseline of up to –0.65% at 12 weeks,43,45 up to –0.48% at 18 weeks,44 up to –0.85% at 24 weeks,42,46,47,50 up to –1.0% at 30 weeks,49 and up to –0.67% at 52 weeks.48 Saxagliptin mean reductions in HbA1c ranged from –0.43% to –1.17%.51–54 Data from four 24-week T2DM studies56–60 showed vildagliptin reducing HbA1c up to –1.4% at 24 weeks, with the greatest reduction in a study that involved drug-naïve patients with a relatively short duration of disease (mean, 1.2 years).59 Reductions in HbA1c of –1.0% were sustained in a 52-week study61 and its 52-week extension.58

DPP-4 inhibitors: weight neutral. The DPP-4 inhibitors appear to have a weight-neutral effect (Table 2). The effects of sitagliptin on weight ranged from a loss of –1.5 kg48 at 52 weeks to a gain of +1.8 kg at 24 weeks.50 Weight changes with saxagliptin ranged from a mean reduction of –1.8 kg53 to a gain of +0.7 kg.51 Two vildagliptin studies showed varying effects on weight ranging from a loss of up to –1.8 kg from baseline56 to a gain of up to +1.3 kg57 relative to placebo, both at 24 weeks.

Potential for CV risk reduction

Potentially beneficial effects on CV risk factors, including blood pressure (ie, reduction) and lipid concentrations (ie, differential effects on low-density lipoprotein and high-density lipoprotein cholesterol), were identified in seven GLP-1 receptor studies—three with exenatide (two with exenatide BID,37,38 and one with the investigational exenatide once weekly21) and four with liraglutide.23,25,26,41 For the DPP-4 inhibitors, three studies were identified—two with sitagliptin45,50 and one with vildagliptin61—in which potentially beneficial effects on CV risk factors were demonstrated.The data have been encouraging, although the clinical implications have yet to be fully understood.

Head-to-head comparison

A recent study compared the effects of the GLP-1 receptor agonist exenatide and the DPP-4 inhibitor sitagliptin on postprandial glucose (PPG) concentrations, insulin and glucagon secretion, gastric intake, and caloric intake.39 Although limited by a short treatment duration (2 weeks), the study showed that the GLP-1 receptor agonist had a greater effect than the DPP-4 inhibitor in reducing PPG concentrations, a more potent effect in increasing insulin secretion and decreasing postprandial glucagon secretion, and a relatively greater effect in reducing caloric intake; and that it decreased the rate of gastric emptying (sitagliptin had no effect). These differences suggest that exenatide may provide a greater degree of GLP-1 receptor activation than the more physiologic concentrations of GLP-1 reached with DPP-4 inhibition.39 Results of a scintigraphic study showed that exenatide substantially slows the gastric emptying that is accelerated in patients with T2DM. This could be another beneficial mechanism in treating postprandial glycemia.63

Adverse effects

Exenatide has shown effects on hepatic injury markers (ie, improvement in alanine and aspartate aminotransferases) for up to 3.5 years of treatment.37 For the GLP-1 receptor agonist and DPP-4 inhibitor studies reviewed, the adverse events were generally mild and included nausea and vomiting, nasopharyngitis, and mild hypoglycemia.

 

 

Meta-analysis conclusions

The published clinical trial data presented in this review expand the body of evidence on the safety and efficacy of incretin-based therapy in patients with T2DM. These data include the results of a meta-analysis by Amori et al,17 which examined randomized controlled trials of 12 weeks’ or longer duration that compared incretin-based therapy with placebo or other diabetes medications and reported HbA1c changes in adults with T2DM. The meta-analysis showed that incretin-based therapies reduced HbA1c more than placebo (weighted mean difference, –0.97% [95% confidence interval (CI), –1.13% to –0.81%] for GLP-1 receptor agonists and –0.74% [95% CI, –0.85% to –0.62%] for DPP-4 inhibitors) and were noninferior to other antidiabetes agents. Treatment with a GLP-1 receptor agonist (ie, exenatide) caused weight loss (–1.4 kg and –4.8 kg vs placebo and insulin, respectively) while DPP-4 inhibitors (ie, sitagliptin, vildagliptin) were weight neutral.17

Beta-cell function

Evidence regarding the effects of incretin-based therapies, particularly the exendin-4 GLP-1 receptor agonists, on beta-cell function in patients with T2DM continues to accumulate. When assessing long-term (1 year) exenatide treatment in patients with T2DM, a trial (n = 69) comparing exenatide with the basal insulin analogue insulin glargine showed that exenatide and insulin glargine resulted in similar reductions in HbA1c (–0.8% vs –0.7%; P = .55).64 However, exenatide significantly reduced body weight while insulin glargine resulted in weight gain (–3.6 kg vs +1.0 kg; P < .0001). In terms of beta-cell function, arginine-stimulated C-peptide secretion during hyperglycemia increased 2.46-fold from baseline after 52 weeks of exenatide treatment compared with 1.31-fold with insulin glargine treatment (P < .0001).64

With respect to the direct beta-cell effects of liraglutide, a preclinical study reported that liraglutide improved glucose homeostasis in marginal mass islet transplantation in diabetic mice.65 In this study, liraglutide was shown, in a mouse model, to reduce the time to normoglycemia after islet cell transplantation (median time, 1 vs 72.5 days; P < .0001). The effects of liraglutide on beta-cell function also were assessed in 13 patients with T2DM. After 7 days of treatment, liraglutide improved beta-cell function, which was associated with improvement in glucose concentration.66 Liraglutide improved potentiation of insulin secretion during the first meal, owing in part to restoration of the potentiation peak (which is markedly blunted in T2DM), in a phenomenon similar to that observed with exenatide.67

Beneficial effects on beta-cell function have also been reported with DPP-4 inhibitors. In a model-based analysis of patients with T2DM, it was shown that sitagliptin improved basal, static, and dynamic responsiveness of pancreatic beta cells to glucose. The results were observed when sitagliptin was administered both as an add-on to metformin therapy and as monotherapy.68 A 52-week, double-blind, randomized, parallel-group study compared vildagliptin 50 mg/day and placebo in 306 patients with T2DM and mild hyperglycemia (HbA1c, 6.2% to 7.5%). Vildagliptin was shown to significantly increase fasting insulin secretory tone, glucose sensitivity, and rate sensitivity, all of which are aspects of beta-cell function.69

Summary

Based on the ability of incretin-based therapies to address various disease mechanisms, including beta-cell defects (ie, hyperglycemia), hormone-related abnormalities (ie, hyperglucagonemia, incretin deficiency/resistance), and accelerated gastric emptying (especially with GLP-1 receptor agonists); their favorable effects on weight (reduction with GLP-1 receptor agonists and neutral with DPP-4 inhibitors); their beneficial effects on CV risk factors; and their good safety profile (ie, hypoglycemia risk comparable with metformin), these agents could be considered therapeutic advances for the treatment of patients with T2DM.

INCRETIN-BASED THERAPIES IN GUIDELINES AND ALGORITHMS

The 2007 AACE medical guidelines for clinical practice for the management of diabetes recognized the place of the incretin-based therapies and included them among the pharmacologic options.5 Exenatide was specifically recommended for combination therapy with metformin, a sulfonylurea (secretagogue), a sulfonylurea plus metformin, or a TZD. Sitagliptin was recommended for use as monotherapy or in combination with metformin or a TZD.5

In 2009, the American Diabetes Association (ADA) and the European Association for the Study of Diabetes convened a consensus panel to produce an algorithm for the initiation and adjustment of therapy for patients with T2DM. In this algorithm, GLP-1 receptor agonists were considered appropriate in certain clinical scenarios (eg, when hypoglycemia was an issue or weight loss was a major consideration during treatment). However, the groups also noted a need for more data on long-term safety and the cost of treatment with incretin-based therapies.70

The AACE and the American College of Endocrinology recently developed “road maps” for managing patients with T2DM. In patients with T2DM who are naïve to therapy, DPP-4 inhibitors are among the recommended first options when the initial HbA1c is 6.0% to 7.0% and as a combination therapy component when HbA1c reaches 7.0% to 9.0%. In patients who have already received monotherapy for 2 to 3 months and whose HbA1c is 6.5% to 8.5%, treatment options include combination therapy with a DPP-4 inhibitor and metformin or a TZD. Another option includes the initiation of treatment with a GLP-1 receptor agonist in combination with a TZD, with metformin or a sulfonylurea, or with metformin and a sulfonylurea.71

The role of GLP-1 receptor agonist therapies and their incorporation into T2DM treatment algorithms was noted at the 2008 annual meeting of the ADA. In the Banting lecture, Ralph A. DeFronzo, MD, advocated the early use of triple-drug therapy with metformin, exenatide, and a TZD in the management of patients with T2DM.9

CONCLUSION

T2DM, which is linked to weight gain and obesity, is a complex disease that predisposes patients to and is associated with CVD. A better understanding and appreciation of the role of the incretin system in the pathogenesis of T2DM has led to the development of incretin-based therapies, such as the GLP-1 receptor agonists and DPP-4 inhibitors. As more experimental and clinical evidence becomes available, subtle nuances are emerging that distinguish the roles of these two therapeutic classes.

The prevalence of type 2 diabetes mellitus (T2DM) is increasing exponentially worldwide. According to the Centers for Disease Control and Prevention, more than 23 million Americans had diabetes in 2007.1 Globally, the prevalence of diabetes, of which T2DM accounts for 90% to 95% of cases,1 is expected to increase from 171 million in 2000 to 366 million in 2030.2 The National Health and Nutrition Examination Survey (NHANES) showed that about 66% of Americans were overweight or obese between 2003–2004.3 Data from a Swedish National Diabetes Register study showed both overweight and obesity as independent risk factors for cardiovascular disease (CVD) in patients with T2DM.4

This article presents an overview of the evolving concepts of the pathophysiology of T2DM, with a focus on two new therapeutic classes: the glucagon-like peptide–1 (GLP-1) receptor agonists and the dipeptidyl peptidase–4 (DPP-4) inhibitors.

THE PATHOPHYSIOLOGY OF T2DM

The American Association of Clinical Endocrinologists (AACE) describes T2DM as “a progressive, complex metabolic disorder characterized by coexisting defects of multiple organ sites including insulin resistance in muscle and adipose tissue, a progressive decline in pancreatic insulin secretion, unrestrained hepatic glucose production, and other hormonal deficiencies.”5 Other defects include accelerated gastric emptying in patients with T2DM, especially those who are obese or who have the disease for a long duration.6,7

Hormonal deficiencies in T2DM are related to abnormalities in the secretion of the beta-cell hormone amylin, the alpha-cell hormone glucagon, and the incretin hormones GLP-1 and glucose-dependent insulinotropic polypeptide (GIP).8,9 In addition to the triumvirate of core defects associated with T2DM (involvement of the pancreatic beta cell, muscle, and liver), other mechanisms of disease onset have been advanced, including accelerated lipolysis, hyperglucagonemia, and incretin deficiency/resistance.9 Also, the rate of basal hepatic glucose production is markedly increased in patients with T2DM, which is closely correlated with elevations in fasting plasma glucagon concentration.9

The incretin effect—the intestinal augmentation of secretion of insulin—attributed to GLP-1 and GIP is reduced in patients with T2DM.10 The secretion of GIP may be normal or elevated in patients with T2DM while the secretion of GLP-1 is deficient; however, cellular responsiveness to GLP-1 is preserved while responsiveness to GIP is diminished.11

Both endogenous and exogenous GLP-1 and GIP are degraded in vivo and in vitro by the enzyme DPP-4,12
a ubiquitous, membrane-spanning, cell-surface aminopeptidase that preferentially cleaves peptides with a proline or alanine residue in the second amino-terminal position. DPP-4 is widely expressed (eg, in the liver, lungs, kidney, lymphocytes, epithelial cells, endothelial cells). The role of DPP-4 in the immune system stems from its exopeptidase activity and its interactions with various molecules, including cyto­kines and chemokines.13

INCRETIN-BASED THERAPIES: GLP-1 RECEPTOR AGONISTS AND DPP-4 INHIBITORS

Exenatide is a GLP-1 receptor agonist that is resistant to DPP-4 degradation. Based on preclinical studies, exenatide, which shares a 53% amino acid sequence identity with human GLP-1, is approximately 5,500 times more potent than endogenous GLP-1 in glucose lowering.14,15 Among the acute actions of exenatide is glucose-dependent insulinotropism, the end result of which may be a reduced risk of hypoglycemia.16 This contrasts with insulin secretagogues (eg, sulfonylureas), which increase insulin secretion regardless of glucose concentrations.

Exenatide received US Food and Drug Administration (FDA) approval in 2005 and is indicated for the treatment of patients with T2DM.13,17 Exenatide is administered BID as a subcutaneous (SC) injection in doses of 5 or 10 μg within 1 hour before the two major meals of the day, which should be eaten about 6 hours apart.18

Approved in 2006, sitagliptin was the first DPP-4 inhibitor indicated for adjunctive therapy to lifestyle modifications for the treatment of patients with T2DM.17 The recommended dosage of oral sitagliptin is 100 mg QD. A single-tablet formulation of the combination of sitagliptin and metformin was approved by the FDA in 2007.19 Another DPP-4 inhibitor, saxagliptin, was approved in July 2009 for treatment of patients with T2DM either as monotherapy or in combination with metformin, sulfonylurea, or a thiazolidinedione (TZD).20 The DPP-4 inhibitor vildagliptin is approved in the European Union and Latin America but not in the United States. Vildagliptin is available as a 50- or 100-mg daily dosage; it has been recommended for use at 50 mg QD in combination with a sulfonylurea or at 50 mg BID with either metformin or a TZD.18

GLP-1 RECEPTOR AGONISTS AND DPP-4 INHIBITORS IN DEVELOPMENT

Exenatide is currently being evaluated as a once-weekly formulation.21,22 Compared with the BID formulation, exenatide once weekly has been shown to produce significantly greater improvements in glycemic control, with similar reductions in body weight and no increased risk of hypoglycemia.21

Also undergoing regulatory review is the partly DPP-4–resistant acylated GLP-1 receptor agonist liraglutide.13 Liraglutide, a human analogue GLP-1 receptor agonist, has 97% linear amino acid sequence homology to human GLP-1.23,24 Based on its prolonged degradation time and resulting 10- to 14-hour half-life, liraglutide is anticipated to be dosed once daily.13,25,26

Other GLP-1 receptor agonists and DPP-4 inhibitors are in varying stages of development.27 Albiglutide is a long-acting GLP-1 receptor agonist that is generated by the genetic fusion of a DPP-4–resistant GLP-1 to human albumin. Based on pharmacokinetic studies, albiglutide has a half-life of 6 to 8 days. AVE0010, an exendin-4-based GLP-1 receptor agonist, was shown in a 28-day T2DM clinical trial to have an affinity four times greater than native GLP-1 for the human GLP-1 receptor.27 Taspoglutide (R1583), a human analogue GLP-1 receptor agonist, was evaluated in three randomized, placebo-controlled studies as a GLP-1 receptor agonist. Alogliptin, a DPP-4 inhibitor currently in development, has been shown to be safe and effective in studies as monotherapy and in combination with other antidiabetes agents.28–30

 

 

CLINICAL TRIALS: GLP-1 RECEPTOR AGONISTS AND DPP-4 INHIBITORS

This section summarizes clinical trials of GLP-1 receptor agonists and DPP-4 inhibitors. The summary is based on literature published from 2005 to 2009 relevant to phase 3 or 4 T2DM clinical trials with currently available agents, or agents with pending new drug applications.

Table 1 summarizes the data on the effects of the GLP-1 receptor agonists on glucose lowering based on glycosylated hemoglobin (HbA1c) mean changes from baseline, body weight, and hypoglycemia. Eleven studies were identified for exenatide, including three pivotal trials,31–33 three insulin-comparator studies,34–36 one long-term study,37 one monotherapy study (a use for which it is not currently indicated),38 one head-to-head study with a DPP-4 inhibitor,39 and two studies with exenatide once weekly (which is currently investigational).21,22 Five primary efficacy studies with liraglutide were also identified.23,25,26,40,41

Table 2 summarizes the corresponding data for the DPP-4 inhibitors. Ten studies with sitagliptin were identified, including four monotherapy studies,42–45 one head-to-head study with a GLP-1 receptor agonist,39 and five studies in which sitagliptin was used in combination or as add-on therapy.46–50 Five saxagliptin studies are reviewed, including two in which saxagliptin was used in combination with metformin and one in combination with glyburide.51–55 Six studies with vildagliptin were reviewed,56–61 but no trials specific to the single-tablet formulation of sitagliptin plus metformin were identified.

Effects on HbA1c and weight

GLP-1 receptor agonists reduced HbA1c. Based on the studies reviewed in Table 1, exenatide BID reduced baseline HbA1c by a maximum of –1.5% at 30 weeks.21,31,32 Exenatide has demonstrated sustained reductions in HbA1c of –0.8% for up to 3.5 years in an open-label extension trial.37 Even greater reductions in HbA1c (–1.4% at 15 weeks and –1.9% at 30 weeks) have been reported with the once-weekly formulation under clinical development.21,22 Liraglutide, another GLP-1 receptor agonist under development, has reported HbA1c reductions from baseline up to –1.67% at 14 weeks,40,41 up to –1.1% at 26 weeks,23,26 and up to –1.14% at 52 weeks.25 The reductions quoted generally refer to means, and individual patients may have greater or lesser responses. Also, baseline HbA1c is a significant determinant of the potential HbA1c reduction. Higher baseline values drop more significantly than do baseline values that are closer to normal.

Weight reduction with GLP-1 receptor agonists. In addition to effective glucose lowering, the GLP-1 receptor agonists, particularly exendin-4 agonists, produced beneficial effects on weight (Table 1). Exenatide BID elicited mean weight reductions up to –3.6 kg at 30 weeks21,31,32 and –5.3 kg at 3.5 years.37 Exenatide once weekly resulted in mean weight reductions of up to –3.8 kg at 15 weeks22 and –3.7 kg at 30 weeks.21 Effects on weight with liraglutide varied from a mean reduction of up to –2.99 kg to a slight gain of up to +0.13 kg at 14 weeks40,41 and with weight loss of up to –2.8 kg at 26 weeks23,26 and up to –2.5 kg at 52 weeks.25 In this review, only exenatide has been assessed in insulin-comparator studies, where it was shown to reduce weight compared with the insulin analogues, which led to weight gain.34–36

Hypoglycemia. Patients receiving exenatide experienced lower rates of hypoglycemia (up to 17%) than patients treated with either insulin glargine or insulin aspart (~25%).34,36 The rate of hypoglycemia with exenatide is comparable to that seen with metformin (up to 21%) in a systematic review of oral antidiabetes agents conducted by the Agency for Healthcare Research and Quality.62 No major hypoglycemic events were reported in the liraglutide studies reviewed. The incidence of hypoglycemia reported with DPP-4 inhibitors (Table 2) is also low (2% or less in most studies). The glucose-dependent mechanisms of the incretin-based therapies minimizes the risk of hypoglycemia.

DPP-4 inhibitors and sustained HbA1c reduction. The effects of the DPP-4 inhibitors on HbA1c and weight, either as monotherapy or in combination with other agents, were evaluated in studies ranging in duration from 12 to 52 weeks (Table 2). No studies were identified that compared the glycemic control effects of DPP-4 inhibitors and insulin analogues. Sitagliptin led to a mean reduction in HbA1c from baseline of up to –0.65% at 12 weeks,43,45 up to –0.48% at 18 weeks,44 up to –0.85% at 24 weeks,42,46,47,50 up to –1.0% at 30 weeks,49 and up to –0.67% at 52 weeks.48 Saxagliptin mean reductions in HbA1c ranged from –0.43% to –1.17%.51–54 Data from four 24-week T2DM studies56–60 showed vildagliptin reducing HbA1c up to –1.4% at 24 weeks, with the greatest reduction in a study that involved drug-naïve patients with a relatively short duration of disease (mean, 1.2 years).59 Reductions in HbA1c of –1.0% were sustained in a 52-week study61 and its 52-week extension.58

DPP-4 inhibitors: weight neutral. The DPP-4 inhibitors appear to have a weight-neutral effect (Table 2). The effects of sitagliptin on weight ranged from a loss of –1.5 kg48 at 52 weeks to a gain of +1.8 kg at 24 weeks.50 Weight changes with saxagliptin ranged from a mean reduction of –1.8 kg53 to a gain of +0.7 kg.51 Two vildagliptin studies showed varying effects on weight ranging from a loss of up to –1.8 kg from baseline56 to a gain of up to +1.3 kg57 relative to placebo, both at 24 weeks.

Potential for CV risk reduction

Potentially beneficial effects on CV risk factors, including blood pressure (ie, reduction) and lipid concentrations (ie, differential effects on low-density lipoprotein and high-density lipoprotein cholesterol), were identified in seven GLP-1 receptor studies—three with exenatide (two with exenatide BID,37,38 and one with the investigational exenatide once weekly21) and four with liraglutide.23,25,26,41 For the DPP-4 inhibitors, three studies were identified—two with sitagliptin45,50 and one with vildagliptin61—in which potentially beneficial effects on CV risk factors were demonstrated.The data have been encouraging, although the clinical implications have yet to be fully understood.

Head-to-head comparison

A recent study compared the effects of the GLP-1 receptor agonist exenatide and the DPP-4 inhibitor sitagliptin on postprandial glucose (PPG) concentrations, insulin and glucagon secretion, gastric intake, and caloric intake.39 Although limited by a short treatment duration (2 weeks), the study showed that the GLP-1 receptor agonist had a greater effect than the DPP-4 inhibitor in reducing PPG concentrations, a more potent effect in increasing insulin secretion and decreasing postprandial glucagon secretion, and a relatively greater effect in reducing caloric intake; and that it decreased the rate of gastric emptying (sitagliptin had no effect). These differences suggest that exenatide may provide a greater degree of GLP-1 receptor activation than the more physiologic concentrations of GLP-1 reached with DPP-4 inhibition.39 Results of a scintigraphic study showed that exenatide substantially slows the gastric emptying that is accelerated in patients with T2DM. This could be another beneficial mechanism in treating postprandial glycemia.63

Adverse effects

Exenatide has shown effects on hepatic injury markers (ie, improvement in alanine and aspartate aminotransferases) for up to 3.5 years of treatment.37 For the GLP-1 receptor agonist and DPP-4 inhibitor studies reviewed, the adverse events were generally mild and included nausea and vomiting, nasopharyngitis, and mild hypoglycemia.

 

 

Meta-analysis conclusions

The published clinical trial data presented in this review expand the body of evidence on the safety and efficacy of incretin-based therapy in patients with T2DM. These data include the results of a meta-analysis by Amori et al,17 which examined randomized controlled trials of 12 weeks’ or longer duration that compared incretin-based therapy with placebo or other diabetes medications and reported HbA1c changes in adults with T2DM. The meta-analysis showed that incretin-based therapies reduced HbA1c more than placebo (weighted mean difference, –0.97% [95% confidence interval (CI), –1.13% to –0.81%] for GLP-1 receptor agonists and –0.74% [95% CI, –0.85% to –0.62%] for DPP-4 inhibitors) and were noninferior to other antidiabetes agents. Treatment with a GLP-1 receptor agonist (ie, exenatide) caused weight loss (–1.4 kg and –4.8 kg vs placebo and insulin, respectively) while DPP-4 inhibitors (ie, sitagliptin, vildagliptin) were weight neutral.17

Beta-cell function

Evidence regarding the effects of incretin-based therapies, particularly the exendin-4 GLP-1 receptor agonists, on beta-cell function in patients with T2DM continues to accumulate. When assessing long-term (1 year) exenatide treatment in patients with T2DM, a trial (n = 69) comparing exenatide with the basal insulin analogue insulin glargine showed that exenatide and insulin glargine resulted in similar reductions in HbA1c (–0.8% vs –0.7%; P = .55).64 However, exenatide significantly reduced body weight while insulin glargine resulted in weight gain (–3.6 kg vs +1.0 kg; P < .0001). In terms of beta-cell function, arginine-stimulated C-peptide secretion during hyperglycemia increased 2.46-fold from baseline after 52 weeks of exenatide treatment compared with 1.31-fold with insulin glargine treatment (P < .0001).64

With respect to the direct beta-cell effects of liraglutide, a preclinical study reported that liraglutide improved glucose homeostasis in marginal mass islet transplantation in diabetic mice.65 In this study, liraglutide was shown, in a mouse model, to reduce the time to normoglycemia after islet cell transplantation (median time, 1 vs 72.5 days; P < .0001). The effects of liraglutide on beta-cell function also were assessed in 13 patients with T2DM. After 7 days of treatment, liraglutide improved beta-cell function, which was associated with improvement in glucose concentration.66 Liraglutide improved potentiation of insulin secretion during the first meal, owing in part to restoration of the potentiation peak (which is markedly blunted in T2DM), in a phenomenon similar to that observed with exenatide.67

Beneficial effects on beta-cell function have also been reported with DPP-4 inhibitors. In a model-based analysis of patients with T2DM, it was shown that sitagliptin improved basal, static, and dynamic responsiveness of pancreatic beta cells to glucose. The results were observed when sitagliptin was administered both as an add-on to metformin therapy and as monotherapy.68 A 52-week, double-blind, randomized, parallel-group study compared vildagliptin 50 mg/day and placebo in 306 patients with T2DM and mild hyperglycemia (HbA1c, 6.2% to 7.5%). Vildagliptin was shown to significantly increase fasting insulin secretory tone, glucose sensitivity, and rate sensitivity, all of which are aspects of beta-cell function.69

Summary

Based on the ability of incretin-based therapies to address various disease mechanisms, including beta-cell defects (ie, hyperglycemia), hormone-related abnormalities (ie, hyperglucagonemia, incretin deficiency/resistance), and accelerated gastric emptying (especially with GLP-1 receptor agonists); their favorable effects on weight (reduction with GLP-1 receptor agonists and neutral with DPP-4 inhibitors); their beneficial effects on CV risk factors; and their good safety profile (ie, hypoglycemia risk comparable with metformin), these agents could be considered therapeutic advances for the treatment of patients with T2DM.

INCRETIN-BASED THERAPIES IN GUIDELINES AND ALGORITHMS

The 2007 AACE medical guidelines for clinical practice for the management of diabetes recognized the place of the incretin-based therapies and included them among the pharmacologic options.5 Exenatide was specifically recommended for combination therapy with metformin, a sulfonylurea (secretagogue), a sulfonylurea plus metformin, or a TZD. Sitagliptin was recommended for use as monotherapy or in combination with metformin or a TZD.5

In 2009, the American Diabetes Association (ADA) and the European Association for the Study of Diabetes convened a consensus panel to produce an algorithm for the initiation and adjustment of therapy for patients with T2DM. In this algorithm, GLP-1 receptor agonists were considered appropriate in certain clinical scenarios (eg, when hypoglycemia was an issue or weight loss was a major consideration during treatment). However, the groups also noted a need for more data on long-term safety and the cost of treatment with incretin-based therapies.70

The AACE and the American College of Endocrinology recently developed “road maps” for managing patients with T2DM. In patients with T2DM who are naïve to therapy, DPP-4 inhibitors are among the recommended first options when the initial HbA1c is 6.0% to 7.0% and as a combination therapy component when HbA1c reaches 7.0% to 9.0%. In patients who have already received monotherapy for 2 to 3 months and whose HbA1c is 6.5% to 8.5%, treatment options include combination therapy with a DPP-4 inhibitor and metformin or a TZD. Another option includes the initiation of treatment with a GLP-1 receptor agonist in combination with a TZD, with metformin or a sulfonylurea, or with metformin and a sulfonylurea.71

The role of GLP-1 receptor agonist therapies and their incorporation into T2DM treatment algorithms was noted at the 2008 annual meeting of the ADA. In the Banting lecture, Ralph A. DeFronzo, MD, advocated the early use of triple-drug therapy with metformin, exenatide, and a TZD in the management of patients with T2DM.9

CONCLUSION

T2DM, which is linked to weight gain and obesity, is a complex disease that predisposes patients to and is associated with CVD. A better understanding and appreciation of the role of the incretin system in the pathogenesis of T2DM has led to the development of incretin-based therapies, such as the GLP-1 receptor agonists and DPP-4 inhibitors. As more experimental and clinical evidence becomes available, subtle nuances are emerging that distinguish the roles of these two therapeutic classes.

References
  1. 2007 National Diabetes Fact Sheet. Centers for Disease Control and Prevention Web site. http://www.cdc.gov/diabetes/pubs/estimates07.htm. Updated: July 23, 2008. Accessed September 25, 2009.
  2. Wild S, Roglic G, Green A, Sicree R, King H. Global prevalence of diabetes: estimates for the year 2000 and projections for 2030. Diabetes Care 2004; 27:1047–1053.
  3. Prevalence of overweight and obesity among adults: United States 2003–2004. Centers for Disease Control and Prevention Web site. http://www.cdc.gov/nchs/products/pubs/pubd/hestats/overweight/overwght_adult_03.htm. Published: April 2006. Accessed September 23, 2009.
  4. Eeg-Olofsson K, Cederholm J, Nilsson PM, et al. Risk of cardiovascular disease and mortality in overweight and obese patients with type 2 diabetes: an observational study in 13,087 patients. Diabetologia 2009; 52:65–73.
  5. AACE Diabetes Mellitus Clinical Practice Guidelines Task Force. American Association of Clinical Endocrinologists medical guidelines for clinical practice for the management of diabetes mellitus. Endocr Pract 2007; 13(suppl 1):S4–S68.
  6. Bertin E, Schneider N, Abdelli N, et al. Gastric emptying is accelerated in obese type 2 diabetic patients without autonomic neuropathy. Diabetes Metab 2001; 27:357–364.
  7. Weytjens C, Keymeulen B, Van Haleweyn C, Somers G, Bossuyt A. Rapid gastric emptying of a liquid meal in long-term type 2 diabetes mellitus. Diabet Med 1998; 15:1022–1027.
  8. Stonehouse AH, Holcombe JH, Kendall DM. GLP-1 analogues, DPP-IV inhibitors and the metabolic syndrome. In: Fonseca V, ed. Therapeutic Strategies in Metabolic Syndrome. Oxford, UK: Atlas Medical Publishing Ltd; 2008: 137–157.
  9. DeFronzo RA. From the triumvirate to the ominous octet: a new paradigm for the treatment of type 2 diabetes mellitus. Diabetes 2009; 58:773–795.
  10. Nauck M, Stöckmann F, Ebert R, Creutzfeldt W. Reduced incretin effect in type 2 (non-insulin-dependent) diabetes. Diabetologia 1986; 29:46–52.
  11. Nauck MA, Heimesaat MM, Orskov C, Holst JJ, Ebert R, Creutzfeldt W. Preserved incretin activity of glucagon-like peptide 1 [7-36 amide] but not of synthetic human gastric inhibitory polypeptide in patients with type-2 diabetes mellitus. J Clin Invest 1993; 91:301–307.
  12. Deacon CF, Johnsen AH, Holst JJ. Degradation of glucagon-like peptide-1 by human plasma in vitro yields an N-terminally truncated peptide that is a major endogenous metabolite in vivo. J Clin Endocrinol Metab 1995; 80:952–957.
  13. Drucker DJ, Nauck MA. The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 2006; 368:1696–1705.
  14. Eng J, Kleinman WA, Singh L, Singh G, Raufman J-P. Isolation and characterization of exendin-4, an exendin-3 analogue, from Heloderma suspectum venom: further evidence for an exendin receptor on dispersed acini from guinea pig pancreas. J Biol Chem 1992; 267:7402–7405.
  15. Young AA, Gedulin BR, Bhavsar S, et al. Glucose-lowering and insulin-sensitizing actions of exendin-4: studies in obese diabetic (ob/ob, db/db) mice, diabetic fatty Zucker rats, and diabetic rhesus monkeys (Macaca mulatta). Diabetes 1999; 48:1026–1034.
  16. Kolterman OG, Buse JB, Fineman MS, et al. Synthetic exendin-4 (exenatide) significantly reduces postprandial and fasting plasma glucose in subjects with type 2 diabetes. J Clin Endocrinol Metab 2003; 88:3082–3089.
  17. Amori RE, Lau J, Pittas AG. Efficacy and safety of incretin therapy in type 2 diabetes: systematic review and meta-analysis. JAMA 2007; 298:194–206.
  18. Davidson JA, Parente EB, Gross JL. Incretin mimetics and dipeptidyl peptidase-4 inhibitors: innovative treatment therapies for type 2 diabetes. Arq Bras Endocrinol Metabol 2008; 52:1039–1049.
  19. Alexander GC, Sehgal NL, Moloney RM, Stafford RS. National trends in treatment of type 2 diabetes mellitus, 1994–2007. Arch Intern Med 2008; 168:2088–2094.
  20. US Department of Health and Human Services. FDA approves new drug treatment for type 2 diabetes. US Food and Drug Administration Web site. http://www.fda.gov/newsevents/newsroom/pressannouncements/ucm174780.htm. Published July 31, 2009. Accessed September 18, 2009.
  21. Drucker DJ, Buse JB, Taylor K, et al; for the DURATION-1 Study Group. Exenatide once weekly versus twice daily for the treatment of type 2 diabetes: a randomised, open-label, non-inferiority study. Lancet 2008; 372:1240–1250.
  22. Kim D, MacConell L, Zhuang D, et al. Effects of once-weekly dosing of a long-acting release formulation of exenatide on glucose control and body weight in subjects with type 2 diabetes. Diabetes Care 2007; 30:1487–1493.
  23. Marre M, Shaw J, Brändle M, et al; for the LEAD-1 SU Study Group. Liraglutide, a once-daily human GLP-1 analogue, added to a sulphonylurea over 26 weeks produces greater improvements in glycaemic and weight control compared with adding rosiglitazone or placebo in subjects with type 2 diabetes (LEAD-1 SU). Diabetes Med 2009; 26:268–278.
  24. Knudsen LB, Nielsen PF, Huusfeldt PO, et al. Potent derivatives of glucagon-like peptide 1 with pharmacokinetic properties suitable for once-daily administration. J Med Chem 2000; 43:1664–1669.
  25. Garber A, Henry R, Ratner R, et al; for the LEAD-3 (Mono) Study Group. Liraglutide versus glimepiride monotherapy for type 2 diabetes (LEAD-3 Mono): a randomised, 52-week, phase III, double-blind, parallel-treatment trial. Lancet 2009; 373:473–481.
  26. Nauck M, Frid A, Hermansen K, et al; for the LEAD-2 Study Group. Efficacy and safety comparison of liraglutide, glimepiride, and placebo, all in combination with metformin, in type 2 diabetes: the LEAD (Liraglutide Effect and Action in Diabetes)-2 study. Diabetes Care 2009; 32:84–90.
  27. Baggio LL, Drucker DJ, Maida A, Lamont BJ. ADA 2008: incretin-based therapeutics. MedscapeCME Web site. http://www.medscape.com/viewprogram/15786. Accessed September 18, 2009.
  28. Fleck P, Christopher R, Covington P, Wilson C, Mekki Q. Efficacy and safety of alogliptin monotherapy over 12 weeks in patients with type 2 diabetes. Paper presented at: 68th Annual Meeting of the American Diabetes Association; June 6–10, 2008; San Francisco, CA. Abstract 479-P.
  29. DeFronzo RA, Burant CF, Fleck P, Wilson C, Mekki Q, Pratley RE. Effect of alogliptin combined with pioglitazone on glycemic control in metformin-treated patients with type 2 diabetes. Paper presented at: 69th Annual Meeting of the American Diabetes Association; June 5–9, 2009; New Orleans, LA. Abstract 2024-PO.
  30. Nauck M, Ellis G, Fleck P, Wilson C, Mekki Q. Efficacy and safety of alogliptin added to metformin therapy in patients with type 2 diabetes. Paper presented at: 68th Annual Meeting of the American Diabetes Association; June 6–10, 2008; San Francisco, CA. Abstract 477-P.
  31. DeFronzo RA, Ratner RE, Han J, Kim DD, Fineman MS, Baron AD. Effects of exenatide (exendin-4) on glycemic control and weight over 30 weeks in metformin-treated patients with type 2 diabetes. Diabetes Care 2005; 28:1092–1100.
  32. Kendall DM, Riddle MC, Rosenstock J, et al. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in patients with type 2 diabetes treated with metformin and a sulfonylurea. Diabetes Care 2005; 28:1083–1091.
  33. Zinman B, Hoogwerf BJ, Durán García S, et al. The effect of adding exenatide to a thiazolidinedione in suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med 2007; 146:477–485.
  34. Barnett AH, Burger J, Johns D, et al. Tolerability and efficacy of exenatide and titrated insulin glargine in adult patients with type 2 diabetes previously uncontrolled with metformin or a sulfonylurea: a multinational, randomized, open-label, two-period, crossover noninferiority trial. Clin Ther 2007; 29:2333–2348.
  35. Heine RJ, Van Gaal LF, Johns D, Mihm MJ, Widel MH, Brodows RG; for the GWAA Study Group. Exenatide versus insulin glargine in patients with suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med 2005; 143:559–569.
  36. Nauck MA, Duran S, Kim D, et al. A comparison of twice-daily exenatide and biphasic insulin aspart in patients with type 2 diabetes who were suboptimally controlled with sulfonylurea and metformin: a non-inferiority study. Diabetologia 2007; 50:259–267.
  37. Klonoff DC, Buse JB, Nielsen LL, et al. Exenatide effects on diabetes, obesity, cardiovascular risk factors and hepatic biomarkers in patients with type 2 diabetes treated for at least 3 years. Curr Med Res Opin 2008; 24:275–286.
  38. Moretto TJ, Milton DR, Ridge TD, et al. Efficacy and tolerability of exenatide monotherapy over 24 weeks in antidiabetic drug-naïve patients with type 2 diabetes: a randomized, double-blind, placebo-controlled, parallel-group study. Clin Ther 2008; 30:1448–1460.
  39. DeFronzo RA, Okerson T, Viswanathan P, Guan X, Holcombe JH, MacConell L. Effects of exenatide versus sitagliptin on postprandial glucose, insulin and glucagon secretion, gastric emptying, and caloric intake: a randomized, cross-over study. Curr Med Res Opin 2008; 24:2943–2952.
  40. Seino Y, Rasmussen MF, Zdravkovic M, Kaku K. Dose-dependent improvement in glycemia with once-daily liraglutide without hypoglycemia or weight gain: a double-blind, randomized, controlled trial in Japanese patients with type 2 diabetes. Diabetes Res Clin Pract 2008; 81:161–168.
  41. Vilsbøll T, Zdravkovic M, Le-Thi T, et al. Liraglutide, a long-acting human glucagon-like peptide-1 analog, given as monotherapy significantly improves glycemic control and lowers body weight without risk of hypoglycemia in patients with type 2 diabetes. Diabetes Care 2007; 30:1608–1610.
  42. Aschner P, Kipnes MS, Lunceford JK, Sanchez M, Mickel C, Williams-Herman DE; for the Sitagliptin Study 021 Group. Effect of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy on glycemic control in patients with type 2 diabetes. Diabetes Care 2006; 29:2632–2637.
  43. Nonaka K, Kakikawa T, Sato A, et al. Efficacy and safety of sitagliptin monotherapy in Japanese patients with type 2 diabetes. Diabetes Res Clin Pract 2008; 79:291–298.
  44. Raz I, Hanefeld M, Xu L, Caria C, Williams-Herman D, Khatami H; for the Sitagliptin Study 023 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy in patients with type 2 diabetes mellitus. Diabetologia 2006; 49:2564–2571.
  45. Scott R, Wu M, Sanchez M, Stein P. Efficacy and tolerability of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy over 12 weeks in patients with type 2 diabetes. Int J Clin Pract 2007; 61:171–180.
  46. Charbonnel B, Karasik A, Liu J, Wu M, Meininger G; for the Sitagliptin Study 020 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin added to ongoing metformin therapy in patients with type 2 diabetes inadequately controlled with metformin alone. Diabetes Care 2006; 29:2638–2643.
  47. Hermansen K, Kipnes M, Luo E, Fanurik D, Khatami H, Stein P; for the Sitagliptin Study 035 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor, sitagliptin, in patients with type 2 diabetes mellitus inadequately controlled on glimepiride alone or on glimepiride and metformin. Diabetes Obes Metab 2007; 9:733–745.
  48. Nauck MA, Meininger G, Sheng D, Terranella L, Stein PP; for the Sitagliptin Study 024 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor, sitagliptin, compared with the sulfonylurea, glipizide, in patients with type 2 diabetes inadequately controlled on metformin alone: a randomized, double-blind, non-inferiority trial. Diabetes Obes Metab 2007; 9:194–205.
  49. Raz I, Chen Y, Wu M, et al. Efficacy and safety of sitagliptin added to ongoing metformin therapy in patients with type 2 diabetes. Curr Med Res Opin 2008; 24:537–550.
  50. Rosenstock J, Brazg R, Andryuk PJ, Lu K, Stein P; for the Sitagliptin Study 019 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin added to ongoing pioglitazone therapy in patients with type 2 diabetes: a 24-week, multicenter, randomized, double-blind, placebo-controlled, parallel-group study. Clin Ther 2006; 28:1556–1568.
  51. Chacra AR, Tan GH, Apanovitch A, Ravichandran S, List J, Chen R; for the CV181-040 Investigators. Saxagliptin added to a submaximal dose of sulphonylurea improves glycaemic control compared with uptitration of sulphonylurea in patients with type 2 diabetes: a randomised controlled trial. Int J Clin Pract 2009; 63:1395–1406.
  52. DeFronzo RA, Hissa MN, Garber AJ, et al. The efficacy and safety of saxagliptin when added to metformin therapy in patients with inadequately controlled type 2 diabetes with metformin alone. Diabetes Care 2009; 32:1649–1655.
  53. Jadzinsky M, Pfützner A, Paz-Pacheco E, Xu Z, Allen E, Chen R; for the CV181-039 Investigators. Saxagliptin given in combination with metformin as initial therapy improves glycaemic control in patients with type 2 diabetes compared with either monotherapy: a randomized controlled trial. Diabetes Obes Metab 2009; 11:611–622.
  54. Rosenstock J, Aguilar-Salinas C, Klein E, Nepal S, List J, Chen R; for the CV181-011 Study Investigators. Effect of saxagliptin monotherapy in treatment-naïve patients with type 2 diabetes. Curr Med Res Opin 2009; 25:2401–2411.
  55. Rosenstock J, Sankoh S, List JF. Glucose-lowering activity of the dipeptidyl peptidase-4 inhibitor saxagliptin in drug-naïve patients with type 2 diabetes. Diabetes Obes Metab 2008; 10:376–386.
  56. Dejager S, Razac S, Foley JE, Schweizer A. Vildagliptin in drug-naïve patients with type 2 diabetes: a 24-week, double-blind, randomized, placebo-controlled, multiple-dose study. Horm Metab Res 2007; 39:218–223.
  57. Garber AJ, Schweizer A, Baron MA, Rochotte E, Dejager S. Vildagliptin in combination with pioglitazone improves glycaemic control in patients with type 2 diabetes failing thiazolidinedione monotherapy: a randomized, placebo-controlled study. Diabetes Obes Metab 2007; 9:166–174.
  58. Göke B, Hershon K, Kerr D, et al. Efficacy and safety of vildagliptin monotherapy during 2-year treatment of drug-naïve patients with type 2 diabetes: comparison with metformin. Horm Metab Res 2008; 40:892–895.
  59. Pan C, Yang W, Barona JP, et al. Comparison of vildagliptin and acarbose monotherapy in patients with type 2 diabetes: a 24-week, double-blind, randomized trial. Diabet Med 2008; 25:435–441.
  60. Pi-Sunyer FX, Schweizer A, Mills D, Dejager S. Efficacy and tolerability of vildagliptin monotherapy in drug-naïve patients with type 2 diabetes. Diabetes Res Clin Pract 2007; 76:132–138.
  61. Schweizer A, Couturier A, Foley JE, Dejager S. Comparison between vildagliptin and metformin to sustain reductions in HbA(1c) over 1 year in drug-naïve patients with type 2 diabetes. Diabetes Med 2007; 24:955–961.
  62. Bolen S, Feldman L, Vassy J, et al. Systematic review: comparative effectiveness and safety of oral medications for type 2 diabetes mellitus. Ann Intern Med 2007; 147:386–399.
  63. Linnebjerg H, Park S, Kothare PA, et al. Effect of exenatide on gastric emptying and relationship to postprandial glycemia in type 2 diabetes. Regul Pept 2008; 151:123–129.
  64. Bunck MC, Diamant M, Cornér A, et al. One-year treatment with exenatide improves beta-cell function, compared with insulin glargine, in metformin-treated type 2 diabetic patients: a randomized, controlled trial. Diabetes Care 2009; 32:762–768.
  65. Merani S, Truong W, Emamaullee JA, Toso C, Knudsen LB, Shapiro AM. Liraglutide, a long-acting human glucagon-like peptide 1 analog, improves glucose homeostasis in marginal mass islet transplantation in mice. Endocrinology 2008; 149:4322–4328.
  66. Mari A, Degn K, Brock B, Rungby J, Ferrannini E, Schmitz O. Effects of the long-acting human glucagon-like peptide-1 analog liraglutide on beta-cell function in normal living conditions. Diabetes Care 2007; 30:2032–2033.
  67. Mari A, Nielsen LL, Nanayakkara N, DeFronzo RA, Ferrannini E, Halseth A. Mathematical modeling shows exenatide improved beta-cell function in patients with type 2 diabetes treated with metformin or metformin and a sulfonylurea. Horm Metab Res 2006; 38:838–844.
  68. Xu L, Man CD, Charbonnel B, et al. Effect of sitagliptin, a dipeptidyl peptidase-4 inhibitor, on beta-cell function in patients with type 2 diabetes: a model-based approach. Diabetes Obes Metab 2008; 10:1212–1220.
  69. Mari A, Scherbaum WA, Nilsson PM, et al. Characterization of the influence of vildagliptin on model-assessed b-cell function in patients with type 2 diabetes and mild hyperglycemia. J Clin Endocrinol Metab 2008; 93:103–109.
  70. Nathan DM, Buse JB, Davidson MB, et al. Medical management of hyperglycemia in type 2 diabetes: a consensus algorithm for the initiation and adjustment of therapy: a consensus statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care 2009; 32:193–203.
  71. Jellinger PS, Davidson JA, Blonde L, et al; for the ACE/AACE Diabetes Road Map Task Force. Road maps to achieve glycemic control in type 2 diabetes mellitus: ACE/AACE Diabetes Road Map Task Force. Endocr Pract 2007; 13:260–268.
References
  1. 2007 National Diabetes Fact Sheet. Centers for Disease Control and Prevention Web site. http://www.cdc.gov/diabetes/pubs/estimates07.htm. Updated: July 23, 2008. Accessed September 25, 2009.
  2. Wild S, Roglic G, Green A, Sicree R, King H. Global prevalence of diabetes: estimates for the year 2000 and projections for 2030. Diabetes Care 2004; 27:1047–1053.
  3. Prevalence of overweight and obesity among adults: United States 2003–2004. Centers for Disease Control and Prevention Web site. http://www.cdc.gov/nchs/products/pubs/pubd/hestats/overweight/overwght_adult_03.htm. Published: April 2006. Accessed September 23, 2009.
  4. Eeg-Olofsson K, Cederholm J, Nilsson PM, et al. Risk of cardiovascular disease and mortality in overweight and obese patients with type 2 diabetes: an observational study in 13,087 patients. Diabetologia 2009; 52:65–73.
  5. AACE Diabetes Mellitus Clinical Practice Guidelines Task Force. American Association of Clinical Endocrinologists medical guidelines for clinical practice for the management of diabetes mellitus. Endocr Pract 2007; 13(suppl 1):S4–S68.
  6. Bertin E, Schneider N, Abdelli N, et al. Gastric emptying is accelerated in obese type 2 diabetic patients without autonomic neuropathy. Diabetes Metab 2001; 27:357–364.
  7. Weytjens C, Keymeulen B, Van Haleweyn C, Somers G, Bossuyt A. Rapid gastric emptying of a liquid meal in long-term type 2 diabetes mellitus. Diabet Med 1998; 15:1022–1027.
  8. Stonehouse AH, Holcombe JH, Kendall DM. GLP-1 analogues, DPP-IV inhibitors and the metabolic syndrome. In: Fonseca V, ed. Therapeutic Strategies in Metabolic Syndrome. Oxford, UK: Atlas Medical Publishing Ltd; 2008: 137–157.
  9. DeFronzo RA. From the triumvirate to the ominous octet: a new paradigm for the treatment of type 2 diabetes mellitus. Diabetes 2009; 58:773–795.
  10. Nauck M, Stöckmann F, Ebert R, Creutzfeldt W. Reduced incretin effect in type 2 (non-insulin-dependent) diabetes. Diabetologia 1986; 29:46–52.
  11. Nauck MA, Heimesaat MM, Orskov C, Holst JJ, Ebert R, Creutzfeldt W. Preserved incretin activity of glucagon-like peptide 1 [7-36 amide] but not of synthetic human gastric inhibitory polypeptide in patients with type-2 diabetes mellitus. J Clin Invest 1993; 91:301–307.
  12. Deacon CF, Johnsen AH, Holst JJ. Degradation of glucagon-like peptide-1 by human plasma in vitro yields an N-terminally truncated peptide that is a major endogenous metabolite in vivo. J Clin Endocrinol Metab 1995; 80:952–957.
  13. Drucker DJ, Nauck MA. The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 2006; 368:1696–1705.
  14. Eng J, Kleinman WA, Singh L, Singh G, Raufman J-P. Isolation and characterization of exendin-4, an exendin-3 analogue, from Heloderma suspectum venom: further evidence for an exendin receptor on dispersed acini from guinea pig pancreas. J Biol Chem 1992; 267:7402–7405.
  15. Young AA, Gedulin BR, Bhavsar S, et al. Glucose-lowering and insulin-sensitizing actions of exendin-4: studies in obese diabetic (ob/ob, db/db) mice, diabetic fatty Zucker rats, and diabetic rhesus monkeys (Macaca mulatta). Diabetes 1999; 48:1026–1034.
  16. Kolterman OG, Buse JB, Fineman MS, et al. Synthetic exendin-4 (exenatide) significantly reduces postprandial and fasting plasma glucose in subjects with type 2 diabetes. J Clin Endocrinol Metab 2003; 88:3082–3089.
  17. Amori RE, Lau J, Pittas AG. Efficacy and safety of incretin therapy in type 2 diabetes: systematic review and meta-analysis. JAMA 2007; 298:194–206.
  18. Davidson JA, Parente EB, Gross JL. Incretin mimetics and dipeptidyl peptidase-4 inhibitors: innovative treatment therapies for type 2 diabetes. Arq Bras Endocrinol Metabol 2008; 52:1039–1049.
  19. Alexander GC, Sehgal NL, Moloney RM, Stafford RS. National trends in treatment of type 2 diabetes mellitus, 1994–2007. Arch Intern Med 2008; 168:2088–2094.
  20. US Department of Health and Human Services. FDA approves new drug treatment for type 2 diabetes. US Food and Drug Administration Web site. http://www.fda.gov/newsevents/newsroom/pressannouncements/ucm174780.htm. Published July 31, 2009. Accessed September 18, 2009.
  21. Drucker DJ, Buse JB, Taylor K, et al; for the DURATION-1 Study Group. Exenatide once weekly versus twice daily for the treatment of type 2 diabetes: a randomised, open-label, non-inferiority study. Lancet 2008; 372:1240–1250.
  22. Kim D, MacConell L, Zhuang D, et al. Effects of once-weekly dosing of a long-acting release formulation of exenatide on glucose control and body weight in subjects with type 2 diabetes. Diabetes Care 2007; 30:1487–1493.
  23. Marre M, Shaw J, Brändle M, et al; for the LEAD-1 SU Study Group. Liraglutide, a once-daily human GLP-1 analogue, added to a sulphonylurea over 26 weeks produces greater improvements in glycaemic and weight control compared with adding rosiglitazone or placebo in subjects with type 2 diabetes (LEAD-1 SU). Diabetes Med 2009; 26:268–278.
  24. Knudsen LB, Nielsen PF, Huusfeldt PO, et al. Potent derivatives of glucagon-like peptide 1 with pharmacokinetic properties suitable for once-daily administration. J Med Chem 2000; 43:1664–1669.
  25. Garber A, Henry R, Ratner R, et al; for the LEAD-3 (Mono) Study Group. Liraglutide versus glimepiride monotherapy for type 2 diabetes (LEAD-3 Mono): a randomised, 52-week, phase III, double-blind, parallel-treatment trial. Lancet 2009; 373:473–481.
  26. Nauck M, Frid A, Hermansen K, et al; for the LEAD-2 Study Group. Efficacy and safety comparison of liraglutide, glimepiride, and placebo, all in combination with metformin, in type 2 diabetes: the LEAD (Liraglutide Effect and Action in Diabetes)-2 study. Diabetes Care 2009; 32:84–90.
  27. Baggio LL, Drucker DJ, Maida A, Lamont BJ. ADA 2008: incretin-based therapeutics. MedscapeCME Web site. http://www.medscape.com/viewprogram/15786. Accessed September 18, 2009.
  28. Fleck P, Christopher R, Covington P, Wilson C, Mekki Q. Efficacy and safety of alogliptin monotherapy over 12 weeks in patients with type 2 diabetes. Paper presented at: 68th Annual Meeting of the American Diabetes Association; June 6–10, 2008; San Francisco, CA. Abstract 479-P.
  29. DeFronzo RA, Burant CF, Fleck P, Wilson C, Mekki Q, Pratley RE. Effect of alogliptin combined with pioglitazone on glycemic control in metformin-treated patients with type 2 diabetes. Paper presented at: 69th Annual Meeting of the American Diabetes Association; June 5–9, 2009; New Orleans, LA. Abstract 2024-PO.
  30. Nauck M, Ellis G, Fleck P, Wilson C, Mekki Q. Efficacy and safety of alogliptin added to metformin therapy in patients with type 2 diabetes. Paper presented at: 68th Annual Meeting of the American Diabetes Association; June 6–10, 2008; San Francisco, CA. Abstract 477-P.
  31. DeFronzo RA, Ratner RE, Han J, Kim DD, Fineman MS, Baron AD. Effects of exenatide (exendin-4) on glycemic control and weight over 30 weeks in metformin-treated patients with type 2 diabetes. Diabetes Care 2005; 28:1092–1100.
  32. Kendall DM, Riddle MC, Rosenstock J, et al. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in patients with type 2 diabetes treated with metformin and a sulfonylurea. Diabetes Care 2005; 28:1083–1091.
  33. Zinman B, Hoogwerf BJ, Durán García S, et al. The effect of adding exenatide to a thiazolidinedione in suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med 2007; 146:477–485.
  34. Barnett AH, Burger J, Johns D, et al. Tolerability and efficacy of exenatide and titrated insulin glargine in adult patients with type 2 diabetes previously uncontrolled with metformin or a sulfonylurea: a multinational, randomized, open-label, two-period, crossover noninferiority trial. Clin Ther 2007; 29:2333–2348.
  35. Heine RJ, Van Gaal LF, Johns D, Mihm MJ, Widel MH, Brodows RG; for the GWAA Study Group. Exenatide versus insulin glargine in patients with suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med 2005; 143:559–569.
  36. Nauck MA, Duran S, Kim D, et al. A comparison of twice-daily exenatide and biphasic insulin aspart in patients with type 2 diabetes who were suboptimally controlled with sulfonylurea and metformin: a non-inferiority study. Diabetologia 2007; 50:259–267.
  37. Klonoff DC, Buse JB, Nielsen LL, et al. Exenatide effects on diabetes, obesity, cardiovascular risk factors and hepatic biomarkers in patients with type 2 diabetes treated for at least 3 years. Curr Med Res Opin 2008; 24:275–286.
  38. Moretto TJ, Milton DR, Ridge TD, et al. Efficacy and tolerability of exenatide monotherapy over 24 weeks in antidiabetic drug-naïve patients with type 2 diabetes: a randomized, double-blind, placebo-controlled, parallel-group study. Clin Ther 2008; 30:1448–1460.
  39. DeFronzo RA, Okerson T, Viswanathan P, Guan X, Holcombe JH, MacConell L. Effects of exenatide versus sitagliptin on postprandial glucose, insulin and glucagon secretion, gastric emptying, and caloric intake: a randomized, cross-over study. Curr Med Res Opin 2008; 24:2943–2952.
  40. Seino Y, Rasmussen MF, Zdravkovic M, Kaku K. Dose-dependent improvement in glycemia with once-daily liraglutide without hypoglycemia or weight gain: a double-blind, randomized, controlled trial in Japanese patients with type 2 diabetes. Diabetes Res Clin Pract 2008; 81:161–168.
  41. Vilsbøll T, Zdravkovic M, Le-Thi T, et al. Liraglutide, a long-acting human glucagon-like peptide-1 analog, given as monotherapy significantly improves glycemic control and lowers body weight without risk of hypoglycemia in patients with type 2 diabetes. Diabetes Care 2007; 30:1608–1610.
  42. Aschner P, Kipnes MS, Lunceford JK, Sanchez M, Mickel C, Williams-Herman DE; for the Sitagliptin Study 021 Group. Effect of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy on glycemic control in patients with type 2 diabetes. Diabetes Care 2006; 29:2632–2637.
  43. Nonaka K, Kakikawa T, Sato A, et al. Efficacy and safety of sitagliptin monotherapy in Japanese patients with type 2 diabetes. Diabetes Res Clin Pract 2008; 79:291–298.
  44. Raz I, Hanefeld M, Xu L, Caria C, Williams-Herman D, Khatami H; for the Sitagliptin Study 023 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy in patients with type 2 diabetes mellitus. Diabetologia 2006; 49:2564–2571.
  45. Scott R, Wu M, Sanchez M, Stein P. Efficacy and tolerability of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy over 12 weeks in patients with type 2 diabetes. Int J Clin Pract 2007; 61:171–180.
  46. Charbonnel B, Karasik A, Liu J, Wu M, Meininger G; for the Sitagliptin Study 020 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin added to ongoing metformin therapy in patients with type 2 diabetes inadequately controlled with metformin alone. Diabetes Care 2006; 29:2638–2643.
  47. Hermansen K, Kipnes M, Luo E, Fanurik D, Khatami H, Stein P; for the Sitagliptin Study 035 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor, sitagliptin, in patients with type 2 diabetes mellitus inadequately controlled on glimepiride alone or on glimepiride and metformin. Diabetes Obes Metab 2007; 9:733–745.
  48. Nauck MA, Meininger G, Sheng D, Terranella L, Stein PP; for the Sitagliptin Study 024 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor, sitagliptin, compared with the sulfonylurea, glipizide, in patients with type 2 diabetes inadequately controlled on metformin alone: a randomized, double-blind, non-inferiority trial. Diabetes Obes Metab 2007; 9:194–205.
  49. Raz I, Chen Y, Wu M, et al. Efficacy and safety of sitagliptin added to ongoing metformin therapy in patients with type 2 diabetes. Curr Med Res Opin 2008; 24:537–550.
  50. Rosenstock J, Brazg R, Andryuk PJ, Lu K, Stein P; for the Sitagliptin Study 019 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin added to ongoing pioglitazone therapy in patients with type 2 diabetes: a 24-week, multicenter, randomized, double-blind, placebo-controlled, parallel-group study. Clin Ther 2006; 28:1556–1568.
  51. Chacra AR, Tan GH, Apanovitch A, Ravichandran S, List J, Chen R; for the CV181-040 Investigators. Saxagliptin added to a submaximal dose of sulphonylurea improves glycaemic control compared with uptitration of sulphonylurea in patients with type 2 diabetes: a randomised controlled trial. Int J Clin Pract 2009; 63:1395–1406.
  52. DeFronzo RA, Hissa MN, Garber AJ, et al. The efficacy and safety of saxagliptin when added to metformin therapy in patients with inadequately controlled type 2 diabetes with metformin alone. Diabetes Care 2009; 32:1649–1655.
  53. Jadzinsky M, Pfützner A, Paz-Pacheco E, Xu Z, Allen E, Chen R; for the CV181-039 Investigators. Saxagliptin given in combination with metformin as initial therapy improves glycaemic control in patients with type 2 diabetes compared with either monotherapy: a randomized controlled trial. Diabetes Obes Metab 2009; 11:611–622.
  54. Rosenstock J, Aguilar-Salinas C, Klein E, Nepal S, List J, Chen R; for the CV181-011 Study Investigators. Effect of saxagliptin monotherapy in treatment-naïve patients with type 2 diabetes. Curr Med Res Opin 2009; 25:2401–2411.
  55. Rosenstock J, Sankoh S, List JF. Glucose-lowering activity of the dipeptidyl peptidase-4 inhibitor saxagliptin in drug-naïve patients with type 2 diabetes. Diabetes Obes Metab 2008; 10:376–386.
  56. Dejager S, Razac S, Foley JE, Schweizer A. Vildagliptin in drug-naïve patients with type 2 diabetes: a 24-week, double-blind, randomized, placebo-controlled, multiple-dose study. Horm Metab Res 2007; 39:218–223.
  57. Garber AJ, Schweizer A, Baron MA, Rochotte E, Dejager S. Vildagliptin in combination with pioglitazone improves glycaemic control in patients with type 2 diabetes failing thiazolidinedione monotherapy: a randomized, placebo-controlled study. Diabetes Obes Metab 2007; 9:166–174.
  58. Göke B, Hershon K, Kerr D, et al. Efficacy and safety of vildagliptin monotherapy during 2-year treatment of drug-naïve patients with type 2 diabetes: comparison with metformin. Horm Metab Res 2008; 40:892–895.
  59. Pan C, Yang W, Barona JP, et al. Comparison of vildagliptin and acarbose monotherapy in patients with type 2 diabetes: a 24-week, double-blind, randomized trial. Diabet Med 2008; 25:435–441.
  60. Pi-Sunyer FX, Schweizer A, Mills D, Dejager S. Efficacy and tolerability of vildagliptin monotherapy in drug-naïve patients with type 2 diabetes. Diabetes Res Clin Pract 2007; 76:132–138.
  61. Schweizer A, Couturier A, Foley JE, Dejager S. Comparison between vildagliptin and metformin to sustain reductions in HbA(1c) over 1 year in drug-naïve patients with type 2 diabetes. Diabetes Med 2007; 24:955–961.
  62. Bolen S, Feldman L, Vassy J, et al. Systematic review: comparative effectiveness and safety of oral medications for type 2 diabetes mellitus. Ann Intern Med 2007; 147:386–399.
  63. Linnebjerg H, Park S, Kothare PA, et al. Effect of exenatide on gastric emptying and relationship to postprandial glycemia in type 2 diabetes. Regul Pept 2008; 151:123–129.
  64. Bunck MC, Diamant M, Cornér A, et al. One-year treatment with exenatide improves beta-cell function, compared with insulin glargine, in metformin-treated type 2 diabetic patients: a randomized, controlled trial. Diabetes Care 2009; 32:762–768.
  65. Merani S, Truong W, Emamaullee JA, Toso C, Knudsen LB, Shapiro AM. Liraglutide, a long-acting human glucagon-like peptide 1 analog, improves glucose homeostasis in marginal mass islet transplantation in mice. Endocrinology 2008; 149:4322–4328.
  66. Mari A, Degn K, Brock B, Rungby J, Ferrannini E, Schmitz O. Effects of the long-acting human glucagon-like peptide-1 analog liraglutide on beta-cell function in normal living conditions. Diabetes Care 2007; 30:2032–2033.
  67. Mari A, Nielsen LL, Nanayakkara N, DeFronzo RA, Ferrannini E, Halseth A. Mathematical modeling shows exenatide improved beta-cell function in patients with type 2 diabetes treated with metformin or metformin and a sulfonylurea. Horm Metab Res 2006; 38:838–844.
  68. Xu L, Man CD, Charbonnel B, et al. Effect of sitagliptin, a dipeptidyl peptidase-4 inhibitor, on beta-cell function in patients with type 2 diabetes: a model-based approach. Diabetes Obes Metab 2008; 10:1212–1220.
  69. Mari A, Scherbaum WA, Nilsson PM, et al. Characterization of the influence of vildagliptin on model-assessed b-cell function in patients with type 2 diabetes and mild hyperglycemia. J Clin Endocrinol Metab 2008; 93:103–109.
  70. Nathan DM, Buse JB, Davidson MB, et al. Medical management of hyperglycemia in type 2 diabetes: a consensus algorithm for the initiation and adjustment of therapy: a consensus statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care 2009; 32:193–203.
  71. Jellinger PS, Davidson JA, Blonde L, et al; for the ACE/AACE Diabetes Road Map Task Force. Road maps to achieve glycemic control in type 2 diabetes mellitus: ACE/AACE Diabetes Road Map Task Force. Endocr Pract 2007; 13:260–268.
Page Number
S28-S38
Page Number
S28-S38
Publications
Publications
Article Type
Display Headline
Advances in therapy for type 2 diabetes: GLP–1 receptor agonists and DPP–4 inhibitors
Display Headline
Advances in therapy for type 2 diabetes: GLP–1 receptor agonists and DPP–4 inhibitors
Citation Override
Cleveland Clinic Journal of Medicine 2009 December;76(suppl 5):S28-S38
Inside the Article

KEY POINTS

  • Hormonal deficiencies in T2DM are related to abnormalities in the secretion of amylin, glucagon, and incretin hormones.
  • In clinical trials, GLP-1 receptor agonists reduced HbA1c levels, had beneficial effects on weight, and caused less hypoglycemia than insulin analogues.
  • Both GLP-1 receptor agonists and DPP-4 inhibitors improve pancreatic beta-cell function.
  • Incretin-based therapies have been incorporated into recently updated clinical guidelines for treatment of T2DM.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Fri, 04/13/2018 - 09:45
Un-Gate On Date
Fri, 04/13/2018 - 09:45
Use ProPublica
CFC Schedule Remove Status
Fri, 04/13/2018 - 09:45
Article PDF Media

Redefining treatment success in type 2 diabetes mellitus: Comprehensive targeting of core defects

Article Type
Changed
Tue, 10/02/2018 - 11:46
Display Headline
Redefining treatment success in type 2 diabetes mellitus: Comprehensive targeting of core defects

According to the American Association of Clinical Endocrinologists (AACE) and the American Diabetes Association (ADA), glycosylated hemoglobin (HbA1c) in patients with diabetes should be maintained at 6.5% or less (AACE) or at less than 7.0% (ADA). Both organizations support an aggressive stepwise approach that includes medication and lifestyle modification, with strategies and clinical attention devoted to avoiding significant hypoglycemia.1,2 Yet, despite the introduction of new antidiabetes agents, most current management strategies are offset by limitations in achieving and maintaining glycemic targets needed to provide optimal care for patients with diabetes, more than 90% of whom have type 2 diabetes mellitus (T2DM).3,4

Nationally, glycemic control among patients with T2DM has improved but is still far from optimal. According to data from the 1999–2000 National Health and Nutrition Examination Survey (NHANES), glycemic control (HbA1c < 7.0%) rates were 35.8% for patients with T2DM.5 In a more recent report (NHANES 1999–2004), fewer than half (48.4%) of adult patients with diagnosed diabetes achieved HbA1c levels below 7.0%.5,6 Factors contributing to these data include earlier onset and earlier detection of T2DM.7

CHANGING TREATMENT TRENDS

Available treatments for patients with T2DM include secretagogues, such as sulfonylureas and “glinides” (repaglinide and nateglinide), metformin, thiazolidinediones (TZDs), and dipeptidyl peptidase–4 (DPP-4) inhibitors among oral medications, and insulin and glucagon-like peptide–1 (GLP-1) receptor agonists among parenterally administered agents. According to the latest published data on prescribing patterns for patients with T2DM, analyses of the National Disease and Therapeutic Index (1994–2007) and the National Prescription Audit (2001–2007), sulfonylurea use decreased from 67% of treatment visits in 1994 to 34% of visits in 2007.8 By 2007, metformin, used in 54% of treatment visits, and TZDs, used in 28%, were the most frequently administered antidiabetes agents. Insulin use declined from 38% of visits during which a treatment was administered in 1994 to 25% of visits in 2000, but had increased subsequently to 28% of visits in 2007.

SIGNIFICANCE OF CARDIOVASCULAR RISK

Clinical research has suggested that focusing solely on improving glycemic control may be insufficient to reduce overall morbidity and mortality associated with diabetes. Specifically, data from recent studies, including the Action to Control Cardiovascular Risk in Diabetes (ACCORD), the Action in Diabetes and Vascular Disease: Preterax and Diamicron Modified Release Controlled Evaluation (ADVANCE), and the Veterans Affairs Diabetes Trial (VADT), emphasized that lowering HbA1c below 7% in a high-risk population of individuals with T2DM did not improve cardiovascular (CV) outcomes.9–11 The observations confirm that risk factors, including weight, blood pressure (BP), and lipid levels, are vitally important in reducing morbidity and mortality in this population. This perception is further underscored by the NHANES 1999–2004 data, which showed poor concurrent control of HbA1c, BP, and lipids; only 13.2% of patients with diagnosed diabetes achieved all three target goals simultaneously.6 Similarly, a nationwide survey in Norway showed that only 13% of patients with T2DM concurrently achieved goals for HbA1c, BP, and lipids.12

In the Danish Steno-2 Study, patients with T2DM and persistent microalbuminuria were treated with either intensive target-driven therapy using multiple drugs or conventional multifactorial treatment. Over a mean period of 13.3 years (7.8 years of treatment plus 5.5 years of follow-up), intensive multifactorial intervention to control multiple CV risk factors, including HbA1c, BP, and lipids, was associated with a lower risk of death from CV causes (hazard ratio [HR], 0.43; 95% confidence interval [CI], 0.19 to 0.94; P = .04) and a lower risk of CV events (HR, 0.41; 95% CI, 0.25 to 0.67; P < .001) than was conventional therapy.13

This article clarifies the redefinition of treatment success in patients with T2DM based on targeting the underlying physiologic defects of the disease.

T2DM, OVERWEIGHT/OBESITY, AND CV DISEASE: CLOSELY LINKED

The incidence and prevalence of T2DM, overweight/obesity, and CV disease (CVD) are increasing worldwide. It is estimated that the worldwide prevalence of diabetes will increase from 171 million in 2000 to 366 million by 203014; T2DM increases the risk of morbidity and mortality from microvascular (eg, neuropathic, retinopathic, nephropathic) and macrovascular (eg, coronary, peripheral vascular disease) complications.15 According to a Michigan health maintenance organization study (N = 1,364), the median annual direct cost of medical care for Caucasian patients with T2DM who were diet controlled, had a body mass index (BMI) of 30 kg/m2 or higher, and had no vascular complications was estimated to be $1,700 for men and $2,100 for women.16 The actual cost of care for patients with T2DM may be much higher, since most patients present with multiple CV risk factors in addition to being overweight.

NHANES data show that approximately two-thirds of Americans are either overweight or obese17; overweight/obesity affects about 80% of adults diagnosed with T2DM.18 Overweight or obesity can increase the risk for developing T2DM by more than 90-fold and, in women, it can increase the risk for developing coronary heart disease (CHD) by sixfold.19 The close link between T2DM and CVD is underscored further with recent data from the Framingham Heart Study, which showed a high lifetime risk of CVD in patients with diabetes, heightened further by obesity. During the 30-year study period, the lifetime risk of CVD in normal-weight people with diabetes was 78.6% in men and 54.8% in women; the risk increased to 86.9% in obese men with diabetes and to 78.8% in obese women with diabetes.20 The NHANES data also showed that the prevalence of T2DM increased in the past decade and that patients are being diagnosed at a younger age, from a mean age of 52 years in 1988–1994 to 46 years in 1999–2000.7

 

 

BRIDGING THE GAP FROM PATHOPHYSIOLOGY TO UNMET NEEDS

The paradigm behind the pathophysiology of T2DM has shifted from its perception as a simple “dual-defect” disease (ie, deficiency in insulin secretion and peripheral tissue insulin resistance) to a multidimensional disorder.1,21 This new model includes overweight/obesity, insulin resistance, qualitative and quantitative defects in insulin secretion, and dysregulation in the secretion of other hormones, including the beta-cell hormone amylin, the alpha-cell hormone glucagon, and the gastrointestinal incretin hormones GLP-1 and glucose-dependent insulinotropic polypeptide.21–23

The major target of antidiabetes agents is glycemic control, assessed by a reduction in HbA1c, but their effects on other metabolic factors and their adverse effects differ with each agent (Table 1).3 Whereas metformin and alpha-glucosidase inhibitors may help normalize glycemia with weight-neutral effects, many other agents, including insulin and its analogues, the “glinides,” first- and second-generation sulfonylureas, and TZDs, are associated with weight gain.23,24 In addition, the propensity to induce hypoglycemia differs among agents and clearly reflects the mechanism of action of each drug. The observed limitations of older therapies treating a progressive disease that is associated with a number of comorbid conditions supports the need for continued development of new antidiabetes agents.

CLINICAL GUIDELINES AND CV RISK FACTOR MANAGEMENT

The best strategy for managing T2DM is a comprehensive approach that addresses the fundamental core defects plus associated factors that contribute to increased CV risk. Several specialty groups have suggested guidelines and algorithms for the management of T2DM and its comorbidities. These guidelines, including the ADA standards of medical care, the AACE standards in tandem with the American College of Endocrinology guidelines, and the recent joint statement from the ADA and the European Association for the Study of Diabetes (EASD), acknowledge that the core defects of T2DM and the associated CV risk factors (eg, weight gain, obesity, hypertension, dyslipidemia) are important in developing optimal treatment strategies.1–3 Medical nutrition guidelines advocate weight loss as a key initial step in managing T2DM and the comorbidities that lead to elevated CV risk.25,26 The National Institutes of Health and the US Department of Health and Human Services/US Department of Agriculture advocate regular physical activity, dietary assessment, and periodic comorbidity and weight assessment for all people, not just those with T2DM or CVD.26,27

Weight reduction

Evidence in support of effective lifestyle intervention was demonstrated in the Action for Health in Diabetes (Look AHEAD) study. After 1 year, patients with T2DM treated with intensive lifestyle intervention lost an average of 8.6% of their initial weight compared with 0.7% in patients treated only with diabetes support and education (P < 0.001). The intensive-intervention patients also had a significant drop in HbA1c (from 7.3% to 6.6%; P < 0.001) and were able to reduce their antidiabetes, antihypertensive, and lipid-lowering medications.28 More recent data from the Look AHEAD study reported that overweight patients with T2DM enrolled in a weight management program experienced significant weight loss, improved physical fitness, reduced physical symptoms, and overall improvement in health-related quality of life.29 Thus, weight reduction appears to be a key component in reducing CV risk and improving quality of life in most patients with T2DM.28–30

Hypertension

Hypertension is a major risk factor for microvascular complications and CVD, and may be associated with, or be the underlying result of, nephropathy.2 BP control is clearly important in reducing the morbidity and mortality associated with T2DM. The recommended BP goal in patients with T2DM is less than 130/80 mm Hg.1,2

Hyperlipidemia

According to the Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III [ATP III]), diabetes is considered a CHD risk equivalent because it confers a high risk of new CHD developing within 10 years.31 In addition to the NCEP–ATP III guidelines, the ADA and the AACE have set target levels for lipids in patients with diabetes, including T2DM.1,2,31 All three organizations have defined 100 mg/dL as the target level for low-density lipoprotein.

HbA1c and lifestyle intervention

Figure 1. Suggested algorithm for the metabolic management of patients with type 2 diabetes mellitus. Clinicians should reinforce lifestyle interventions at every visit and check glycosylated hemoglobin (HbA1c) every 3 months until it is less than 7.0%, and then check it at least every 6 months. The interventions should be adjusted if HbA1c is 7.0% or greater.
The American Heart Association and the ADA initiated a call to action for global risk assessment for CVD and diabetes.32 According to their joint scientific statement, lifestyle intervention should be reinforced at every physician visit, and HbA1c should be monitored every 3 months until it is less than 7.0% and then rechecked every 6 months. Adjustments in intervention should be made if the HbA1c level is 7.0% or higher.3 A recent joint statement from the ADA and the EASD revised an earlier treatment algorithm for the initiation of therapy in patients with T2DM; the revision includes incretin therapies (ie, GLP-1 receptor agonists) as a tier 2 option, especially in patients in whom hypoglycemia and weight gain are concerns (Figure 1).3

 

 

EVOLUTION OF ANTIDIABETES THERAPIES

Traditional antidiabetes agents used in the treatment of patients with T2DM have focused mainly on insulin secretion and insulin resistance, with treatment success defined as achieving HbA1c goals with a reduced incidence of hypoglycemia.23 Secretagogues, such as sulfonylureas and glinides, stimulate the pancreas to release insulin. Insulin sensitizers, such as TZDs and metformin, enhance the action of insulin in muscle and fat1,3,23 and lower hepatic glucose production. The alpha-glucosidase inhibitors alter carbohydrate absorption from the gastrointestinal tract.1 The extent to which each agent achieves treatment success in terms of glucose lowering depends on several factors, including intrinsic attributes, duration of disease, and baseline glycemic control.3

Newer agents for the treatment of T2DM include the incretin-based therapies—GLP-1 receptor agonists and DPP-4 inhibitors—which influence mechanisms beyond increasing pancreatic insulin secretion and decreasing peripheral insulin resistance (Table 2).22 The GLP-1 signaling pathway has been leveraged by two distinct pharmacologic approaches. The first involves the use of synthetic peptides with glucoregulatory effects similar to those of endogenous GLP-1 (GLP-1 receptor agonists). The second involves the use of DPP-4 inhibitors, small molecules that inhibit the proteolytic activity of DPP-4, leading to enhanced endogenous GLP-1 concentrations.22

GLP-1 receptor agonists

Exenatide effects. Although many agents are in development, to date exenatide is the only GLP-1 receptor agonist approved by the US Food and Drug Administration (FDA).8,33 Exenatide is an exendin-4 GLP-1 receptor agonist with multiple glucoregulatory effects, including enhanced glucose-dependent insulin secretion, reduced glucagon secretion and food intake, and slowed gastric emptying.22,34 Exenatide is detectable in the circulation for up to 10 hours following subcutaneous (SC) administration22 and has a greater potency in reducing plasma glucose than GLP-1 in preclinical studies.35,36

By virtue of its beneficial effects on glycemic control, weight, BP, and lipids, exenatide addresses some of the components of the metabolic syndrome.37–41 In pivotal 30-week studies, exenatide was associated with HbA1c reductions that ranged from –0.40% to –0.86% from baseline and decreases in body weight of approximately –1 kg to –3 kg from baseline, without severe hypoglycemia.37–39 The percentage of patients who reached the ADA goal of HbA1c less than 7.0% at 30 weeks ranged from 24% to 34%. The addition of exenatide to TZD therapy in a 16-week study was associated with mean reductions in HbA1c of –0.98%, fasting plasma glucose (FPG) concentration of –1.69 mmol/L (–30.42 mg/dL), and body weight of –1.51 kg.40

A posthoc analysis of an open-label extension study involving patients who completed the original 30-week placebo-controlled studies showed that 46% of patients who remained on exenatide achieved the ADA goal of HbA1c less than 7.0% at 3 years.41 Exenatide administered for up to 3.5 years was associated with sustained reductions in HbA1c of –1.0% (P < .0001) and body weight of –5.3 kg (P < .001). Pancreatic beta-cell function, assessed by homeostasis model assessment, improved, as did BP, triglyceride, high-density lipoprotein, low-density lipoprotein, and aspartate aminotransferase levels.41

Comparison with insulin analogues. Comparative studies have highlighted the contrasting effects of exenatide and insulin analogues (eg, insulin glargine and fixed-ratio insulin).42–45 In a 26-week trial comparing exenatide with insulin glargine in subjects with T2DM, both agents resulted in similar decreases in HbA1c. Exenatide was also associated with a –2.3-kg weight reduction, whereas insulin glargine was associated with a +1.8-kg weight gain.42 Although rates of symptomatic hypoglycemia were similar, there were fewer cases of nocturnal hypoglycemia with exenatide (0.9 event/patient-year vs 2.4 events/patient-year with insulin).

In a 32-week study comparing exenatide BID with titrated insulin glargine QD, the HbA1c reductions for exenatide and insulin glargine were comparable. However, body weight decreased –4.2 kg over two 16-week treatment periods with exenatide, but increased +3.3 kg over the same periods with the basal insulin analogue.43 The incidence of hypoglycemia was lower with exenatide than with insulin glargine (14.7% vs 25.2%), although the difference was not statistically significant.

In another study that compared exenatide with biphasic insulin aspart, patients who were treated with exenatide also lost weight while those who received the fast-acting insulin analogue gained weight (between-group difference, –5.4 kg). Patients treated with exenatide also demonstrated greater reductions in postprandial plasma glucose (PPG) excursions following their morning (P < .001), midday (P = .002), and evening meals (P < .001).44 Overall, hypoglycemia rates were similar at study end between exenatide and insulin aspart (4.7 events/patient-year vs 5.6 events/patient-year). In all of these studies, significant gastrointestinal adverse events (nausea and vomiting) occurred more frequently with exenatide, and more patients withdrew from exenatide than from insulin.

Formulations in development. Other advances in GLP-1 receptor agonist therapy include novel formulations under clinical development, such as exenatide once weekly36,46 and liraglutide, a human analogue GLP-1 receptor agonist formulated for once-daily administration.47,48 In a 52-week study in patients with T2DM, liraglutide significantly reduced HbA1c; the 1.2-mg SC QD dosage reduced HBA1c by –0.84% (P = .0014) and the 1.8-mg SC QD dosage by –1.14% (P < .0001). In comparison, glimepiride 8 mg orally QD achieved a –0.51% reduction. Liraglutide was also associated with greater reductions in weight, hypoglycemia, and systolic BP than glimepiride.47

A 26-week study compared liraglutide (0.6, 1.2, and 1.8 mg SC QD), placebo, and glimepiride 4 mg QD in combination with metformin 1 g BID. HbA1c was reduced significantly in all liraglutide groups compared with placebo (P < .0001). Mean HbA1c decreased –1.0% with liraglutide 1.2 mg and 1.8 mg and with glimepiride; it decreased –0.7% with liraglutide 0.6 mg; and it increased +0.1% with placebo. Body weight decreased –1.8 kg to –2.8 kg in all liraglutide groups but increased +1.0 kg in the glimepiride group (P < .0001). The incidence of minor hypoglycemia with liraglutide (~3%) was comparable to that observed with placebo but less than that with glimepiride (17%; P < .001).48

A once-weekly long-acting release (LAR) formulation of exenatide submitted to the FDA for approval may provide enhanced glycemic and weight control, potentially improving patient acceptance and adherence.36,46 In a 15-week study, exenatide once weekly produced significant reductions in HbA1c, FPG, PPG, and body weight. There were no withdrawals due to adverse events, and the formation of anti-exenatide antibodies was not predictive of therapeutic end point response or adverse safety outcome. Instances of hypoglycemia were mild and not dose related.36 In a 30-week study comparing exenatide LAR once weekly with exenatide BID, patients given exenatide LAR once weekly had significantly greater HbA1c reductions than did patients given exenatide BID (–1.9% vs –1.5%; P = .0023). Treatment adherence was 98% with both exenatide regimens, and no episodes of major hypoglycemia occurred with either formulation regardless of background sulfonylurea use. Favorable effects on BP and lipid profile were observed with both exenatide regimens.46

 

 

DPP-4 inhibitors

The DPP-4 inhibitors (commonly called gliptins) inhibit the proteolytic cleavage of circulating GLP-1 by binding to the DPP-4 enzyme, increasing the concentration of endogenous GLP-1 approximately two- to threefold.49–51 These concentrations result in more prompt and appropriate secretion of insulin and suppression of glucagon in response to a carbohydrate-containing snack or meal, with the change in glucagon correlating linearly with improved glucose tolerance.51

DPP-4 inhibitors, which are given orally, include sitagliptin and saxagliptin (approved in the United States) and vildagliptin (not approved in the United States but used in the European Union and Latin America).8,22,33,52 Sitagliptin can be used either as monotherapy or in combination with metformin or a TZD.8,49–55 Recently, a single-tablet formulation of sitagliptin plus metformin was granted regulatory approval.8

When used alone or in combination with metformin or pioglitazone, sitagliptin has been associated with significant reductions in HbA1c (of ~0.5% to 0.6% when used alone, ~0.7% with metformin, and ~0.9% with pioglitazone [P < .001 vs placebo]), with hypoglycemia occurring in 1.3% or less of the population.54 In an 18-week study in which patients with T2DM who were inadequately controlled with metformin monotherapy were randomized to receive add-on sitagliptin (100 mg QD), rosiglitazone (8 mg QD), or placebo, sitagliptin reduced HbA1c –0.73% (P < .001 vs placebo) and reduced body weight –0.4 kg, while rosiglitazone reduced HbA1c –0.79% and increased body weight +1.5 kg.55

To evaluate the effectiveness of sitagliptin and metformin as initial therapy, a 54-week study was completed in 885 patients with T2DM and inadequate glycemic control (HbA1c 7.5–11%) on diet and exercise.56 Patients were evaluated on monotherapy with either sitagliptin (100 mg QD) or metformin (1 g or 2 g QD), or on initial therapy with the two in combination (sitagliptin 100 mg + metformin 1 mg or 2 mg QD). At week 54, in the all-patients-treated analysis, mean changes in HbA1c from baseline were –1.8% with sitagliptin plus metformin 2 g QD, –1.4% with sitagliptin plus metformin 1 g QD, –1.3% with metformin 2 g QD monotherapy, –1.0% with metformin 1 g QD monotherapy, and –0.8% with sitagliptin 100 mg QD monotherapy.

All treatments improved measures of beta-cell function (eg, homeostasis model assessment [HOMA]-beta, proinsulin/insulin ratio). Mean body weight decreased from baseline in the combination and metformin monotherapy groups and was unchanged from baseline in the sitagliptin monotherapy group. The incidence of hypoglycemia was low (1%–3%) across treatment groups. The incidence of gastrointestinal adverse experiences was evaluated with the coadministration of sitagliptin and metformin and appeared similar to that observed with use of metformin as monotherapy.56 Thus, this study suggested that an initial combination of a DPP-IV inhibitor with metformin can improve glycemic control and markers of beta-cell function in patients with T2DM.

Incretin-based therapies compared

Studies in both healthy individuals and in patients with T2DM have shown that oral DPP-4 inhibitors such as sitagliptin increase endogenous GLP-1 concentrations by about twofold compared with placebo.22,50 The pharma­cologic concentration of subcutaneously administered exenatide available for activating the GLP-1 receptor is significantly greater than the increased endogenous GLP-1 concentrations achieved with sitagliptin. In a recent clinical study comparing exenatide and sitagliptin in patients with T2DM, the mean 2-hour plasma concentration for exenatide was 64 pM compared with the mean 2-hour postprandial GLP-1 concentration of 15 pM for sitagliptin (baseline GLP-1 concentration was 7.2 pM).57 While both agents were shown to be effective, exenatide appeared to have had a greater effect than sitagliptin in increasing insulin secretion and reducing postprandial glucagon secretion, leading to significantly (P < 0.0001) greater reductions in PPG.57

Sitagliptin has been minimally associated with nausea, whereas patients who take exenatide need to be informed of the risk of usually mild to moderate, but sometimes severe, nausea and vomiting that tends to decrease over time.

For a detailed comparison of the effects of GLP-1 receptor agonists and DPP-4 inhibitors on HbA1c, weight, and hypoglycemia, see “Advances in therapy for type 2 diabetes: GLP–1 receptor agonists and DPP–4 inhibitors.”

CONCLUSION

Despite advances in diagnosis and treatment, T2DM, overweight/obesity, CVD, and their complications remain major public health burdens worldwide. The concepts that explain the pathophysiology of T2DM include the contribution of various factors beyond insulin secretion and insulin resistance, such as the role of incretin hormones in disease progression. A comprehensive approach to managing patients with T2DM requires targeting the fundamental defects of the disease and its comorbidities. Newer agents, including incretin-based therapies such as GLP-1 receptor agonists and DPP-4 inhibitors, address the fundamental defects of T2DM. The definition of treatment success in the management of T2DM will be redefined as more data become available on agents that exert beneficial effects not only on glycemia but on parameters that may influence overall CV health, such as weight, BP, and lipid profiles.

References
  1. AACE Diabetes Mellitus Clinical Practice Guidelines Task Force. American Association of Clinical Endocrinologists medical guidelines for clinical practice for the management of diabetes mellitus. Endocr Pract 2007; 13(suppl 1):S4–S68.
  2. American Diabetes Association. Standards of medical care in diabetes—2009. Diabetes Care 2009; 32(suppl 1):S13–S61.
  3. Nathan DM, Buse JB, Davidson MB, et al. Medical management of hyperglycemia in type 2 diabetes: a consensus algorithm for the initiation and adjustment of therapy: a consensus statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care 2009; 32:193–203.
  4. National Institute of Diabetes and Digestive and Kidney Diseases. National Diabetes Statistics, 2007 fact sheet. National Institutes of Health Web site. http://www.diabetes.niddk.nih.gov/dm/pubs/statistics/index.htm. Published 2008. Accessed September 16, 2009.
  5. Koro CE, Bowlin SJ, Bourgeois N, Fedder DO. Glycemic control from 1988 to 2000 among US adults diagnosed with type 2 diabetes: a preliminary report. Diabetes Care 2004; 27:17–20.
  6. Ong KL, Cheung BM, Wong LY, Wat NM, Tan KC, Lam KS. Prevalence, treatment, and control of diagnosed diabetes in the US National Health and Nutrition Examination Survey 1999–2004. Ann Epidemiol 2008; 18:222–229.
  7. Koopman RJ, Mainous AG III, Diaz VA, Geesey ME. Changes in age at diagnosis of type 2 diabetes mellitus in the United States, 1988 to 2000. Ann Fam Med 2005; 3:60–63.
  8. Alexander GC, Sehgal NL, Moloney RM, Stafford RS. National trends in treatment of type 2 diabetes mellitus, 1994–2007. Arch Intern Med 2008; 168:2088–2094.
  9. The Action to Control Cardiovascular Risk in Diabetes Study Group. Effects of intensive glucose lowering in type 2 diabetes. N Engl J Med 2008; 358:2545–2559.
  10. The ADVANCE Collaborative Group. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N Engl J Med 2008; 358:2560–2572.
  11. Duckworth W, Abraira C, Moritz T, et al. Glucose control and vascular complications in veterans with type 2 diabetes. N Engl J Med 2009; 360:129–139.
  12. Jenssen TG, Tonstad S, Claudi T, Midthjell K, Cooper J. The gap between guidelines and practice in the treatment of type 2 diabetes: a nationwide survey in Norway. Diabetes Res Clin Pract 2008; 80:314–320.
  13. Gaede P, Lund-Andersen H, Parving HH, Pedersen O. Effect of a multifactorial intervention on mortality in type 2 diabetes. N Engl J Med 2008; 358:580–591.
  14. Wild S, Roglic G, Green A, Sicree R, King H. Global prevalence of diabetes: estimates for the year 2000 and projections for 2030. Diabetes Care 2004; 27:1047–1053.
  15. Rosenstock J. Management of type 2 diabetes mellitus in the elderly: special considerations. Drugs Aging 2001; 18:31–44.
  16. Brandle M, Zhou H, Smith BR, et al. The direct medical cost of type 2 diabetes. Diabetes Care 2003; 26:2300–2304.
  17. National Center for Health Statistics. Prevalence of overweight and obesity among adults: United States 2003–2004. Centers for Disease Contral and Prevention Web site. http://www.cdc.gov/nchs/products/pubs/pubd/hestats/overweight/overwght_adult_03.htm. Published: April 2006. Accessed September 23, 2009.
  18. Van Gaal LF, Gutkin SW, Nauck MA. Exploiting the antidiabetic properties of incretins to treat type 2 diabetes mellitus: glucagon-like peptide 1 receptor agonists or insulin for patients with inadequate glycemic control. Eur J Endocrinol 2008; 158:773–784.
  19. Anderson JW, Kendall CW, Jenkins DJ. Importance of weight management in type 2 diabetes: review with meta-analysis of clinical studies. J Am Coll Nutr 2003; 22:331–339.
  20. Fox CS, Pencina MJ, Wilson PW, Paynter NP, Vasan RS, D’Agostino RB Sr. Lifetime risk of cardiovascular disease among individuals with and without diabetes stratified by obesity status in the Framingham heart study. Diabetes Care 2008; 31:1582–1584.
  21. DeFronzo RA. From the triumvirate to the ominous octet: a new paradigm for the treatment of type 2 diabetes mellitus. Diabetes 2009; 58:773–795.
  22. Stonehouse A, Okerson T, Kendall D, Maggs D. Emerging incretin based therapies for type 2 diabetes: incretin mimetics and DPP-4 inhibitors. Curr Diabetes Rev 2008; 4:101–109.
  23. Cefalu WT. Pharmacotherapy for the treatment of patients with type 2 diabetes mellitus: rationale and specific agents. Clin Pharmacol Ther 2007; 81:636–649.
  24. Henry RR. Evolving concepts of type 2 diabetes management with oral medications: new approaches to an old disease. Curr Med Res Opin 2008; 24:2189–2202.
  25. American Diabetes Association. Nutrition recommendations and interventions for diabetes: a position statement of the American Diabetes Association. Diabetes Care 2008; 31(suppl 1):S61−S78.
  26. US Department of Health and Human Services (HHS) and US Department of Agriculture. Dietary guidelines for Americans, 2005. US Department of HHS Web site. http://www.health.gov/DietaryGuidelines/dga2005/document/default.htm. Published January 2005. Accessed September 25, 2009.
  27. National Heart, Lung, and Blood Institute. The practical guide: identification, evaluation, and treatment of overweight and obesity in adults. National Institutes of Health Web site. http://www.nhlbi.nih.gov/guidelines/obesity/prctgd_c.pdf. Updated: October 2000. Accessed September 28, 2009.
  28. Look AHEAD Research Group. Reduction in weight and cardio­vascular disease risk factors in individuals with type 2 diabetes: one-year results of the Look AHEAD trial. Diabetes Care 2007; 30:1374–1383.
  29. Williamson DA, Rejeski J, Lang W, Van Dorsten B, Fabricatore AN, Toledo K; for the Look AHEAD Research Group. Impact of a weight management program on health-related quality of life in overweight adults with type 2 diabetes. Arch Intern Med 2009; 169:163–171.
  30. Klein S, Sheard NF, Pi-Sunyer X, et al; for the American Diabetes Association; North American Association for the Study of Obesity; American Society for Clinical Nutrition. Weight management through lifestyle modification for the prevention and management of type 2 diabetes: rationale and strategies: a statement of the American Diabetes Association, the North American Association for the Study of Obesity, and the American Society for Clinical Nutrition. Diabetes Care 2004; 27:2067–2073.
  31. Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults. Executive summary of the Third Report of The National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III). JAMA 2001; 285:2486–2497.
  32. Buse JB, Ginsberg HN, Bakris GL, et al. Primary prevention of cardiovascular diseases in people with diabetes mellitus: a scientific statement from the American Heart Association and the American Diabetes Association. Circulation 2007; 115:114–126.
  33. Amori RE, Lau J, Pittas AG. Efficacy and safety of incretin therapy in type 2 diabetes: systematic review and meta-analysis. JAMA 2007; 298:194–206.
  34. Nielsen LL, Young AA, Parkes DG. Pharmacology of exenatide (synthetic exendin-4): a potential therapeutic for improved glycemic control of type 2 diabetes. Regul Pept 2004; 117:77–88.
  35. Young AA, Gedulin BR, Bhavsar S, et al. Glucose-lowering and insulin-sensitizing actions of exendin-4: studies in obese diabetic (ob/ob, db/db) mice, diabetic fatty Zucker rats, and diabetic rhesus monkeys (Macaca mulatta). Diabetes 1999; 48:1026–1034.
  36. Kim D, MacConell L, Zhuang D, et al. Effects of once-weekly dosing of a long-acting release formulation of exenatide on glucose control and body weight in subjects with type 2 diabetes. Diabetes Care 2007; 30:1487–1493.
  37. Buse JB, Henry RR, Han J, et al. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in sulfonylurea-treated patients with type 2 diabetes. Diabetes Care 2004; 27:2628–2635.
  38. DeFronzo RA, Ratner RE, Han J, Kim DD, Fineman MS, Baron AD. Effects of exenatide (exendin-4) on glycemic control and weight over 30 weeks in metformin-treated patients with type 2 diabetes. Diabetes Care 2005; 28:1092–1100.
  39. Kendall DM, Riddle MC, Rosenstock J, et al. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in patients with type 2 diabetes treated with metformin and a sulfonylurea. Diabetes Care 2005; 28:1083–1091.
  40. Zinman B, Hoogwerf BJ, Durán García S, et al. The effect of adding exenatide to a thiazolidinedione in suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med 2007; 146:477–485.
  41. Klonoff DC, Buse JB, Nielsen LL, et al. Exenatide effects on diabetes, obesity, cardiovascular risk factors and hepatic biomarkers in patients with type 2 diabetes treated for at least 3 years. Curr Med Res Opin 2008; 24:275–286.
  42. Heine RJ, Van Gaal LF, Johns D, Mihm MJ, Widel MH, Brodows RG; for the GWAA Study Group. Exenatide versus insulin glargine in patients with suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med 2005; 143:559–569.
  43. Barnett AH, Burger J, Johns D, et al. Tolerability and efficacy of exenatide and titrated insulin glargine in adult patients with type 2 diabetes previously uncontrolled with metformin or a sulfonylurea: a multinational, randomized, open-label, two-period, crossover noninferiority trial. Clin Ther 2007; 29:2333–2348.
  44. Nauck MA, Duran S, Kim D, et al. A comparison of twice-daily exenatide and biphasic insulin aspart in patients with type 2 diabetes who were suboptimally controlled with sulfonylurea and metformin: a non-inferiority study. Diabetologia 2007; 50:259–267.
  45. Glass LC, Qu Y, Lenox S, et al. Effects of exenatide versus insulin analogues on weight change in subjects with type 2 diabetes: a pooled post-hoc analysis. Curr Med Res Opin 2008; 24:639–644.
  46. Drucker DJ, Buse JB, Taylor K, et al; for the DURATION-1 Study Group. Exenatide once weekly versus twice daily for the treatment of type 2 diabetes: a randomised, open-label, non-inferiority study. Lancet 2008; 372:1240–1250.
  47. Garber A, Henry R, Ratner R, et al; for the LEAD-3 (Mono) Study Group. Liraglutide versus glimepiride monotherapy for type 2 diabetes (LEAD-3 Mono): a randomised, 52-week, phase III, double-blind, parallel-treatment trial. Lancet 2009; 373:473–481.
  48. Nauck M, Frid A, Hermansen K, et al; for the LEAD-2 Study Group. Efficacy and safety comparison of liraglutide, glimepiride, and placebo, all in combination with metformin, in type 2 diabetes: the LEAD (Liraglutide Effect and Action in Diabetes)-2 study. Diabetes Care 2009; 32:84–90.
  49. Ahrén B, Landin-Olsson M, Jansson PA, Svensson M, Holmes D, Schweizer A. Inhibition of dipeptidyl peptidase-4 reduces glycemia, sustains insulin levels, and reduces glucagon levels in type 2 diabetes. J Clin Endocrinol Metab 2004; 89:2078–2084.
  50. Herman GA, Stevens C, Van Dyck K, et al. Pharmacokinetics and pharmacodynamics of sitagliptin, an inhibitor of dipeptidyl peptidase IV, in healthy subjects: results from two randomized, double-blind, placebo-controlled studies with single oral doses. Clin Pharmacol Ther 2005; 78:675–688.
  51. Bohannon N. Overview of the gliptin class (dipeptidyl peptidase-4 inhibitors) in clinical practice. Postgrad Med 2009; 121:40–45.
  52. US Department of Health and Human Services. FDA approves new drug treatment for type 2 diabetes. US Food and Drug Administration Web site. http://www.fda.gov/newsevents/newsroom/pressannouncements/ucm174780.htm. Published July 31, 2009. Accessed September 18, 2009.
  53. Raz I, Hanefeld M, Xu L, Caria C, Williams-Herman D, Khatami H; for the Sitagliptin Study 023 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy in patients with type 2 diabetes mellitus. Diabetologia 2006; 49:2564–2571.
  54. Zerilli T, Pyon EY. Sitagliptin phosphate: a DPP-4 inhibitor for the treatment of type 2 diabetes mellitus. Clin Ther 2007; 29:2614–2634.
  55. Scott R, Loeys T, Davies MJ, Engel SS; for the Sitagliptin Study 801 Group. Efficacy and safety of sitagliptin when added to ongoing metformin therapy in patients with type 2 diabetes. Diabetes Obes Metab 2008; 10:959–969.
  56. Williams-Herman D, Johnson J, Teng R, et al. Efficacy and safety of initial combination therapy with sitagliptin and metformin in patients with type 2 diabetes: a 54-week study. Curr Med Res Opin 2009; 25:569–583.
  57. DeFronzo RA, Okerson T, Viswanathan P, Guan X, Holcombe JH, MacConell L. Effects of exenatide versus sitagliptin on postprandial glucose, insulin and glucagon secretion, gastric emptying, and caloric intake: a randomized, cross-over study. Curr Med Res Opin 2008; 24:2943–2952.
Article PDF
Author and Disclosure Information

William T. Cefalu, MD
Joint Program on Diabetes, Endocrinology and Metabolism, Louisiana State University Health Science Center and Pennington Biomedical Research Center, New Orleans and Baton Rouge, LA

Robert J. Richards, MD
Joint Program on Diabetes, Endocrinology and Metabolism, Louisiana State University Health Science Center and Pennington Biomedical Research Center, New Orleans and Baton Rouge, LA

Lydia Y. Melendez-Ramirez, MD
Joint Program on Diabetes, Endocrinology and Metabolism, Louisiana State University Health Science Center and Pennington Biomedical Research Center, New Orleans and Baton Rouge, LA

Correspondence: William T. Cefalu, MD, 6400 Perkins Road, Baton Rouge, LA 70808; [email protected]

Dr. Cefalu reported that he has received research and grant support from Amylin Pharmaceuticals, Inc., Eli Lilly and Company, Hollis-Eden Pharmaceuticals, Johnson & Johnson, and Merck & Co., Inc.; consulting/advisory fees from Amylin Pharmaceuticals, Inc., Eli Lilly and Company, Halozyme Therapeutics, Johnson & Johnson, and Merck & Co., Inc.; and honoraria from Amylin Pharmaceuticals, Inc., Eli Lilly and Company, and Merck & Co., Inc. Drs. Melendez-Ramirez and Richards reported that they have no financial interests or relationships that pose a potential conflict of interest with this article. Drs. Cefalu, Melendez-Ramirez, and Richards reported that they received no honoraria for writing this article.

Dr. Cefalu and his coauthors reported that they wrote this article and received no assistance with content development from unnamed contributors. They reported that BlueSpark Healthcare Communications, a medical communications company, assisted with preliminary literature searches, reference verification, proofing for grammar and style, and table and figure rendering based on author instructions.

Publications
Page Number
S39-S47
Author and Disclosure Information

William T. Cefalu, MD
Joint Program on Diabetes, Endocrinology and Metabolism, Louisiana State University Health Science Center and Pennington Biomedical Research Center, New Orleans and Baton Rouge, LA

Robert J. Richards, MD
Joint Program on Diabetes, Endocrinology and Metabolism, Louisiana State University Health Science Center and Pennington Biomedical Research Center, New Orleans and Baton Rouge, LA

Lydia Y. Melendez-Ramirez, MD
Joint Program on Diabetes, Endocrinology and Metabolism, Louisiana State University Health Science Center and Pennington Biomedical Research Center, New Orleans and Baton Rouge, LA

Correspondence: William T. Cefalu, MD, 6400 Perkins Road, Baton Rouge, LA 70808; [email protected]

Dr. Cefalu reported that he has received research and grant support from Amylin Pharmaceuticals, Inc., Eli Lilly and Company, Hollis-Eden Pharmaceuticals, Johnson & Johnson, and Merck & Co., Inc.; consulting/advisory fees from Amylin Pharmaceuticals, Inc., Eli Lilly and Company, Halozyme Therapeutics, Johnson & Johnson, and Merck & Co., Inc.; and honoraria from Amylin Pharmaceuticals, Inc., Eli Lilly and Company, and Merck & Co., Inc. Drs. Melendez-Ramirez and Richards reported that they have no financial interests or relationships that pose a potential conflict of interest with this article. Drs. Cefalu, Melendez-Ramirez, and Richards reported that they received no honoraria for writing this article.

Dr. Cefalu and his coauthors reported that they wrote this article and received no assistance with content development from unnamed contributors. They reported that BlueSpark Healthcare Communications, a medical communications company, assisted with preliminary literature searches, reference verification, proofing for grammar and style, and table and figure rendering based on author instructions.

Author and Disclosure Information

William T. Cefalu, MD
Joint Program on Diabetes, Endocrinology and Metabolism, Louisiana State University Health Science Center and Pennington Biomedical Research Center, New Orleans and Baton Rouge, LA

Robert J. Richards, MD
Joint Program on Diabetes, Endocrinology and Metabolism, Louisiana State University Health Science Center and Pennington Biomedical Research Center, New Orleans and Baton Rouge, LA

Lydia Y. Melendez-Ramirez, MD
Joint Program on Diabetes, Endocrinology and Metabolism, Louisiana State University Health Science Center and Pennington Biomedical Research Center, New Orleans and Baton Rouge, LA

Correspondence: William T. Cefalu, MD, 6400 Perkins Road, Baton Rouge, LA 70808; [email protected]

Dr. Cefalu reported that he has received research and grant support from Amylin Pharmaceuticals, Inc., Eli Lilly and Company, Hollis-Eden Pharmaceuticals, Johnson & Johnson, and Merck & Co., Inc.; consulting/advisory fees from Amylin Pharmaceuticals, Inc., Eli Lilly and Company, Halozyme Therapeutics, Johnson & Johnson, and Merck & Co., Inc.; and honoraria from Amylin Pharmaceuticals, Inc., Eli Lilly and Company, and Merck & Co., Inc. Drs. Melendez-Ramirez and Richards reported that they have no financial interests or relationships that pose a potential conflict of interest with this article. Drs. Cefalu, Melendez-Ramirez, and Richards reported that they received no honoraria for writing this article.

Dr. Cefalu and his coauthors reported that they wrote this article and received no assistance with content development from unnamed contributors. They reported that BlueSpark Healthcare Communications, a medical communications company, assisted with preliminary literature searches, reference verification, proofing for grammar and style, and table and figure rendering based on author instructions.

Article PDF
Article PDF
Related Articles

According to the American Association of Clinical Endocrinologists (AACE) and the American Diabetes Association (ADA), glycosylated hemoglobin (HbA1c) in patients with diabetes should be maintained at 6.5% or less (AACE) or at less than 7.0% (ADA). Both organizations support an aggressive stepwise approach that includes medication and lifestyle modification, with strategies and clinical attention devoted to avoiding significant hypoglycemia.1,2 Yet, despite the introduction of new antidiabetes agents, most current management strategies are offset by limitations in achieving and maintaining glycemic targets needed to provide optimal care for patients with diabetes, more than 90% of whom have type 2 diabetes mellitus (T2DM).3,4

Nationally, glycemic control among patients with T2DM has improved but is still far from optimal. According to data from the 1999–2000 National Health and Nutrition Examination Survey (NHANES), glycemic control (HbA1c < 7.0%) rates were 35.8% for patients with T2DM.5 In a more recent report (NHANES 1999–2004), fewer than half (48.4%) of adult patients with diagnosed diabetes achieved HbA1c levels below 7.0%.5,6 Factors contributing to these data include earlier onset and earlier detection of T2DM.7

CHANGING TREATMENT TRENDS

Available treatments for patients with T2DM include secretagogues, such as sulfonylureas and “glinides” (repaglinide and nateglinide), metformin, thiazolidinediones (TZDs), and dipeptidyl peptidase–4 (DPP-4) inhibitors among oral medications, and insulin and glucagon-like peptide–1 (GLP-1) receptor agonists among parenterally administered agents. According to the latest published data on prescribing patterns for patients with T2DM, analyses of the National Disease and Therapeutic Index (1994–2007) and the National Prescription Audit (2001–2007), sulfonylurea use decreased from 67% of treatment visits in 1994 to 34% of visits in 2007.8 By 2007, metformin, used in 54% of treatment visits, and TZDs, used in 28%, were the most frequently administered antidiabetes agents. Insulin use declined from 38% of visits during which a treatment was administered in 1994 to 25% of visits in 2000, but had increased subsequently to 28% of visits in 2007.

SIGNIFICANCE OF CARDIOVASCULAR RISK

Clinical research has suggested that focusing solely on improving glycemic control may be insufficient to reduce overall morbidity and mortality associated with diabetes. Specifically, data from recent studies, including the Action to Control Cardiovascular Risk in Diabetes (ACCORD), the Action in Diabetes and Vascular Disease: Preterax and Diamicron Modified Release Controlled Evaluation (ADVANCE), and the Veterans Affairs Diabetes Trial (VADT), emphasized that lowering HbA1c below 7% in a high-risk population of individuals with T2DM did not improve cardiovascular (CV) outcomes.9–11 The observations confirm that risk factors, including weight, blood pressure (BP), and lipid levels, are vitally important in reducing morbidity and mortality in this population. This perception is further underscored by the NHANES 1999–2004 data, which showed poor concurrent control of HbA1c, BP, and lipids; only 13.2% of patients with diagnosed diabetes achieved all three target goals simultaneously.6 Similarly, a nationwide survey in Norway showed that only 13% of patients with T2DM concurrently achieved goals for HbA1c, BP, and lipids.12

In the Danish Steno-2 Study, patients with T2DM and persistent microalbuminuria were treated with either intensive target-driven therapy using multiple drugs or conventional multifactorial treatment. Over a mean period of 13.3 years (7.8 years of treatment plus 5.5 years of follow-up), intensive multifactorial intervention to control multiple CV risk factors, including HbA1c, BP, and lipids, was associated with a lower risk of death from CV causes (hazard ratio [HR], 0.43; 95% confidence interval [CI], 0.19 to 0.94; P = .04) and a lower risk of CV events (HR, 0.41; 95% CI, 0.25 to 0.67; P < .001) than was conventional therapy.13

This article clarifies the redefinition of treatment success in patients with T2DM based on targeting the underlying physiologic defects of the disease.

T2DM, OVERWEIGHT/OBESITY, AND CV DISEASE: CLOSELY LINKED

The incidence and prevalence of T2DM, overweight/obesity, and CV disease (CVD) are increasing worldwide. It is estimated that the worldwide prevalence of diabetes will increase from 171 million in 2000 to 366 million by 203014; T2DM increases the risk of morbidity and mortality from microvascular (eg, neuropathic, retinopathic, nephropathic) and macrovascular (eg, coronary, peripheral vascular disease) complications.15 According to a Michigan health maintenance organization study (N = 1,364), the median annual direct cost of medical care for Caucasian patients with T2DM who were diet controlled, had a body mass index (BMI) of 30 kg/m2 or higher, and had no vascular complications was estimated to be $1,700 for men and $2,100 for women.16 The actual cost of care for patients with T2DM may be much higher, since most patients present with multiple CV risk factors in addition to being overweight.

NHANES data show that approximately two-thirds of Americans are either overweight or obese17; overweight/obesity affects about 80% of adults diagnosed with T2DM.18 Overweight or obesity can increase the risk for developing T2DM by more than 90-fold and, in women, it can increase the risk for developing coronary heart disease (CHD) by sixfold.19 The close link between T2DM and CVD is underscored further with recent data from the Framingham Heart Study, which showed a high lifetime risk of CVD in patients with diabetes, heightened further by obesity. During the 30-year study period, the lifetime risk of CVD in normal-weight people with diabetes was 78.6% in men and 54.8% in women; the risk increased to 86.9% in obese men with diabetes and to 78.8% in obese women with diabetes.20 The NHANES data also showed that the prevalence of T2DM increased in the past decade and that patients are being diagnosed at a younger age, from a mean age of 52 years in 1988–1994 to 46 years in 1999–2000.7

 

 

BRIDGING THE GAP FROM PATHOPHYSIOLOGY TO UNMET NEEDS

The paradigm behind the pathophysiology of T2DM has shifted from its perception as a simple “dual-defect” disease (ie, deficiency in insulin secretion and peripheral tissue insulin resistance) to a multidimensional disorder.1,21 This new model includes overweight/obesity, insulin resistance, qualitative and quantitative defects in insulin secretion, and dysregulation in the secretion of other hormones, including the beta-cell hormone amylin, the alpha-cell hormone glucagon, and the gastrointestinal incretin hormones GLP-1 and glucose-dependent insulinotropic polypeptide.21–23

The major target of antidiabetes agents is glycemic control, assessed by a reduction in HbA1c, but their effects on other metabolic factors and their adverse effects differ with each agent (Table 1).3 Whereas metformin and alpha-glucosidase inhibitors may help normalize glycemia with weight-neutral effects, many other agents, including insulin and its analogues, the “glinides,” first- and second-generation sulfonylureas, and TZDs, are associated with weight gain.23,24 In addition, the propensity to induce hypoglycemia differs among agents and clearly reflects the mechanism of action of each drug. The observed limitations of older therapies treating a progressive disease that is associated with a number of comorbid conditions supports the need for continued development of new antidiabetes agents.

CLINICAL GUIDELINES AND CV RISK FACTOR MANAGEMENT

The best strategy for managing T2DM is a comprehensive approach that addresses the fundamental core defects plus associated factors that contribute to increased CV risk. Several specialty groups have suggested guidelines and algorithms for the management of T2DM and its comorbidities. These guidelines, including the ADA standards of medical care, the AACE standards in tandem with the American College of Endocrinology guidelines, and the recent joint statement from the ADA and the European Association for the Study of Diabetes (EASD), acknowledge that the core defects of T2DM and the associated CV risk factors (eg, weight gain, obesity, hypertension, dyslipidemia) are important in developing optimal treatment strategies.1–3 Medical nutrition guidelines advocate weight loss as a key initial step in managing T2DM and the comorbidities that lead to elevated CV risk.25,26 The National Institutes of Health and the US Department of Health and Human Services/US Department of Agriculture advocate regular physical activity, dietary assessment, and periodic comorbidity and weight assessment for all people, not just those with T2DM or CVD.26,27

Weight reduction

Evidence in support of effective lifestyle intervention was demonstrated in the Action for Health in Diabetes (Look AHEAD) study. After 1 year, patients with T2DM treated with intensive lifestyle intervention lost an average of 8.6% of their initial weight compared with 0.7% in patients treated only with diabetes support and education (P < 0.001). The intensive-intervention patients also had a significant drop in HbA1c (from 7.3% to 6.6%; P < 0.001) and were able to reduce their antidiabetes, antihypertensive, and lipid-lowering medications.28 More recent data from the Look AHEAD study reported that overweight patients with T2DM enrolled in a weight management program experienced significant weight loss, improved physical fitness, reduced physical symptoms, and overall improvement in health-related quality of life.29 Thus, weight reduction appears to be a key component in reducing CV risk and improving quality of life in most patients with T2DM.28–30

Hypertension

Hypertension is a major risk factor for microvascular complications and CVD, and may be associated with, or be the underlying result of, nephropathy.2 BP control is clearly important in reducing the morbidity and mortality associated with T2DM. The recommended BP goal in patients with T2DM is less than 130/80 mm Hg.1,2

Hyperlipidemia

According to the Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III [ATP III]), diabetes is considered a CHD risk equivalent because it confers a high risk of new CHD developing within 10 years.31 In addition to the NCEP–ATP III guidelines, the ADA and the AACE have set target levels for lipids in patients with diabetes, including T2DM.1,2,31 All three organizations have defined 100 mg/dL as the target level for low-density lipoprotein.

HbA1c and lifestyle intervention

Figure 1. Suggested algorithm for the metabolic management of patients with type 2 diabetes mellitus. Clinicians should reinforce lifestyle interventions at every visit and check glycosylated hemoglobin (HbA1c) every 3 months until it is less than 7.0%, and then check it at least every 6 months. The interventions should be adjusted if HbA1c is 7.0% or greater.
The American Heart Association and the ADA initiated a call to action for global risk assessment for CVD and diabetes.32 According to their joint scientific statement, lifestyle intervention should be reinforced at every physician visit, and HbA1c should be monitored every 3 months until it is less than 7.0% and then rechecked every 6 months. Adjustments in intervention should be made if the HbA1c level is 7.0% or higher.3 A recent joint statement from the ADA and the EASD revised an earlier treatment algorithm for the initiation of therapy in patients with T2DM; the revision includes incretin therapies (ie, GLP-1 receptor agonists) as a tier 2 option, especially in patients in whom hypoglycemia and weight gain are concerns (Figure 1).3

 

 

EVOLUTION OF ANTIDIABETES THERAPIES

Traditional antidiabetes agents used in the treatment of patients with T2DM have focused mainly on insulin secretion and insulin resistance, with treatment success defined as achieving HbA1c goals with a reduced incidence of hypoglycemia.23 Secretagogues, such as sulfonylureas and glinides, stimulate the pancreas to release insulin. Insulin sensitizers, such as TZDs and metformin, enhance the action of insulin in muscle and fat1,3,23 and lower hepatic glucose production. The alpha-glucosidase inhibitors alter carbohydrate absorption from the gastrointestinal tract.1 The extent to which each agent achieves treatment success in terms of glucose lowering depends on several factors, including intrinsic attributes, duration of disease, and baseline glycemic control.3

Newer agents for the treatment of T2DM include the incretin-based therapies—GLP-1 receptor agonists and DPP-4 inhibitors—which influence mechanisms beyond increasing pancreatic insulin secretion and decreasing peripheral insulin resistance (Table 2).22 The GLP-1 signaling pathway has been leveraged by two distinct pharmacologic approaches. The first involves the use of synthetic peptides with glucoregulatory effects similar to those of endogenous GLP-1 (GLP-1 receptor agonists). The second involves the use of DPP-4 inhibitors, small molecules that inhibit the proteolytic activity of DPP-4, leading to enhanced endogenous GLP-1 concentrations.22

GLP-1 receptor agonists

Exenatide effects. Although many agents are in development, to date exenatide is the only GLP-1 receptor agonist approved by the US Food and Drug Administration (FDA).8,33 Exenatide is an exendin-4 GLP-1 receptor agonist with multiple glucoregulatory effects, including enhanced glucose-dependent insulin secretion, reduced glucagon secretion and food intake, and slowed gastric emptying.22,34 Exenatide is detectable in the circulation for up to 10 hours following subcutaneous (SC) administration22 and has a greater potency in reducing plasma glucose than GLP-1 in preclinical studies.35,36

By virtue of its beneficial effects on glycemic control, weight, BP, and lipids, exenatide addresses some of the components of the metabolic syndrome.37–41 In pivotal 30-week studies, exenatide was associated with HbA1c reductions that ranged from –0.40% to –0.86% from baseline and decreases in body weight of approximately –1 kg to –3 kg from baseline, without severe hypoglycemia.37–39 The percentage of patients who reached the ADA goal of HbA1c less than 7.0% at 30 weeks ranged from 24% to 34%. The addition of exenatide to TZD therapy in a 16-week study was associated with mean reductions in HbA1c of –0.98%, fasting plasma glucose (FPG) concentration of –1.69 mmol/L (–30.42 mg/dL), and body weight of –1.51 kg.40

A posthoc analysis of an open-label extension study involving patients who completed the original 30-week placebo-controlled studies showed that 46% of patients who remained on exenatide achieved the ADA goal of HbA1c less than 7.0% at 3 years.41 Exenatide administered for up to 3.5 years was associated with sustained reductions in HbA1c of –1.0% (P < .0001) and body weight of –5.3 kg (P < .001). Pancreatic beta-cell function, assessed by homeostasis model assessment, improved, as did BP, triglyceride, high-density lipoprotein, low-density lipoprotein, and aspartate aminotransferase levels.41

Comparison with insulin analogues. Comparative studies have highlighted the contrasting effects of exenatide and insulin analogues (eg, insulin glargine and fixed-ratio insulin).42–45 In a 26-week trial comparing exenatide with insulin glargine in subjects with T2DM, both agents resulted in similar decreases in HbA1c. Exenatide was also associated with a –2.3-kg weight reduction, whereas insulin glargine was associated with a +1.8-kg weight gain.42 Although rates of symptomatic hypoglycemia were similar, there were fewer cases of nocturnal hypoglycemia with exenatide (0.9 event/patient-year vs 2.4 events/patient-year with insulin).

In a 32-week study comparing exenatide BID with titrated insulin glargine QD, the HbA1c reductions for exenatide and insulin glargine were comparable. However, body weight decreased –4.2 kg over two 16-week treatment periods with exenatide, but increased +3.3 kg over the same periods with the basal insulin analogue.43 The incidence of hypoglycemia was lower with exenatide than with insulin glargine (14.7% vs 25.2%), although the difference was not statistically significant.

In another study that compared exenatide with biphasic insulin aspart, patients who were treated with exenatide also lost weight while those who received the fast-acting insulin analogue gained weight (between-group difference, –5.4 kg). Patients treated with exenatide also demonstrated greater reductions in postprandial plasma glucose (PPG) excursions following their morning (P < .001), midday (P = .002), and evening meals (P < .001).44 Overall, hypoglycemia rates were similar at study end between exenatide and insulin aspart (4.7 events/patient-year vs 5.6 events/patient-year). In all of these studies, significant gastrointestinal adverse events (nausea and vomiting) occurred more frequently with exenatide, and more patients withdrew from exenatide than from insulin.

Formulations in development. Other advances in GLP-1 receptor agonist therapy include novel formulations under clinical development, such as exenatide once weekly36,46 and liraglutide, a human analogue GLP-1 receptor agonist formulated for once-daily administration.47,48 In a 52-week study in patients with T2DM, liraglutide significantly reduced HbA1c; the 1.2-mg SC QD dosage reduced HBA1c by –0.84% (P = .0014) and the 1.8-mg SC QD dosage by –1.14% (P < .0001). In comparison, glimepiride 8 mg orally QD achieved a –0.51% reduction. Liraglutide was also associated with greater reductions in weight, hypoglycemia, and systolic BP than glimepiride.47

A 26-week study compared liraglutide (0.6, 1.2, and 1.8 mg SC QD), placebo, and glimepiride 4 mg QD in combination with metformin 1 g BID. HbA1c was reduced significantly in all liraglutide groups compared with placebo (P < .0001). Mean HbA1c decreased –1.0% with liraglutide 1.2 mg and 1.8 mg and with glimepiride; it decreased –0.7% with liraglutide 0.6 mg; and it increased +0.1% with placebo. Body weight decreased –1.8 kg to –2.8 kg in all liraglutide groups but increased +1.0 kg in the glimepiride group (P < .0001). The incidence of minor hypoglycemia with liraglutide (~3%) was comparable to that observed with placebo but less than that with glimepiride (17%; P < .001).48

A once-weekly long-acting release (LAR) formulation of exenatide submitted to the FDA for approval may provide enhanced glycemic and weight control, potentially improving patient acceptance and adherence.36,46 In a 15-week study, exenatide once weekly produced significant reductions in HbA1c, FPG, PPG, and body weight. There were no withdrawals due to adverse events, and the formation of anti-exenatide antibodies was not predictive of therapeutic end point response or adverse safety outcome. Instances of hypoglycemia were mild and not dose related.36 In a 30-week study comparing exenatide LAR once weekly with exenatide BID, patients given exenatide LAR once weekly had significantly greater HbA1c reductions than did patients given exenatide BID (–1.9% vs –1.5%; P = .0023). Treatment adherence was 98% with both exenatide regimens, and no episodes of major hypoglycemia occurred with either formulation regardless of background sulfonylurea use. Favorable effects on BP and lipid profile were observed with both exenatide regimens.46

 

 

DPP-4 inhibitors

The DPP-4 inhibitors (commonly called gliptins) inhibit the proteolytic cleavage of circulating GLP-1 by binding to the DPP-4 enzyme, increasing the concentration of endogenous GLP-1 approximately two- to threefold.49–51 These concentrations result in more prompt and appropriate secretion of insulin and suppression of glucagon in response to a carbohydrate-containing snack or meal, with the change in glucagon correlating linearly with improved glucose tolerance.51

DPP-4 inhibitors, which are given orally, include sitagliptin and saxagliptin (approved in the United States) and vildagliptin (not approved in the United States but used in the European Union and Latin America).8,22,33,52 Sitagliptin can be used either as monotherapy or in combination with metformin or a TZD.8,49–55 Recently, a single-tablet formulation of sitagliptin plus metformin was granted regulatory approval.8

When used alone or in combination with metformin or pioglitazone, sitagliptin has been associated with significant reductions in HbA1c (of ~0.5% to 0.6% when used alone, ~0.7% with metformin, and ~0.9% with pioglitazone [P < .001 vs placebo]), with hypoglycemia occurring in 1.3% or less of the population.54 In an 18-week study in which patients with T2DM who were inadequately controlled with metformin monotherapy were randomized to receive add-on sitagliptin (100 mg QD), rosiglitazone (8 mg QD), or placebo, sitagliptin reduced HbA1c –0.73% (P < .001 vs placebo) and reduced body weight –0.4 kg, while rosiglitazone reduced HbA1c –0.79% and increased body weight +1.5 kg.55

To evaluate the effectiveness of sitagliptin and metformin as initial therapy, a 54-week study was completed in 885 patients with T2DM and inadequate glycemic control (HbA1c 7.5–11%) on diet and exercise.56 Patients were evaluated on monotherapy with either sitagliptin (100 mg QD) or metformin (1 g or 2 g QD), or on initial therapy with the two in combination (sitagliptin 100 mg + metformin 1 mg or 2 mg QD). At week 54, in the all-patients-treated analysis, mean changes in HbA1c from baseline were –1.8% with sitagliptin plus metformin 2 g QD, –1.4% with sitagliptin plus metformin 1 g QD, –1.3% with metformin 2 g QD monotherapy, –1.0% with metformin 1 g QD monotherapy, and –0.8% with sitagliptin 100 mg QD monotherapy.

All treatments improved measures of beta-cell function (eg, homeostasis model assessment [HOMA]-beta, proinsulin/insulin ratio). Mean body weight decreased from baseline in the combination and metformin monotherapy groups and was unchanged from baseline in the sitagliptin monotherapy group. The incidence of hypoglycemia was low (1%–3%) across treatment groups. The incidence of gastrointestinal adverse experiences was evaluated with the coadministration of sitagliptin and metformin and appeared similar to that observed with use of metformin as monotherapy.56 Thus, this study suggested that an initial combination of a DPP-IV inhibitor with metformin can improve glycemic control and markers of beta-cell function in patients with T2DM.

Incretin-based therapies compared

Studies in both healthy individuals and in patients with T2DM have shown that oral DPP-4 inhibitors such as sitagliptin increase endogenous GLP-1 concentrations by about twofold compared with placebo.22,50 The pharma­cologic concentration of subcutaneously administered exenatide available for activating the GLP-1 receptor is significantly greater than the increased endogenous GLP-1 concentrations achieved with sitagliptin. In a recent clinical study comparing exenatide and sitagliptin in patients with T2DM, the mean 2-hour plasma concentration for exenatide was 64 pM compared with the mean 2-hour postprandial GLP-1 concentration of 15 pM for sitagliptin (baseline GLP-1 concentration was 7.2 pM).57 While both agents were shown to be effective, exenatide appeared to have had a greater effect than sitagliptin in increasing insulin secretion and reducing postprandial glucagon secretion, leading to significantly (P < 0.0001) greater reductions in PPG.57

Sitagliptin has been minimally associated with nausea, whereas patients who take exenatide need to be informed of the risk of usually mild to moderate, but sometimes severe, nausea and vomiting that tends to decrease over time.

For a detailed comparison of the effects of GLP-1 receptor agonists and DPP-4 inhibitors on HbA1c, weight, and hypoglycemia, see “Advances in therapy for type 2 diabetes: GLP–1 receptor agonists and DPP–4 inhibitors.”

CONCLUSION

Despite advances in diagnosis and treatment, T2DM, overweight/obesity, CVD, and their complications remain major public health burdens worldwide. The concepts that explain the pathophysiology of T2DM include the contribution of various factors beyond insulin secretion and insulin resistance, such as the role of incretin hormones in disease progression. A comprehensive approach to managing patients with T2DM requires targeting the fundamental defects of the disease and its comorbidities. Newer agents, including incretin-based therapies such as GLP-1 receptor agonists and DPP-4 inhibitors, address the fundamental defects of T2DM. The definition of treatment success in the management of T2DM will be redefined as more data become available on agents that exert beneficial effects not only on glycemia but on parameters that may influence overall CV health, such as weight, BP, and lipid profiles.

According to the American Association of Clinical Endocrinologists (AACE) and the American Diabetes Association (ADA), glycosylated hemoglobin (HbA1c) in patients with diabetes should be maintained at 6.5% or less (AACE) or at less than 7.0% (ADA). Both organizations support an aggressive stepwise approach that includes medication and lifestyle modification, with strategies and clinical attention devoted to avoiding significant hypoglycemia.1,2 Yet, despite the introduction of new antidiabetes agents, most current management strategies are offset by limitations in achieving and maintaining glycemic targets needed to provide optimal care for patients with diabetes, more than 90% of whom have type 2 diabetes mellitus (T2DM).3,4

Nationally, glycemic control among patients with T2DM has improved but is still far from optimal. According to data from the 1999–2000 National Health and Nutrition Examination Survey (NHANES), glycemic control (HbA1c < 7.0%) rates were 35.8% for patients with T2DM.5 In a more recent report (NHANES 1999–2004), fewer than half (48.4%) of adult patients with diagnosed diabetes achieved HbA1c levels below 7.0%.5,6 Factors contributing to these data include earlier onset and earlier detection of T2DM.7

CHANGING TREATMENT TRENDS

Available treatments for patients with T2DM include secretagogues, such as sulfonylureas and “glinides” (repaglinide and nateglinide), metformin, thiazolidinediones (TZDs), and dipeptidyl peptidase–4 (DPP-4) inhibitors among oral medications, and insulin and glucagon-like peptide–1 (GLP-1) receptor agonists among parenterally administered agents. According to the latest published data on prescribing patterns for patients with T2DM, analyses of the National Disease and Therapeutic Index (1994–2007) and the National Prescription Audit (2001–2007), sulfonylurea use decreased from 67% of treatment visits in 1994 to 34% of visits in 2007.8 By 2007, metformin, used in 54% of treatment visits, and TZDs, used in 28%, were the most frequently administered antidiabetes agents. Insulin use declined from 38% of visits during which a treatment was administered in 1994 to 25% of visits in 2000, but had increased subsequently to 28% of visits in 2007.

SIGNIFICANCE OF CARDIOVASCULAR RISK

Clinical research has suggested that focusing solely on improving glycemic control may be insufficient to reduce overall morbidity and mortality associated with diabetes. Specifically, data from recent studies, including the Action to Control Cardiovascular Risk in Diabetes (ACCORD), the Action in Diabetes and Vascular Disease: Preterax and Diamicron Modified Release Controlled Evaluation (ADVANCE), and the Veterans Affairs Diabetes Trial (VADT), emphasized that lowering HbA1c below 7% in a high-risk population of individuals with T2DM did not improve cardiovascular (CV) outcomes.9–11 The observations confirm that risk factors, including weight, blood pressure (BP), and lipid levels, are vitally important in reducing morbidity and mortality in this population. This perception is further underscored by the NHANES 1999–2004 data, which showed poor concurrent control of HbA1c, BP, and lipids; only 13.2% of patients with diagnosed diabetes achieved all three target goals simultaneously.6 Similarly, a nationwide survey in Norway showed that only 13% of patients with T2DM concurrently achieved goals for HbA1c, BP, and lipids.12

In the Danish Steno-2 Study, patients with T2DM and persistent microalbuminuria were treated with either intensive target-driven therapy using multiple drugs or conventional multifactorial treatment. Over a mean period of 13.3 years (7.8 years of treatment plus 5.5 years of follow-up), intensive multifactorial intervention to control multiple CV risk factors, including HbA1c, BP, and lipids, was associated with a lower risk of death from CV causes (hazard ratio [HR], 0.43; 95% confidence interval [CI], 0.19 to 0.94; P = .04) and a lower risk of CV events (HR, 0.41; 95% CI, 0.25 to 0.67; P < .001) than was conventional therapy.13

This article clarifies the redefinition of treatment success in patients with T2DM based on targeting the underlying physiologic defects of the disease.

T2DM, OVERWEIGHT/OBESITY, AND CV DISEASE: CLOSELY LINKED

The incidence and prevalence of T2DM, overweight/obesity, and CV disease (CVD) are increasing worldwide. It is estimated that the worldwide prevalence of diabetes will increase from 171 million in 2000 to 366 million by 203014; T2DM increases the risk of morbidity and mortality from microvascular (eg, neuropathic, retinopathic, nephropathic) and macrovascular (eg, coronary, peripheral vascular disease) complications.15 According to a Michigan health maintenance organization study (N = 1,364), the median annual direct cost of medical care for Caucasian patients with T2DM who were diet controlled, had a body mass index (BMI) of 30 kg/m2 or higher, and had no vascular complications was estimated to be $1,700 for men and $2,100 for women.16 The actual cost of care for patients with T2DM may be much higher, since most patients present with multiple CV risk factors in addition to being overweight.

NHANES data show that approximately two-thirds of Americans are either overweight or obese17; overweight/obesity affects about 80% of adults diagnosed with T2DM.18 Overweight or obesity can increase the risk for developing T2DM by more than 90-fold and, in women, it can increase the risk for developing coronary heart disease (CHD) by sixfold.19 The close link between T2DM and CVD is underscored further with recent data from the Framingham Heart Study, which showed a high lifetime risk of CVD in patients with diabetes, heightened further by obesity. During the 30-year study period, the lifetime risk of CVD in normal-weight people with diabetes was 78.6% in men and 54.8% in women; the risk increased to 86.9% in obese men with diabetes and to 78.8% in obese women with diabetes.20 The NHANES data also showed that the prevalence of T2DM increased in the past decade and that patients are being diagnosed at a younger age, from a mean age of 52 years in 1988–1994 to 46 years in 1999–2000.7

 

 

BRIDGING THE GAP FROM PATHOPHYSIOLOGY TO UNMET NEEDS

The paradigm behind the pathophysiology of T2DM has shifted from its perception as a simple “dual-defect” disease (ie, deficiency in insulin secretion and peripheral tissue insulin resistance) to a multidimensional disorder.1,21 This new model includes overweight/obesity, insulin resistance, qualitative and quantitative defects in insulin secretion, and dysregulation in the secretion of other hormones, including the beta-cell hormone amylin, the alpha-cell hormone glucagon, and the gastrointestinal incretin hormones GLP-1 and glucose-dependent insulinotropic polypeptide.21–23

The major target of antidiabetes agents is glycemic control, assessed by a reduction in HbA1c, but their effects on other metabolic factors and their adverse effects differ with each agent (Table 1).3 Whereas metformin and alpha-glucosidase inhibitors may help normalize glycemia with weight-neutral effects, many other agents, including insulin and its analogues, the “glinides,” first- and second-generation sulfonylureas, and TZDs, are associated with weight gain.23,24 In addition, the propensity to induce hypoglycemia differs among agents and clearly reflects the mechanism of action of each drug. The observed limitations of older therapies treating a progressive disease that is associated with a number of comorbid conditions supports the need for continued development of new antidiabetes agents.

CLINICAL GUIDELINES AND CV RISK FACTOR MANAGEMENT

The best strategy for managing T2DM is a comprehensive approach that addresses the fundamental core defects plus associated factors that contribute to increased CV risk. Several specialty groups have suggested guidelines and algorithms for the management of T2DM and its comorbidities. These guidelines, including the ADA standards of medical care, the AACE standards in tandem with the American College of Endocrinology guidelines, and the recent joint statement from the ADA and the European Association for the Study of Diabetes (EASD), acknowledge that the core defects of T2DM and the associated CV risk factors (eg, weight gain, obesity, hypertension, dyslipidemia) are important in developing optimal treatment strategies.1–3 Medical nutrition guidelines advocate weight loss as a key initial step in managing T2DM and the comorbidities that lead to elevated CV risk.25,26 The National Institutes of Health and the US Department of Health and Human Services/US Department of Agriculture advocate regular physical activity, dietary assessment, and periodic comorbidity and weight assessment for all people, not just those with T2DM or CVD.26,27

Weight reduction

Evidence in support of effective lifestyle intervention was demonstrated in the Action for Health in Diabetes (Look AHEAD) study. After 1 year, patients with T2DM treated with intensive lifestyle intervention lost an average of 8.6% of their initial weight compared with 0.7% in patients treated only with diabetes support and education (P < 0.001). The intensive-intervention patients also had a significant drop in HbA1c (from 7.3% to 6.6%; P < 0.001) and were able to reduce their antidiabetes, antihypertensive, and lipid-lowering medications.28 More recent data from the Look AHEAD study reported that overweight patients with T2DM enrolled in a weight management program experienced significant weight loss, improved physical fitness, reduced physical symptoms, and overall improvement in health-related quality of life.29 Thus, weight reduction appears to be a key component in reducing CV risk and improving quality of life in most patients with T2DM.28–30

Hypertension

Hypertension is a major risk factor for microvascular complications and CVD, and may be associated with, or be the underlying result of, nephropathy.2 BP control is clearly important in reducing the morbidity and mortality associated with T2DM. The recommended BP goal in patients with T2DM is less than 130/80 mm Hg.1,2

Hyperlipidemia

According to the Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III [ATP III]), diabetes is considered a CHD risk equivalent because it confers a high risk of new CHD developing within 10 years.31 In addition to the NCEP–ATP III guidelines, the ADA and the AACE have set target levels for lipids in patients with diabetes, including T2DM.1,2,31 All three organizations have defined 100 mg/dL as the target level for low-density lipoprotein.

HbA1c and lifestyle intervention

Figure 1. Suggested algorithm for the metabolic management of patients with type 2 diabetes mellitus. Clinicians should reinforce lifestyle interventions at every visit and check glycosylated hemoglobin (HbA1c) every 3 months until it is less than 7.0%, and then check it at least every 6 months. The interventions should be adjusted if HbA1c is 7.0% or greater.
The American Heart Association and the ADA initiated a call to action for global risk assessment for CVD and diabetes.32 According to their joint scientific statement, lifestyle intervention should be reinforced at every physician visit, and HbA1c should be monitored every 3 months until it is less than 7.0% and then rechecked every 6 months. Adjustments in intervention should be made if the HbA1c level is 7.0% or higher.3 A recent joint statement from the ADA and the EASD revised an earlier treatment algorithm for the initiation of therapy in patients with T2DM; the revision includes incretin therapies (ie, GLP-1 receptor agonists) as a tier 2 option, especially in patients in whom hypoglycemia and weight gain are concerns (Figure 1).3

 

 

EVOLUTION OF ANTIDIABETES THERAPIES

Traditional antidiabetes agents used in the treatment of patients with T2DM have focused mainly on insulin secretion and insulin resistance, with treatment success defined as achieving HbA1c goals with a reduced incidence of hypoglycemia.23 Secretagogues, such as sulfonylureas and glinides, stimulate the pancreas to release insulin. Insulin sensitizers, such as TZDs and metformin, enhance the action of insulin in muscle and fat1,3,23 and lower hepatic glucose production. The alpha-glucosidase inhibitors alter carbohydrate absorption from the gastrointestinal tract.1 The extent to which each agent achieves treatment success in terms of glucose lowering depends on several factors, including intrinsic attributes, duration of disease, and baseline glycemic control.3

Newer agents for the treatment of T2DM include the incretin-based therapies—GLP-1 receptor agonists and DPP-4 inhibitors—which influence mechanisms beyond increasing pancreatic insulin secretion and decreasing peripheral insulin resistance (Table 2).22 The GLP-1 signaling pathway has been leveraged by two distinct pharmacologic approaches. The first involves the use of synthetic peptides with glucoregulatory effects similar to those of endogenous GLP-1 (GLP-1 receptor agonists). The second involves the use of DPP-4 inhibitors, small molecules that inhibit the proteolytic activity of DPP-4, leading to enhanced endogenous GLP-1 concentrations.22

GLP-1 receptor agonists

Exenatide effects. Although many agents are in development, to date exenatide is the only GLP-1 receptor agonist approved by the US Food and Drug Administration (FDA).8,33 Exenatide is an exendin-4 GLP-1 receptor agonist with multiple glucoregulatory effects, including enhanced glucose-dependent insulin secretion, reduced glucagon secretion and food intake, and slowed gastric emptying.22,34 Exenatide is detectable in the circulation for up to 10 hours following subcutaneous (SC) administration22 and has a greater potency in reducing plasma glucose than GLP-1 in preclinical studies.35,36

By virtue of its beneficial effects on glycemic control, weight, BP, and lipids, exenatide addresses some of the components of the metabolic syndrome.37–41 In pivotal 30-week studies, exenatide was associated with HbA1c reductions that ranged from –0.40% to –0.86% from baseline and decreases in body weight of approximately –1 kg to –3 kg from baseline, without severe hypoglycemia.37–39 The percentage of patients who reached the ADA goal of HbA1c less than 7.0% at 30 weeks ranged from 24% to 34%. The addition of exenatide to TZD therapy in a 16-week study was associated with mean reductions in HbA1c of –0.98%, fasting plasma glucose (FPG) concentration of –1.69 mmol/L (–30.42 mg/dL), and body weight of –1.51 kg.40

A posthoc analysis of an open-label extension study involving patients who completed the original 30-week placebo-controlled studies showed that 46% of patients who remained on exenatide achieved the ADA goal of HbA1c less than 7.0% at 3 years.41 Exenatide administered for up to 3.5 years was associated with sustained reductions in HbA1c of –1.0% (P < .0001) and body weight of –5.3 kg (P < .001). Pancreatic beta-cell function, assessed by homeostasis model assessment, improved, as did BP, triglyceride, high-density lipoprotein, low-density lipoprotein, and aspartate aminotransferase levels.41

Comparison with insulin analogues. Comparative studies have highlighted the contrasting effects of exenatide and insulin analogues (eg, insulin glargine and fixed-ratio insulin).42–45 In a 26-week trial comparing exenatide with insulin glargine in subjects with T2DM, both agents resulted in similar decreases in HbA1c. Exenatide was also associated with a –2.3-kg weight reduction, whereas insulin glargine was associated with a +1.8-kg weight gain.42 Although rates of symptomatic hypoglycemia were similar, there were fewer cases of nocturnal hypoglycemia with exenatide (0.9 event/patient-year vs 2.4 events/patient-year with insulin).

In a 32-week study comparing exenatide BID with titrated insulin glargine QD, the HbA1c reductions for exenatide and insulin glargine were comparable. However, body weight decreased –4.2 kg over two 16-week treatment periods with exenatide, but increased +3.3 kg over the same periods with the basal insulin analogue.43 The incidence of hypoglycemia was lower with exenatide than with insulin glargine (14.7% vs 25.2%), although the difference was not statistically significant.

In another study that compared exenatide with biphasic insulin aspart, patients who were treated with exenatide also lost weight while those who received the fast-acting insulin analogue gained weight (between-group difference, –5.4 kg). Patients treated with exenatide also demonstrated greater reductions in postprandial plasma glucose (PPG) excursions following their morning (P < .001), midday (P = .002), and evening meals (P < .001).44 Overall, hypoglycemia rates were similar at study end between exenatide and insulin aspart (4.7 events/patient-year vs 5.6 events/patient-year). In all of these studies, significant gastrointestinal adverse events (nausea and vomiting) occurred more frequently with exenatide, and more patients withdrew from exenatide than from insulin.

Formulations in development. Other advances in GLP-1 receptor agonist therapy include novel formulations under clinical development, such as exenatide once weekly36,46 and liraglutide, a human analogue GLP-1 receptor agonist formulated for once-daily administration.47,48 In a 52-week study in patients with T2DM, liraglutide significantly reduced HbA1c; the 1.2-mg SC QD dosage reduced HBA1c by –0.84% (P = .0014) and the 1.8-mg SC QD dosage by –1.14% (P < .0001). In comparison, glimepiride 8 mg orally QD achieved a –0.51% reduction. Liraglutide was also associated with greater reductions in weight, hypoglycemia, and systolic BP than glimepiride.47

A 26-week study compared liraglutide (0.6, 1.2, and 1.8 mg SC QD), placebo, and glimepiride 4 mg QD in combination with metformin 1 g BID. HbA1c was reduced significantly in all liraglutide groups compared with placebo (P < .0001). Mean HbA1c decreased –1.0% with liraglutide 1.2 mg and 1.8 mg and with glimepiride; it decreased –0.7% with liraglutide 0.6 mg; and it increased +0.1% with placebo. Body weight decreased –1.8 kg to –2.8 kg in all liraglutide groups but increased +1.0 kg in the glimepiride group (P < .0001). The incidence of minor hypoglycemia with liraglutide (~3%) was comparable to that observed with placebo but less than that with glimepiride (17%; P < .001).48

A once-weekly long-acting release (LAR) formulation of exenatide submitted to the FDA for approval may provide enhanced glycemic and weight control, potentially improving patient acceptance and adherence.36,46 In a 15-week study, exenatide once weekly produced significant reductions in HbA1c, FPG, PPG, and body weight. There were no withdrawals due to adverse events, and the formation of anti-exenatide antibodies was not predictive of therapeutic end point response or adverse safety outcome. Instances of hypoglycemia were mild and not dose related.36 In a 30-week study comparing exenatide LAR once weekly with exenatide BID, patients given exenatide LAR once weekly had significantly greater HbA1c reductions than did patients given exenatide BID (–1.9% vs –1.5%; P = .0023). Treatment adherence was 98% with both exenatide regimens, and no episodes of major hypoglycemia occurred with either formulation regardless of background sulfonylurea use. Favorable effects on BP and lipid profile were observed with both exenatide regimens.46

 

 

DPP-4 inhibitors

The DPP-4 inhibitors (commonly called gliptins) inhibit the proteolytic cleavage of circulating GLP-1 by binding to the DPP-4 enzyme, increasing the concentration of endogenous GLP-1 approximately two- to threefold.49–51 These concentrations result in more prompt and appropriate secretion of insulin and suppression of glucagon in response to a carbohydrate-containing snack or meal, with the change in glucagon correlating linearly with improved glucose tolerance.51

DPP-4 inhibitors, which are given orally, include sitagliptin and saxagliptin (approved in the United States) and vildagliptin (not approved in the United States but used in the European Union and Latin America).8,22,33,52 Sitagliptin can be used either as monotherapy or in combination with metformin or a TZD.8,49–55 Recently, a single-tablet formulation of sitagliptin plus metformin was granted regulatory approval.8

When used alone or in combination with metformin or pioglitazone, sitagliptin has been associated with significant reductions in HbA1c (of ~0.5% to 0.6% when used alone, ~0.7% with metformin, and ~0.9% with pioglitazone [P < .001 vs placebo]), with hypoglycemia occurring in 1.3% or less of the population.54 In an 18-week study in which patients with T2DM who were inadequately controlled with metformin monotherapy were randomized to receive add-on sitagliptin (100 mg QD), rosiglitazone (8 mg QD), or placebo, sitagliptin reduced HbA1c –0.73% (P < .001 vs placebo) and reduced body weight –0.4 kg, while rosiglitazone reduced HbA1c –0.79% and increased body weight +1.5 kg.55

To evaluate the effectiveness of sitagliptin and metformin as initial therapy, a 54-week study was completed in 885 patients with T2DM and inadequate glycemic control (HbA1c 7.5–11%) on diet and exercise.56 Patients were evaluated on monotherapy with either sitagliptin (100 mg QD) or metformin (1 g or 2 g QD), or on initial therapy with the two in combination (sitagliptin 100 mg + metformin 1 mg or 2 mg QD). At week 54, in the all-patients-treated analysis, mean changes in HbA1c from baseline were –1.8% with sitagliptin plus metformin 2 g QD, –1.4% with sitagliptin plus metformin 1 g QD, –1.3% with metformin 2 g QD monotherapy, –1.0% with metformin 1 g QD monotherapy, and –0.8% with sitagliptin 100 mg QD monotherapy.

All treatments improved measures of beta-cell function (eg, homeostasis model assessment [HOMA]-beta, proinsulin/insulin ratio). Mean body weight decreased from baseline in the combination and metformin monotherapy groups and was unchanged from baseline in the sitagliptin monotherapy group. The incidence of hypoglycemia was low (1%–3%) across treatment groups. The incidence of gastrointestinal adverse experiences was evaluated with the coadministration of sitagliptin and metformin and appeared similar to that observed with use of metformin as monotherapy.56 Thus, this study suggested that an initial combination of a DPP-IV inhibitor with metformin can improve glycemic control and markers of beta-cell function in patients with T2DM.

Incretin-based therapies compared

Studies in both healthy individuals and in patients with T2DM have shown that oral DPP-4 inhibitors such as sitagliptin increase endogenous GLP-1 concentrations by about twofold compared with placebo.22,50 The pharma­cologic concentration of subcutaneously administered exenatide available for activating the GLP-1 receptor is significantly greater than the increased endogenous GLP-1 concentrations achieved with sitagliptin. In a recent clinical study comparing exenatide and sitagliptin in patients with T2DM, the mean 2-hour plasma concentration for exenatide was 64 pM compared with the mean 2-hour postprandial GLP-1 concentration of 15 pM for sitagliptin (baseline GLP-1 concentration was 7.2 pM).57 While both agents were shown to be effective, exenatide appeared to have had a greater effect than sitagliptin in increasing insulin secretion and reducing postprandial glucagon secretion, leading to significantly (P < 0.0001) greater reductions in PPG.57

Sitagliptin has been minimally associated with nausea, whereas patients who take exenatide need to be informed of the risk of usually mild to moderate, but sometimes severe, nausea and vomiting that tends to decrease over time.

For a detailed comparison of the effects of GLP-1 receptor agonists and DPP-4 inhibitors on HbA1c, weight, and hypoglycemia, see “Advances in therapy for type 2 diabetes: GLP–1 receptor agonists and DPP–4 inhibitors.”

CONCLUSION

Despite advances in diagnosis and treatment, T2DM, overweight/obesity, CVD, and their complications remain major public health burdens worldwide. The concepts that explain the pathophysiology of T2DM include the contribution of various factors beyond insulin secretion and insulin resistance, such as the role of incretin hormones in disease progression. A comprehensive approach to managing patients with T2DM requires targeting the fundamental defects of the disease and its comorbidities. Newer agents, including incretin-based therapies such as GLP-1 receptor agonists and DPP-4 inhibitors, address the fundamental defects of T2DM. The definition of treatment success in the management of T2DM will be redefined as more data become available on agents that exert beneficial effects not only on glycemia but on parameters that may influence overall CV health, such as weight, BP, and lipid profiles.

References
  1. AACE Diabetes Mellitus Clinical Practice Guidelines Task Force. American Association of Clinical Endocrinologists medical guidelines for clinical practice for the management of diabetes mellitus. Endocr Pract 2007; 13(suppl 1):S4–S68.
  2. American Diabetes Association. Standards of medical care in diabetes—2009. Diabetes Care 2009; 32(suppl 1):S13–S61.
  3. Nathan DM, Buse JB, Davidson MB, et al. Medical management of hyperglycemia in type 2 diabetes: a consensus algorithm for the initiation and adjustment of therapy: a consensus statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care 2009; 32:193–203.
  4. National Institute of Diabetes and Digestive and Kidney Diseases. National Diabetes Statistics, 2007 fact sheet. National Institutes of Health Web site. http://www.diabetes.niddk.nih.gov/dm/pubs/statistics/index.htm. Published 2008. Accessed September 16, 2009.
  5. Koro CE, Bowlin SJ, Bourgeois N, Fedder DO. Glycemic control from 1988 to 2000 among US adults diagnosed with type 2 diabetes: a preliminary report. Diabetes Care 2004; 27:17–20.
  6. Ong KL, Cheung BM, Wong LY, Wat NM, Tan KC, Lam KS. Prevalence, treatment, and control of diagnosed diabetes in the US National Health and Nutrition Examination Survey 1999–2004. Ann Epidemiol 2008; 18:222–229.
  7. Koopman RJ, Mainous AG III, Diaz VA, Geesey ME. Changes in age at diagnosis of type 2 diabetes mellitus in the United States, 1988 to 2000. Ann Fam Med 2005; 3:60–63.
  8. Alexander GC, Sehgal NL, Moloney RM, Stafford RS. National trends in treatment of type 2 diabetes mellitus, 1994–2007. Arch Intern Med 2008; 168:2088–2094.
  9. The Action to Control Cardiovascular Risk in Diabetes Study Group. Effects of intensive glucose lowering in type 2 diabetes. N Engl J Med 2008; 358:2545–2559.
  10. The ADVANCE Collaborative Group. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N Engl J Med 2008; 358:2560–2572.
  11. Duckworth W, Abraira C, Moritz T, et al. Glucose control and vascular complications in veterans with type 2 diabetes. N Engl J Med 2009; 360:129–139.
  12. Jenssen TG, Tonstad S, Claudi T, Midthjell K, Cooper J. The gap between guidelines and practice in the treatment of type 2 diabetes: a nationwide survey in Norway. Diabetes Res Clin Pract 2008; 80:314–320.
  13. Gaede P, Lund-Andersen H, Parving HH, Pedersen O. Effect of a multifactorial intervention on mortality in type 2 diabetes. N Engl J Med 2008; 358:580–591.
  14. Wild S, Roglic G, Green A, Sicree R, King H. Global prevalence of diabetes: estimates for the year 2000 and projections for 2030. Diabetes Care 2004; 27:1047–1053.
  15. Rosenstock J. Management of type 2 diabetes mellitus in the elderly: special considerations. Drugs Aging 2001; 18:31–44.
  16. Brandle M, Zhou H, Smith BR, et al. The direct medical cost of type 2 diabetes. Diabetes Care 2003; 26:2300–2304.
  17. National Center for Health Statistics. Prevalence of overweight and obesity among adults: United States 2003–2004. Centers for Disease Contral and Prevention Web site. http://www.cdc.gov/nchs/products/pubs/pubd/hestats/overweight/overwght_adult_03.htm. Published: April 2006. Accessed September 23, 2009.
  18. Van Gaal LF, Gutkin SW, Nauck MA. Exploiting the antidiabetic properties of incretins to treat type 2 diabetes mellitus: glucagon-like peptide 1 receptor agonists or insulin for patients with inadequate glycemic control. Eur J Endocrinol 2008; 158:773–784.
  19. Anderson JW, Kendall CW, Jenkins DJ. Importance of weight management in type 2 diabetes: review with meta-analysis of clinical studies. J Am Coll Nutr 2003; 22:331–339.
  20. Fox CS, Pencina MJ, Wilson PW, Paynter NP, Vasan RS, D’Agostino RB Sr. Lifetime risk of cardiovascular disease among individuals with and without diabetes stratified by obesity status in the Framingham heart study. Diabetes Care 2008; 31:1582–1584.
  21. DeFronzo RA. From the triumvirate to the ominous octet: a new paradigm for the treatment of type 2 diabetes mellitus. Diabetes 2009; 58:773–795.
  22. Stonehouse A, Okerson T, Kendall D, Maggs D. Emerging incretin based therapies for type 2 diabetes: incretin mimetics and DPP-4 inhibitors. Curr Diabetes Rev 2008; 4:101–109.
  23. Cefalu WT. Pharmacotherapy for the treatment of patients with type 2 diabetes mellitus: rationale and specific agents. Clin Pharmacol Ther 2007; 81:636–649.
  24. Henry RR. Evolving concepts of type 2 diabetes management with oral medications: new approaches to an old disease. Curr Med Res Opin 2008; 24:2189–2202.
  25. American Diabetes Association. Nutrition recommendations and interventions for diabetes: a position statement of the American Diabetes Association. Diabetes Care 2008; 31(suppl 1):S61−S78.
  26. US Department of Health and Human Services (HHS) and US Department of Agriculture. Dietary guidelines for Americans, 2005. US Department of HHS Web site. http://www.health.gov/DietaryGuidelines/dga2005/document/default.htm. Published January 2005. Accessed September 25, 2009.
  27. National Heart, Lung, and Blood Institute. The practical guide: identification, evaluation, and treatment of overweight and obesity in adults. National Institutes of Health Web site. http://www.nhlbi.nih.gov/guidelines/obesity/prctgd_c.pdf. Updated: October 2000. Accessed September 28, 2009.
  28. Look AHEAD Research Group. Reduction in weight and cardio­vascular disease risk factors in individuals with type 2 diabetes: one-year results of the Look AHEAD trial. Diabetes Care 2007; 30:1374–1383.
  29. Williamson DA, Rejeski J, Lang W, Van Dorsten B, Fabricatore AN, Toledo K; for the Look AHEAD Research Group. Impact of a weight management program on health-related quality of life in overweight adults with type 2 diabetes. Arch Intern Med 2009; 169:163–171.
  30. Klein S, Sheard NF, Pi-Sunyer X, et al; for the American Diabetes Association; North American Association for the Study of Obesity; American Society for Clinical Nutrition. Weight management through lifestyle modification for the prevention and management of type 2 diabetes: rationale and strategies: a statement of the American Diabetes Association, the North American Association for the Study of Obesity, and the American Society for Clinical Nutrition. Diabetes Care 2004; 27:2067–2073.
  31. Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults. Executive summary of the Third Report of The National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III). JAMA 2001; 285:2486–2497.
  32. Buse JB, Ginsberg HN, Bakris GL, et al. Primary prevention of cardiovascular diseases in people with diabetes mellitus: a scientific statement from the American Heart Association and the American Diabetes Association. Circulation 2007; 115:114–126.
  33. Amori RE, Lau J, Pittas AG. Efficacy and safety of incretin therapy in type 2 diabetes: systematic review and meta-analysis. JAMA 2007; 298:194–206.
  34. Nielsen LL, Young AA, Parkes DG. Pharmacology of exenatide (synthetic exendin-4): a potential therapeutic for improved glycemic control of type 2 diabetes. Regul Pept 2004; 117:77–88.
  35. Young AA, Gedulin BR, Bhavsar S, et al. Glucose-lowering and insulin-sensitizing actions of exendin-4: studies in obese diabetic (ob/ob, db/db) mice, diabetic fatty Zucker rats, and diabetic rhesus monkeys (Macaca mulatta). Diabetes 1999; 48:1026–1034.
  36. Kim D, MacConell L, Zhuang D, et al. Effects of once-weekly dosing of a long-acting release formulation of exenatide on glucose control and body weight in subjects with type 2 diabetes. Diabetes Care 2007; 30:1487–1493.
  37. Buse JB, Henry RR, Han J, et al. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in sulfonylurea-treated patients with type 2 diabetes. Diabetes Care 2004; 27:2628–2635.
  38. DeFronzo RA, Ratner RE, Han J, Kim DD, Fineman MS, Baron AD. Effects of exenatide (exendin-4) on glycemic control and weight over 30 weeks in metformin-treated patients with type 2 diabetes. Diabetes Care 2005; 28:1092–1100.
  39. Kendall DM, Riddle MC, Rosenstock J, et al. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in patients with type 2 diabetes treated with metformin and a sulfonylurea. Diabetes Care 2005; 28:1083–1091.
  40. Zinman B, Hoogwerf BJ, Durán García S, et al. The effect of adding exenatide to a thiazolidinedione in suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med 2007; 146:477–485.
  41. Klonoff DC, Buse JB, Nielsen LL, et al. Exenatide effects on diabetes, obesity, cardiovascular risk factors and hepatic biomarkers in patients with type 2 diabetes treated for at least 3 years. Curr Med Res Opin 2008; 24:275–286.
  42. Heine RJ, Van Gaal LF, Johns D, Mihm MJ, Widel MH, Brodows RG; for the GWAA Study Group. Exenatide versus insulin glargine in patients with suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med 2005; 143:559–569.
  43. Barnett AH, Burger J, Johns D, et al. Tolerability and efficacy of exenatide and titrated insulin glargine in adult patients with type 2 diabetes previously uncontrolled with metformin or a sulfonylurea: a multinational, randomized, open-label, two-period, crossover noninferiority trial. Clin Ther 2007; 29:2333–2348.
  44. Nauck MA, Duran S, Kim D, et al. A comparison of twice-daily exenatide and biphasic insulin aspart in patients with type 2 diabetes who were suboptimally controlled with sulfonylurea and metformin: a non-inferiority study. Diabetologia 2007; 50:259–267.
  45. Glass LC, Qu Y, Lenox S, et al. Effects of exenatide versus insulin analogues on weight change in subjects with type 2 diabetes: a pooled post-hoc analysis. Curr Med Res Opin 2008; 24:639–644.
  46. Drucker DJ, Buse JB, Taylor K, et al; for the DURATION-1 Study Group. Exenatide once weekly versus twice daily for the treatment of type 2 diabetes: a randomised, open-label, non-inferiority study. Lancet 2008; 372:1240–1250.
  47. Garber A, Henry R, Ratner R, et al; for the LEAD-3 (Mono) Study Group. Liraglutide versus glimepiride monotherapy for type 2 diabetes (LEAD-3 Mono): a randomised, 52-week, phase III, double-blind, parallel-treatment trial. Lancet 2009; 373:473–481.
  48. Nauck M, Frid A, Hermansen K, et al; for the LEAD-2 Study Group. Efficacy and safety comparison of liraglutide, glimepiride, and placebo, all in combination with metformin, in type 2 diabetes: the LEAD (Liraglutide Effect and Action in Diabetes)-2 study. Diabetes Care 2009; 32:84–90.
  49. Ahrén B, Landin-Olsson M, Jansson PA, Svensson M, Holmes D, Schweizer A. Inhibition of dipeptidyl peptidase-4 reduces glycemia, sustains insulin levels, and reduces glucagon levels in type 2 diabetes. J Clin Endocrinol Metab 2004; 89:2078–2084.
  50. Herman GA, Stevens C, Van Dyck K, et al. Pharmacokinetics and pharmacodynamics of sitagliptin, an inhibitor of dipeptidyl peptidase IV, in healthy subjects: results from two randomized, double-blind, placebo-controlled studies with single oral doses. Clin Pharmacol Ther 2005; 78:675–688.
  51. Bohannon N. Overview of the gliptin class (dipeptidyl peptidase-4 inhibitors) in clinical practice. Postgrad Med 2009; 121:40–45.
  52. US Department of Health and Human Services. FDA approves new drug treatment for type 2 diabetes. US Food and Drug Administration Web site. http://www.fda.gov/newsevents/newsroom/pressannouncements/ucm174780.htm. Published July 31, 2009. Accessed September 18, 2009.
  53. Raz I, Hanefeld M, Xu L, Caria C, Williams-Herman D, Khatami H; for the Sitagliptin Study 023 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy in patients with type 2 diabetes mellitus. Diabetologia 2006; 49:2564–2571.
  54. Zerilli T, Pyon EY. Sitagliptin phosphate: a DPP-4 inhibitor for the treatment of type 2 diabetes mellitus. Clin Ther 2007; 29:2614–2634.
  55. Scott R, Loeys T, Davies MJ, Engel SS; for the Sitagliptin Study 801 Group. Efficacy and safety of sitagliptin when added to ongoing metformin therapy in patients with type 2 diabetes. Diabetes Obes Metab 2008; 10:959–969.
  56. Williams-Herman D, Johnson J, Teng R, et al. Efficacy and safety of initial combination therapy with sitagliptin and metformin in patients with type 2 diabetes: a 54-week study. Curr Med Res Opin 2009; 25:569–583.
  57. DeFronzo RA, Okerson T, Viswanathan P, Guan X, Holcombe JH, MacConell L. Effects of exenatide versus sitagliptin on postprandial glucose, insulin and glucagon secretion, gastric emptying, and caloric intake: a randomized, cross-over study. Curr Med Res Opin 2008; 24:2943–2952.
References
  1. AACE Diabetes Mellitus Clinical Practice Guidelines Task Force. American Association of Clinical Endocrinologists medical guidelines for clinical practice for the management of diabetes mellitus. Endocr Pract 2007; 13(suppl 1):S4–S68.
  2. American Diabetes Association. Standards of medical care in diabetes—2009. Diabetes Care 2009; 32(suppl 1):S13–S61.
  3. Nathan DM, Buse JB, Davidson MB, et al. Medical management of hyperglycemia in type 2 diabetes: a consensus algorithm for the initiation and adjustment of therapy: a consensus statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care 2009; 32:193–203.
  4. National Institute of Diabetes and Digestive and Kidney Diseases. National Diabetes Statistics, 2007 fact sheet. National Institutes of Health Web site. http://www.diabetes.niddk.nih.gov/dm/pubs/statistics/index.htm. Published 2008. Accessed September 16, 2009.
  5. Koro CE, Bowlin SJ, Bourgeois N, Fedder DO. Glycemic control from 1988 to 2000 among US adults diagnosed with type 2 diabetes: a preliminary report. Diabetes Care 2004; 27:17–20.
  6. Ong KL, Cheung BM, Wong LY, Wat NM, Tan KC, Lam KS. Prevalence, treatment, and control of diagnosed diabetes in the US National Health and Nutrition Examination Survey 1999–2004. Ann Epidemiol 2008; 18:222–229.
  7. Koopman RJ, Mainous AG III, Diaz VA, Geesey ME. Changes in age at diagnosis of type 2 diabetes mellitus in the United States, 1988 to 2000. Ann Fam Med 2005; 3:60–63.
  8. Alexander GC, Sehgal NL, Moloney RM, Stafford RS. National trends in treatment of type 2 diabetes mellitus, 1994–2007. Arch Intern Med 2008; 168:2088–2094.
  9. The Action to Control Cardiovascular Risk in Diabetes Study Group. Effects of intensive glucose lowering in type 2 diabetes. N Engl J Med 2008; 358:2545–2559.
  10. The ADVANCE Collaborative Group. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N Engl J Med 2008; 358:2560–2572.
  11. Duckworth W, Abraira C, Moritz T, et al. Glucose control and vascular complications in veterans with type 2 diabetes. N Engl J Med 2009; 360:129–139.
  12. Jenssen TG, Tonstad S, Claudi T, Midthjell K, Cooper J. The gap between guidelines and practice in the treatment of type 2 diabetes: a nationwide survey in Norway. Diabetes Res Clin Pract 2008; 80:314–320.
  13. Gaede P, Lund-Andersen H, Parving HH, Pedersen O. Effect of a multifactorial intervention on mortality in type 2 diabetes. N Engl J Med 2008; 358:580–591.
  14. Wild S, Roglic G, Green A, Sicree R, King H. Global prevalence of diabetes: estimates for the year 2000 and projections for 2030. Diabetes Care 2004; 27:1047–1053.
  15. Rosenstock J. Management of type 2 diabetes mellitus in the elderly: special considerations. Drugs Aging 2001; 18:31–44.
  16. Brandle M, Zhou H, Smith BR, et al. The direct medical cost of type 2 diabetes. Diabetes Care 2003; 26:2300–2304.
  17. National Center for Health Statistics. Prevalence of overweight and obesity among adults: United States 2003–2004. Centers for Disease Contral and Prevention Web site. http://www.cdc.gov/nchs/products/pubs/pubd/hestats/overweight/overwght_adult_03.htm. Published: April 2006. Accessed September 23, 2009.
  18. Van Gaal LF, Gutkin SW, Nauck MA. Exploiting the antidiabetic properties of incretins to treat type 2 diabetes mellitus: glucagon-like peptide 1 receptor agonists or insulin for patients with inadequate glycemic control. Eur J Endocrinol 2008; 158:773–784.
  19. Anderson JW, Kendall CW, Jenkins DJ. Importance of weight management in type 2 diabetes: review with meta-analysis of clinical studies. J Am Coll Nutr 2003; 22:331–339.
  20. Fox CS, Pencina MJ, Wilson PW, Paynter NP, Vasan RS, D’Agostino RB Sr. Lifetime risk of cardiovascular disease among individuals with and without diabetes stratified by obesity status in the Framingham heart study. Diabetes Care 2008; 31:1582–1584.
  21. DeFronzo RA. From the triumvirate to the ominous octet: a new paradigm for the treatment of type 2 diabetes mellitus. Diabetes 2009; 58:773–795.
  22. Stonehouse A, Okerson T, Kendall D, Maggs D. Emerging incretin based therapies for type 2 diabetes: incretin mimetics and DPP-4 inhibitors. Curr Diabetes Rev 2008; 4:101–109.
  23. Cefalu WT. Pharmacotherapy for the treatment of patients with type 2 diabetes mellitus: rationale and specific agents. Clin Pharmacol Ther 2007; 81:636–649.
  24. Henry RR. Evolving concepts of type 2 diabetes management with oral medications: new approaches to an old disease. Curr Med Res Opin 2008; 24:2189–2202.
  25. American Diabetes Association. Nutrition recommendations and interventions for diabetes: a position statement of the American Diabetes Association. Diabetes Care 2008; 31(suppl 1):S61−S78.
  26. US Department of Health and Human Services (HHS) and US Department of Agriculture. Dietary guidelines for Americans, 2005. US Department of HHS Web site. http://www.health.gov/DietaryGuidelines/dga2005/document/default.htm. Published January 2005. Accessed September 25, 2009.
  27. National Heart, Lung, and Blood Institute. The practical guide: identification, evaluation, and treatment of overweight and obesity in adults. National Institutes of Health Web site. http://www.nhlbi.nih.gov/guidelines/obesity/prctgd_c.pdf. Updated: October 2000. Accessed September 28, 2009.
  28. Look AHEAD Research Group. Reduction in weight and cardio­vascular disease risk factors in individuals with type 2 diabetes: one-year results of the Look AHEAD trial. Diabetes Care 2007; 30:1374–1383.
  29. Williamson DA, Rejeski J, Lang W, Van Dorsten B, Fabricatore AN, Toledo K; for the Look AHEAD Research Group. Impact of a weight management program on health-related quality of life in overweight adults with type 2 diabetes. Arch Intern Med 2009; 169:163–171.
  30. Klein S, Sheard NF, Pi-Sunyer X, et al; for the American Diabetes Association; North American Association for the Study of Obesity; American Society for Clinical Nutrition. Weight management through lifestyle modification for the prevention and management of type 2 diabetes: rationale and strategies: a statement of the American Diabetes Association, the North American Association for the Study of Obesity, and the American Society for Clinical Nutrition. Diabetes Care 2004; 27:2067–2073.
  31. Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults. Executive summary of the Third Report of The National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III). JAMA 2001; 285:2486–2497.
  32. Buse JB, Ginsberg HN, Bakris GL, et al. Primary prevention of cardiovascular diseases in people with diabetes mellitus: a scientific statement from the American Heart Association and the American Diabetes Association. Circulation 2007; 115:114–126.
  33. Amori RE, Lau J, Pittas AG. Efficacy and safety of incretin therapy in type 2 diabetes: systematic review and meta-analysis. JAMA 2007; 298:194–206.
  34. Nielsen LL, Young AA, Parkes DG. Pharmacology of exenatide (synthetic exendin-4): a potential therapeutic for improved glycemic control of type 2 diabetes. Regul Pept 2004; 117:77–88.
  35. Young AA, Gedulin BR, Bhavsar S, et al. Glucose-lowering and insulin-sensitizing actions of exendin-4: studies in obese diabetic (ob/ob, db/db) mice, diabetic fatty Zucker rats, and diabetic rhesus monkeys (Macaca mulatta). Diabetes 1999; 48:1026–1034.
  36. Kim D, MacConell L, Zhuang D, et al. Effects of once-weekly dosing of a long-acting release formulation of exenatide on glucose control and body weight in subjects with type 2 diabetes. Diabetes Care 2007; 30:1487–1493.
  37. Buse JB, Henry RR, Han J, et al. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in sulfonylurea-treated patients with type 2 diabetes. Diabetes Care 2004; 27:2628–2635.
  38. DeFronzo RA, Ratner RE, Han J, Kim DD, Fineman MS, Baron AD. Effects of exenatide (exendin-4) on glycemic control and weight over 30 weeks in metformin-treated patients with type 2 diabetes. Diabetes Care 2005; 28:1092–1100.
  39. Kendall DM, Riddle MC, Rosenstock J, et al. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in patients with type 2 diabetes treated with metformin and a sulfonylurea. Diabetes Care 2005; 28:1083–1091.
  40. Zinman B, Hoogwerf BJ, Durán García S, et al. The effect of adding exenatide to a thiazolidinedione in suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med 2007; 146:477–485.
  41. Klonoff DC, Buse JB, Nielsen LL, et al. Exenatide effects on diabetes, obesity, cardiovascular risk factors and hepatic biomarkers in patients with type 2 diabetes treated for at least 3 years. Curr Med Res Opin 2008; 24:275–286.
  42. Heine RJ, Van Gaal LF, Johns D, Mihm MJ, Widel MH, Brodows RG; for the GWAA Study Group. Exenatide versus insulin glargine in patients with suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med 2005; 143:559–569.
  43. Barnett AH, Burger J, Johns D, et al. Tolerability and efficacy of exenatide and titrated insulin glargine in adult patients with type 2 diabetes previously uncontrolled with metformin or a sulfonylurea: a multinational, randomized, open-label, two-period, crossover noninferiority trial. Clin Ther 2007; 29:2333–2348.
  44. Nauck MA, Duran S, Kim D, et al. A comparison of twice-daily exenatide and biphasic insulin aspart in patients with type 2 diabetes who were suboptimally controlled with sulfonylurea and metformin: a non-inferiority study. Diabetologia 2007; 50:259–267.
  45. Glass LC, Qu Y, Lenox S, et al. Effects of exenatide versus insulin analogues on weight change in subjects with type 2 diabetes: a pooled post-hoc analysis. Curr Med Res Opin 2008; 24:639–644.
  46. Drucker DJ, Buse JB, Taylor K, et al; for the DURATION-1 Study Group. Exenatide once weekly versus twice daily for the treatment of type 2 diabetes: a randomised, open-label, non-inferiority study. Lancet 2008; 372:1240–1250.
  47. Garber A, Henry R, Ratner R, et al; for the LEAD-3 (Mono) Study Group. Liraglutide versus glimepiride monotherapy for type 2 diabetes (LEAD-3 Mono): a randomised, 52-week, phase III, double-blind, parallel-treatment trial. Lancet 2009; 373:473–481.
  48. Nauck M, Frid A, Hermansen K, et al; for the LEAD-2 Study Group. Efficacy and safety comparison of liraglutide, glimepiride, and placebo, all in combination with metformin, in type 2 diabetes: the LEAD (Liraglutide Effect and Action in Diabetes)-2 study. Diabetes Care 2009; 32:84–90.
  49. Ahrén B, Landin-Olsson M, Jansson PA, Svensson M, Holmes D, Schweizer A. Inhibition of dipeptidyl peptidase-4 reduces glycemia, sustains insulin levels, and reduces glucagon levels in type 2 diabetes. J Clin Endocrinol Metab 2004; 89:2078–2084.
  50. Herman GA, Stevens C, Van Dyck K, et al. Pharmacokinetics and pharmacodynamics of sitagliptin, an inhibitor of dipeptidyl peptidase IV, in healthy subjects: results from two randomized, double-blind, placebo-controlled studies with single oral doses. Clin Pharmacol Ther 2005; 78:675–688.
  51. Bohannon N. Overview of the gliptin class (dipeptidyl peptidase-4 inhibitors) in clinical practice. Postgrad Med 2009; 121:40–45.
  52. US Department of Health and Human Services. FDA approves new drug treatment for type 2 diabetes. US Food and Drug Administration Web site. http://www.fda.gov/newsevents/newsroom/pressannouncements/ucm174780.htm. Published July 31, 2009. Accessed September 18, 2009.
  53. Raz I, Hanefeld M, Xu L, Caria C, Williams-Herman D, Khatami H; for the Sitagliptin Study 023 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy in patients with type 2 diabetes mellitus. Diabetologia 2006; 49:2564–2571.
  54. Zerilli T, Pyon EY. Sitagliptin phosphate: a DPP-4 inhibitor for the treatment of type 2 diabetes mellitus. Clin Ther 2007; 29:2614–2634.
  55. Scott R, Loeys T, Davies MJ, Engel SS; for the Sitagliptin Study 801 Group. Efficacy and safety of sitagliptin when added to ongoing metformin therapy in patients with type 2 diabetes. Diabetes Obes Metab 2008; 10:959–969.
  56. Williams-Herman D, Johnson J, Teng R, et al. Efficacy and safety of initial combination therapy with sitagliptin and metformin in patients with type 2 diabetes: a 54-week study. Curr Med Res Opin 2009; 25:569–583.
  57. DeFronzo RA, Okerson T, Viswanathan P, Guan X, Holcombe JH, MacConell L. Effects of exenatide versus sitagliptin on postprandial glucose, insulin and glucagon secretion, gastric emptying, and caloric intake: a randomized, cross-over study. Curr Med Res Opin 2008; 24:2943–2952.
Page Number
S39-S47
Page Number
S39-S47
Publications
Publications
Article Type
Display Headline
Redefining treatment success in type 2 diabetes mellitus: Comprehensive targeting of core defects
Display Headline
Redefining treatment success in type 2 diabetes mellitus: Comprehensive targeting of core defects
Citation Override
Cleveland Clinic Journal of Medicine 2009 December;76(suppl 5):S39-S47
Inside the Article

KEY POINTS

  • The NHANES 1999–2004 data showed that only 13.2% of patients with diagnosed diabetes achieved concurrent weight, blood pressure, and lipid level goals.
  • Among patients with T2DM, lifestyle intervention (control of weight, blood pressure, lipid levels) should be reinforced at every physician visit; glycosylated hemoglobin (HbA1c) should be monitored every 3 months until it is less than 7.0%, and then rechecked every 6 months.
  • The effects of GLP-1 agonists on HbA1c are comparable to insulin analogues, but GLP-1 agonists are associated with weight reduction, while insulin is associated with weight gain.
  • DPP-4 inhibitors have been associated with significant reductions in HbA1c when used alone or with metformin or pioglitazone.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Fri, 04/13/2018 - 10:30
Un-Gate On Date
Fri, 04/13/2018 - 10:30
Use ProPublica
CFC Schedule Remove Status
Fri, 04/13/2018 - 10:30
Article PDF Media

Author and subject indexes: 2009

Article Type
Changed
Mon, 01/28/2019 - 16:48
Display Headline
Author and subject indexes: 2009
Article PDF
Issue
Cleveland Clinic Journal of Medicine - 76(12)
Publications
Page Number
731-734
Sections
Article PDF
Article PDF
Issue
Cleveland Clinic Journal of Medicine - 76(12)
Issue
Cleveland Clinic Journal of Medicine - 76(12)
Page Number
731-734
Page Number
731-734
Publications
Publications
Article Type
Display Headline
Author and subject indexes: 2009
Display Headline
Author and subject indexes: 2009
Sections
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media

An algorithm for managing warfarin resistance

Article Type
Changed
Thu, 04/12/2018 - 12:08
Display Headline
An algorithm for managing warfarin resistance

Warfarin (coumadin) differs from most other drugs in that the dosage required to achieve a desired therapeutic effect varies greatly among individuals. This variability can lead to therapeutic failure, potentially resulting in new thrombosis, or, at the other extreme, to life-threatening bleeding.

Further, there is no reliable means to identify patients who require unusually high doses of warfarin, although genetic testing may become available in the future.

See related patient information

Warfarin, a coumarin derivative first synthesized in 1948, is still the only oral anticoagulant available for long-term use in the United States. Indications for its use include the treatment and, to a lesser extent, the prevention of arterial and venous thromboembolism. It is also used for long-term anticoagulation in patients with atrial arrhythmias (atrial fibrillation and atrial flutter) and mechanical heart valves.

In the paragraphs that follow, we review the causes of warfarin resistance and how to recognize and manage it.

WHAT IS WARFARIN RESISTANCE?

Resistance to warfarin has been described as the inability to prolong the prothrombin time or raise the international normalized ratio (INR) into the therapeutic range when the drug is given at normally prescribed doses.1

However, a higher warfarin requirement does not itself establish the diagnosis of warfarin resistance. The prevalence of warfarin resistance varies by patient population and is difficult to determine. The difficulty lies largely in accounting for dietary factors and in defining normal metabolic variations among individuals.

The range of normally recommended daily or weekly warfarin doses to maintain a therapeutic prothrombin time or INR depends on the study population. Nevertheless, patients who need more than 105 mg per week (15 mg/day) should be considered warfarin-resistant. These patients are likely to be in the top 5% for warfarin doses within an anticoagulated cohort.

Warfarin resistance is different than warfarin failure, which is defined as a new thrombotic event despite a therapeutic prothrombin time and INR. This situation is commonly seen in patients with malignant diseases.

An important characteristic of warfarin resistance is that patients need much smaller doses of vitamin K to reverse the effect of warfarin.2 Thijssen3 showed that, in warfarin-resistant rats, warfarin did not irreversibly inhibit vitamin K1 2,3-epoxide reductase (VKORC1) activity. This is consistent with the vitamin K hypersensitivity observed in warfarin-resistant people.2,3

WHAT CAUSES WARFARIN RESISTANCE?

Warfarin resistance can be classified in practical terms as acquired vs hereditary, or in mechanistic terms as pharmacokinetic vs pharmacodynamic.

Acquired vs hereditary resistance

Hulse4 categorizes warfarin resistance as either acquired or hereditary.

Acquired resistance to warfarin may result from:

  • Poor patient compliance (the most common cause)
  • High consumption of vitamin K
  • Decreased absorption of warfarin
  • Increased clearance (see Warfarin is metabolized by P450 enzymes5–11)
  • Drug interactions (Table 1).12,13

Hereditary resistance has been postulated to be caused by genetic factors that result either in faster metabolism of the drug (a form of pharmacokinetic resistance) or in lower activity of the drug (pharmacodynamic resistance). Polymorphisms may play a role, as some VKORC1 and CYP2C9 variant alleles are known to be associated with increased sensitivity to warfarin.14

However, the genetic mechanisms of warfarin resistance are not clearly understood, despite several case reports of hereditary resistance confirmed by similar patterns of resistance in immediate family members.15–19 More than one mechanism is likely. There is ample room for further insight into genetic polymorphisms underlying hereditary warfarin resistance. More on this topic is included in the sections below.

 

 

Pharmacokinetic resistance

Pharmacokinetic resistance can result from diminished absorption or increased elimination of the drug. Causes of diminished absorption include emesis, diarrhea, and malabsorption syndrome.

The mechanism of increased warfarin clearance has not been delineated, although the following have been implicated.

Genetic factors. Duplication or multiplication of cytochrome P450 enzyme genes has been described as contributing to a phenotype of ultrarapid metabolism. Some people may carry multiple copies of the CYP2C9 gene, as has already been reported for cytochrome P450 CYP2D6 and CYP2A6.7,8 It is also plausible that rare allelic variants of CYP2C9 exist that are associated with higher-than-normal activity, given that there are alleles known to predispose to warfarin sensitivity.

Hypoalbuminemia may increase the free fraction of warfarin, leading to enhanced rates of clearance and a shorter plasma half-life.15

Hyperalbuminemia may paradoxically also contribute to warfarin resistance via drug binding.

Hyperlipidemia. Several observers have found that lowering serum lipids, primarily triglycerides, increases the sensitivity to warfarin irrespective of the means used to achieve this decrease.20 This most likely results in a decreased pool of vitamin K, some of which is bound to triglycerides.21 Conversely, patients receiving intravenous lipids with total parenteral nutrition have also been diagnosed clinically with warfarin resistance,22 and rat models have shown an association between a lipidrich diet and increased vitamin K-dependent factor activity.23

Diuretics may decrease the response to warfarin by reducing the plasma volume, with a subsequent increase in clotting factor activity.24

Pharmacodynamic resistance

Potential mechanisms of pharmacodynamic warfarin resistance described in rats and in people include:

  • Increased affinity of vitamin K1, 2,3-epoxide reductase complex (VKOR) for vitamin K25,26 (see How warfarin works2,10,11,27–30)
  • Prolongation of normal clotting factor activity16
  • Production of clotting factors that is not dependent on vitamin K16
  • Decreased VKOR sensitivity to warfarin.26

In rats, these mechanisms are manifested by relatively high doses of warfarin being required to achieve poisoning. In humans, they result in high doses being needed to achieve a therapeutic effect in the setting of normal warfarin pharmacokinetics, normal warfarin concentration, and normal half-lives of blood clotting proteins.

Figure 1.

Genetics of pharmacodynamic resistance. Pharmacodynamic warfarin resistance has also been described with inheritance of a monogenetic dominant trait. An early study by O’Reilly24 traced anticoagulation resistance to a genetically linked abnormality of interaction between warfarin and a putative vitamin K receptor.

In one patient with hereditary resistance and high warfarin requirements, a heterozygous point mutation in the VKORC1 gene was identified.31 This results in a substitution that lies in a conserved (normally constant or unchanging DNA sequence in a genome) region of VKORC1 that contains three of four previously identified amino acid substitutions associated with warfarin resistance (Val29Leu, Val45Ala, and Arg58Gly). Further investigation is required to fully characterize the structure-function relationship for VKORC1 and to determine the relationship between the VKORC1 genotype and other pharmacogenetic determinants of warfarin dose-response.

Separately, Loebstein et al32 reported a new mutation, Asp36Tyr, which was common in Jewish ethnic groups of Ethiopian descent (in whom the prevalence is 5%) and Ashkenazi descent (prevalence 4%). In that study, Asp36Tyr carriers needed doses of more than 70 mg per week, placing them towards the high end of the usual warfarin dosing range.

Daly and Aithal7 discovered that warfarinresistant rats overexpressed a protein known as calumenin. This protein is situated in the endoplasmic reticulum and appears to interact with VKOR, decreasing the binding of warfarin. In mice, the calumenin gene is located on chromosome 7, where the gene for VKORC1 is also located.

 

 

DIAGNOSIS BY HISTORY AND LABORATORY STUDIES

A full drug and diet history is invaluable in diagnosing potential causes of warfarin resistance (Table 1).

Plasma warfarin levels that are subtherapeutic should raise suspicion of intestinal malabsorption or poor compliance. Poor compliance might be more appropriately seen as a mimic of warfarin resistance. Studies in humans suggest that a therapeutic total plasma warfarin level lies between 0.5 μg/mL and 3.0 μg/mL,10 though the range may vary among laboratories and patient populations.

Warfarin absorption and clearance can be evaluated by analyzing plasma levels at specific intervals after administration, eg, every 60 to 180 minutes. The drug’s half-life can be determined on the basis of its concentrations in different time samples. Normally, the S-enantiomer of warfarin is cleared at twice the rate of the R-enantiomer (5.2 vs 2.5 mL/min/70 kg).8 A normal clearance rate confirms that resistance to warfarin is not due to enhanced elimination.

Clotting assays of factors II, VII, IX, and X may be a more precise way to assess the pharmacodynamics of warfarin,10 although there is no strong evidence to support routine use of such assays. Some studies suggest targeting factor II and factor X activity levels of 10% to 30% of normal biologic activity for a therapeutic warfarin effect in patients with an unreliable or prolonged baseline prothrombin time and INR, such as those with lupus anticoagulant.

Figure 2. Algorithm for evaluating suspected warfarin resistance.
An algorithm. Bentley et al33 suggest using the plasma warfarin level in an algorithm to determine the type of resistance pattern. Plasma warfarin levels are typically measured by regional specialized reference laboratories with a turnaround time of 2 to 7 days, as opposed to 24 hours for factor II and X activity. Our suggested algorithm for evaluation of suspected warfarin resistance is shown in Figure 2.

TREAT THE CAUSE

Once the type of warfarin resistance has been determined, treatment should be oriented toward the cause.

Educate the patient

The importance of compliance should be reinforced. Educating the patient about diet and other medications that may interact with warfarin is also important. (See an example of patient education material.)

Increase the warfarin dose

If the patient truly has hereditary resistance, there are two approaches to treatment.

The first is to increase the warfarin dose until the prothrombin time and INR are in the therapeutic ranges. When indicated, the warfarin dose can be safely titrated upward to more than 100 mg per day in patients who are monitored regularly—as all patients on chronic warfarin therapy should be—and whose other medications are otherwise stable. One such example is reported in a warfarinresistant patient who needed 145 mg/day to maintain a therapeutic prothrombin time.22

Try other anticoagulants?

The second approach is to change to another type of anticoagulant. However, there is no strong evidence in favor of this approach over prescribing larger dosages of warfarin.

Other anticoagulant drugs currently available in the United States include subcutaneous heparins (unfractionated and low-molecular-weight heparins) and the subcutaneous factor Xa inhibitor fondaparinux (Arixtra).

Agents not available in the United States include the following.

Dabigatran, an oral direct thrombin inhibitor, is undergoing phase 3 studies of its use for long-term anticoagulation.

Rivaroxaban (a direct factor Xa inhibitor) and dabigatran have been approved in Canada and the European Union to prevent venous thromboembolism after knee and hip arthroplasty, based on prospective comparisons with enoxaparin (Lovenox).34–37

Vitamin K antagonists other than warfarin that are not available in the United States include bishydroxycoumarin (which has limitations including slow absorption and high frequency of gastrointestinal side effects), phenprocoumon, and acenocoumarol. Another is phenindione, which has been associated with serious hypersensitivity reactions, some of which proved fatal and occurred within a few weeks of initiating therapy.

References
  1. Lefrere JJ, Horellou MH, Conard J, Samama M. Proposed classification of resistance to oral anticoagulant therapy. J Clin Pathol 1987; 40:242.
  2. Linder MW. Genetic mechanisms for hypersensitivity and resistance to the anticoagulant warfarin. Clin Chim Acta 2001; 308:915.
  3. Thijssen HH. Warfarin resistance. Vitamin K epoxide reductase of Scottish resistance gene is not irreversibly blocked by warfarin. Biochem Pharmacol 1987; 36:27532757.
  4. Hulse ML. Warfarin resistance: diagnosis and therapeutic alternative. Pharmacotherapy 1996; 16:10091017.
  5. Hirsh J, Dalen JE, Deykin D, Poller L, Bussey H. Oral anticoagulants. Mechanism of action, clinical effectiveness, and optimal therapeutic range. Chest 1995; 108( suppl 4):231S234S.
  6. Daly AK, King BP. Pharmacogenetics of oral anticoagulants. Pharmacogenetics 2003; 13:247252.
  7. Daly AK, Aithal GP. Genetic regulation of warfarin metabolism and response. Semin Vasc Med 2003; 3:231238.
  8. Takahashi H, Echizen H. Pharmacogenetics of warfarin elimination and its clinical implications. Clin Pharmacokinet 2001; 40:587603.
  9. Retti AE, Wienkers LC, Gonzalez FJ, Trager WF, Korezekwa KR. Impaired (S)-warfarin metabolism catalysed by the R144C allele variant of CYP2C9. Pharmacogenetics 1994; 4:3942.
  10. Porter RS, Sawyer WR. Warfarin. In:Evans WE, Shentag JJ, Jusko WJ, editors. Applied Pharmacokinetics. Principles of Therapeutics Drug Monitoring, 3rd ed. Washington, DC: Applied Therapeutics, 1992: 31.131.46.
  11. Warrell DA, Cox TM, Firth JD. Oxford Textbook of Medicine, 4th ed. Oxford University Press, 2003:734.
  12. Holbrook AM, Pereira JA, Labiris R, et al. Systematic overview of warfarin and its drug and food interactions. Arch Intern Med 2005; 165:10951106.
  13. Medical Economics Staff. Physicians’ Desk Reference, 55th Ed. Medical Economics, 2001:11391140.
  14. Schwarz UI, Ritchie MD, Bradford Y, et al. Genetic determinants of response to warfarin during initial anticoagulation. N Engl J Med 2008; 358:9991008.
  15. Diab F, Feffer S. Hereditary warfarin resistance. South Med J 1994; 87:407409.
  16. O’Reilly RA. The second reported kindred with hereditary resistance to oral anticoagulant drugs. N Engl J Med 1970; 282:14481451.
  17. O’Reilly RA, Aggeler PM, Hoag MS, Leong LS, Kropatkin ML. Hereditary transmission of exceptional resistance to coumarin anticoagulant drugs. The first reported kindred. N Engl J Med 1964; 271:809815.
  18. Alving BM, Strickler MP, Knight RD, Barr CF, Berenberg JL, Peek CC. Hereditary warfarin resistance. Investigation of rare phenomenon. Arch Intern Med 1985; 145:499501.
  19. Warrier L, Brennan CA, Lusher JM. Familial warfarin resistance in a black child. Am J Pediatr Hematol Oncol 1986; 8:346347.
  20. Nikkila EA, Pelkonen R. Serum lipid-reducing agents and anticoagulant requirement. Lancet 1963; 1:332.
  21. Robinson A, Liau FO, Routledge PA, Backhouse G, Spragg BP, Bentley DP. Lipids and warfarin requirements. Thromb Haemost 1990; 63:148149.
  22. MacLaren R, Wachsman BA, Swift DK, Kuhl DA. Warfarin resistance associated with intravenous lipid administration: discussion of propofol and review of the literature. Pharmacotherapy 1997; 17:13311337.
  23. DeCurtis A, D’Adamo MC, Amore C, et al. Experimental arterial thrombosis in genetically or diet induced hyperlipidemia in rats—role of vitamin K-dependent clotting factors and prevention by low-intensity oral anticoagulation. Thromb Haemost 2001; 86:14401448.
  24. O’Reilly RA. Drug interaction involving oral anticoagulation. In:Melmon KL, editor. Cardiovascular Drug Therapy, Philadelphia; FA Davis, 1975:2341.
  25. O’ Reilly RA, Pool JG, Aggeler PM. Hereditary resistance to coumarin anticoagulation drugs in man and rat. Ann N Y Acad Sci 1968; 151:913931.
  26. Cain D, Hutson SM, Wallin R. Warfarin resistance is associated with a protein component of the vitamin K 2,3-epoxide reductase enzyme complex in rat liver. Thromb Haemost 1998; 80:128133.
  27. Rodvold KA, Quandt CM, Friedenberg WR. Thromboembolic disorders. In:DiPiro JT, Talbert RL, editors. Pharmacotherapy. A Pathophysiologic Approach, 2nd ed. New York: Elsevier, 1992:312335.
  28. Park BK. Warfarin: metabolism and mode of action. Biochem Pharmacol 1988; 37:1927.
  29. Cain D, Hutson SM, Wallin R. Assembly of the warfarin-sensitive vitamin K 2,3-epoxide reductase enzyme complex in the endoplasmic reticulum membrane. J Biol Chem 1997; 272:2906829075.
  30. Gallop PM, Lian JB, Hauschka PV. Carboxylated calcium binding proteins and vitamin K. N Engl J Med 1980; 302:14601466.
  31. Rost S, Fregin A, Ivaskevicius V, et al. Mutations in VKORC1 cause warfarin resistance and multiple coagulation factor deficiency type 2. Nature 2004; 427:537541.
  32. Loebstein R, Dovskin I, Halkin H, et al. A coding VKORC1 Asp36-Tyr polymorphism predisposes to warfarin resistance. Blood 2007; 109:24772480.
  33. Bentley DP, Backhouse G, Hutchings A, Haddon RL, Spragg B, Routledge PA. Investigation of patients with abnormal response to warfarin. Br J Clin Pharmacol 1986; 22:3741.
  34. Eriksson BI, Borris LC, Friedman RJ, et al. RECORD1 Study Group. Rivaroxaban versus enoxaparin for thromboprophylaxis after hip arthroplasty. N Engl J Med 2008; 358:27652775.
  35. Kakkar AK, Brenner B, Dahl OE, et al; RECORD2 Investigators. Extended duration rivaroxaban versus short-term enoxaparin for the prevention of venous thromboembolism after total hip arthroplasty: a double-blind, randomised controlled trial. Lancet 2008; 372:3139.
  36. Lassen MR, Ageno W, Borris LC, et al; RECORD3 Investigators. Rivaroxaban versus enoxaparin for thromboprophylaxis after total knee arthroplasty. N Engl J Med 2008; 358:27762786.
  37. Wolowacz SE, Roskell NS, Plumb JM, Caprini JA, Eriksson BI. Efficacy and safety of dabigatran etexilate for the prevention of venous thromboembolism following total hip or knee arthroplasty. A meta-analysis. Thromb Haemost 2009; 101:7785.
Article PDF
Author and Disclosure Information

Olusegun Osinbowale, MD, MBA, RPVI
Department of Cardiology, Section of Noninvasive Cardiology, Ochsner Clinic Foundation, New Orleans, LA

Monzr Al Malki, MD
Biotherapeutics Department Laboratory, Division of Surgical Research, Boston University School of Medicine, Roger Williams Medical Center, Providence, RI

Andrew Schade, MD, PhD
Division of Pathology and Laboratory Medicine, Department of Clinical Pathology, Cleveland Clinic

John R. Bartholomew, MD
Department of Cardiovascular Medicine, Head, Section of Vascular Medicine, Cleveland Clinic

Address: John R. Bartholomew, MD, Department of Cardiovascular Medicine, Section of Vascular Medicine, J3-5, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail [email protected]

Issue
Cleveland Clinic Journal of Medicine - 76(12)
Publications
Topics
Page Number
724-730
Sections
Author and Disclosure Information

Olusegun Osinbowale, MD, MBA, RPVI
Department of Cardiology, Section of Noninvasive Cardiology, Ochsner Clinic Foundation, New Orleans, LA

Monzr Al Malki, MD
Biotherapeutics Department Laboratory, Division of Surgical Research, Boston University School of Medicine, Roger Williams Medical Center, Providence, RI

Andrew Schade, MD, PhD
Division of Pathology and Laboratory Medicine, Department of Clinical Pathology, Cleveland Clinic

John R. Bartholomew, MD
Department of Cardiovascular Medicine, Head, Section of Vascular Medicine, Cleveland Clinic

Address: John R. Bartholomew, MD, Department of Cardiovascular Medicine, Section of Vascular Medicine, J3-5, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail [email protected]

Author and Disclosure Information

Olusegun Osinbowale, MD, MBA, RPVI
Department of Cardiology, Section of Noninvasive Cardiology, Ochsner Clinic Foundation, New Orleans, LA

Monzr Al Malki, MD
Biotherapeutics Department Laboratory, Division of Surgical Research, Boston University School of Medicine, Roger Williams Medical Center, Providence, RI

Andrew Schade, MD, PhD
Division of Pathology and Laboratory Medicine, Department of Clinical Pathology, Cleveland Clinic

John R. Bartholomew, MD
Department of Cardiovascular Medicine, Head, Section of Vascular Medicine, Cleveland Clinic

Address: John R. Bartholomew, MD, Department of Cardiovascular Medicine, Section of Vascular Medicine, J3-5, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail [email protected]

Article PDF
Article PDF

Warfarin (coumadin) differs from most other drugs in that the dosage required to achieve a desired therapeutic effect varies greatly among individuals. This variability can lead to therapeutic failure, potentially resulting in new thrombosis, or, at the other extreme, to life-threatening bleeding.

Further, there is no reliable means to identify patients who require unusually high doses of warfarin, although genetic testing may become available in the future.

See related patient information

Warfarin, a coumarin derivative first synthesized in 1948, is still the only oral anticoagulant available for long-term use in the United States. Indications for its use include the treatment and, to a lesser extent, the prevention of arterial and venous thromboembolism. It is also used for long-term anticoagulation in patients with atrial arrhythmias (atrial fibrillation and atrial flutter) and mechanical heart valves.

In the paragraphs that follow, we review the causes of warfarin resistance and how to recognize and manage it.

WHAT IS WARFARIN RESISTANCE?

Resistance to warfarin has been described as the inability to prolong the prothrombin time or raise the international normalized ratio (INR) into the therapeutic range when the drug is given at normally prescribed doses.1

However, a higher warfarin requirement does not itself establish the diagnosis of warfarin resistance. The prevalence of warfarin resistance varies by patient population and is difficult to determine. The difficulty lies largely in accounting for dietary factors and in defining normal metabolic variations among individuals.

The range of normally recommended daily or weekly warfarin doses to maintain a therapeutic prothrombin time or INR depends on the study population. Nevertheless, patients who need more than 105 mg per week (15 mg/day) should be considered warfarin-resistant. These patients are likely to be in the top 5% for warfarin doses within an anticoagulated cohort.

Warfarin resistance is different than warfarin failure, which is defined as a new thrombotic event despite a therapeutic prothrombin time and INR. This situation is commonly seen in patients with malignant diseases.

An important characteristic of warfarin resistance is that patients need much smaller doses of vitamin K to reverse the effect of warfarin.2 Thijssen3 showed that, in warfarin-resistant rats, warfarin did not irreversibly inhibit vitamin K1 2,3-epoxide reductase (VKORC1) activity. This is consistent with the vitamin K hypersensitivity observed in warfarin-resistant people.2,3

WHAT CAUSES WARFARIN RESISTANCE?

Warfarin resistance can be classified in practical terms as acquired vs hereditary, or in mechanistic terms as pharmacokinetic vs pharmacodynamic.

Acquired vs hereditary resistance

Hulse4 categorizes warfarin resistance as either acquired or hereditary.

Acquired resistance to warfarin may result from:

  • Poor patient compliance (the most common cause)
  • High consumption of vitamin K
  • Decreased absorption of warfarin
  • Increased clearance (see Warfarin is metabolized by P450 enzymes5–11)
  • Drug interactions (Table 1).12,13

Hereditary resistance has been postulated to be caused by genetic factors that result either in faster metabolism of the drug (a form of pharmacokinetic resistance) or in lower activity of the drug (pharmacodynamic resistance). Polymorphisms may play a role, as some VKORC1 and CYP2C9 variant alleles are known to be associated with increased sensitivity to warfarin.14

However, the genetic mechanisms of warfarin resistance are not clearly understood, despite several case reports of hereditary resistance confirmed by similar patterns of resistance in immediate family members.15–19 More than one mechanism is likely. There is ample room for further insight into genetic polymorphisms underlying hereditary warfarin resistance. More on this topic is included in the sections below.

 

 

Pharmacokinetic resistance

Pharmacokinetic resistance can result from diminished absorption or increased elimination of the drug. Causes of diminished absorption include emesis, diarrhea, and malabsorption syndrome.

The mechanism of increased warfarin clearance has not been delineated, although the following have been implicated.

Genetic factors. Duplication or multiplication of cytochrome P450 enzyme genes has been described as contributing to a phenotype of ultrarapid metabolism. Some people may carry multiple copies of the CYP2C9 gene, as has already been reported for cytochrome P450 CYP2D6 and CYP2A6.7,8 It is also plausible that rare allelic variants of CYP2C9 exist that are associated with higher-than-normal activity, given that there are alleles known to predispose to warfarin sensitivity.

Hypoalbuminemia may increase the free fraction of warfarin, leading to enhanced rates of clearance and a shorter plasma half-life.15

Hyperalbuminemia may paradoxically also contribute to warfarin resistance via drug binding.

Hyperlipidemia. Several observers have found that lowering serum lipids, primarily triglycerides, increases the sensitivity to warfarin irrespective of the means used to achieve this decrease.20 This most likely results in a decreased pool of vitamin K, some of which is bound to triglycerides.21 Conversely, patients receiving intravenous lipids with total parenteral nutrition have also been diagnosed clinically with warfarin resistance,22 and rat models have shown an association between a lipidrich diet and increased vitamin K-dependent factor activity.23

Diuretics may decrease the response to warfarin by reducing the plasma volume, with a subsequent increase in clotting factor activity.24

Pharmacodynamic resistance

Potential mechanisms of pharmacodynamic warfarin resistance described in rats and in people include:

  • Increased affinity of vitamin K1, 2,3-epoxide reductase complex (VKOR) for vitamin K25,26 (see How warfarin works2,10,11,27–30)
  • Prolongation of normal clotting factor activity16
  • Production of clotting factors that is not dependent on vitamin K16
  • Decreased VKOR sensitivity to warfarin.26

In rats, these mechanisms are manifested by relatively high doses of warfarin being required to achieve poisoning. In humans, they result in high doses being needed to achieve a therapeutic effect in the setting of normal warfarin pharmacokinetics, normal warfarin concentration, and normal half-lives of blood clotting proteins.

Figure 1.

Genetics of pharmacodynamic resistance. Pharmacodynamic warfarin resistance has also been described with inheritance of a monogenetic dominant trait. An early study by O’Reilly24 traced anticoagulation resistance to a genetically linked abnormality of interaction between warfarin and a putative vitamin K receptor.

In one patient with hereditary resistance and high warfarin requirements, a heterozygous point mutation in the VKORC1 gene was identified.31 This results in a substitution that lies in a conserved (normally constant or unchanging DNA sequence in a genome) region of VKORC1 that contains three of four previously identified amino acid substitutions associated with warfarin resistance (Val29Leu, Val45Ala, and Arg58Gly). Further investigation is required to fully characterize the structure-function relationship for VKORC1 and to determine the relationship between the VKORC1 genotype and other pharmacogenetic determinants of warfarin dose-response.

Separately, Loebstein et al32 reported a new mutation, Asp36Tyr, which was common in Jewish ethnic groups of Ethiopian descent (in whom the prevalence is 5%) and Ashkenazi descent (prevalence 4%). In that study, Asp36Tyr carriers needed doses of more than 70 mg per week, placing them towards the high end of the usual warfarin dosing range.

Daly and Aithal7 discovered that warfarinresistant rats overexpressed a protein known as calumenin. This protein is situated in the endoplasmic reticulum and appears to interact with VKOR, decreasing the binding of warfarin. In mice, the calumenin gene is located on chromosome 7, where the gene for VKORC1 is also located.

 

 

DIAGNOSIS BY HISTORY AND LABORATORY STUDIES

A full drug and diet history is invaluable in diagnosing potential causes of warfarin resistance (Table 1).

Plasma warfarin levels that are subtherapeutic should raise suspicion of intestinal malabsorption or poor compliance. Poor compliance might be more appropriately seen as a mimic of warfarin resistance. Studies in humans suggest that a therapeutic total plasma warfarin level lies between 0.5 μg/mL and 3.0 μg/mL,10 though the range may vary among laboratories and patient populations.

Warfarin absorption and clearance can be evaluated by analyzing plasma levels at specific intervals after administration, eg, every 60 to 180 minutes. The drug’s half-life can be determined on the basis of its concentrations in different time samples. Normally, the S-enantiomer of warfarin is cleared at twice the rate of the R-enantiomer (5.2 vs 2.5 mL/min/70 kg).8 A normal clearance rate confirms that resistance to warfarin is not due to enhanced elimination.

Clotting assays of factors II, VII, IX, and X may be a more precise way to assess the pharmacodynamics of warfarin,10 although there is no strong evidence to support routine use of such assays. Some studies suggest targeting factor II and factor X activity levels of 10% to 30% of normal biologic activity for a therapeutic warfarin effect in patients with an unreliable or prolonged baseline prothrombin time and INR, such as those with lupus anticoagulant.

Figure 2. Algorithm for evaluating suspected warfarin resistance.
An algorithm. Bentley et al33 suggest using the plasma warfarin level in an algorithm to determine the type of resistance pattern. Plasma warfarin levels are typically measured by regional specialized reference laboratories with a turnaround time of 2 to 7 days, as opposed to 24 hours for factor II and X activity. Our suggested algorithm for evaluation of suspected warfarin resistance is shown in Figure 2.

TREAT THE CAUSE

Once the type of warfarin resistance has been determined, treatment should be oriented toward the cause.

Educate the patient

The importance of compliance should be reinforced. Educating the patient about diet and other medications that may interact with warfarin is also important. (See an example of patient education material.)

Increase the warfarin dose

If the patient truly has hereditary resistance, there are two approaches to treatment.

The first is to increase the warfarin dose until the prothrombin time and INR are in the therapeutic ranges. When indicated, the warfarin dose can be safely titrated upward to more than 100 mg per day in patients who are monitored regularly—as all patients on chronic warfarin therapy should be—and whose other medications are otherwise stable. One such example is reported in a warfarinresistant patient who needed 145 mg/day to maintain a therapeutic prothrombin time.22

Try other anticoagulants?

The second approach is to change to another type of anticoagulant. However, there is no strong evidence in favor of this approach over prescribing larger dosages of warfarin.

Other anticoagulant drugs currently available in the United States include subcutaneous heparins (unfractionated and low-molecular-weight heparins) and the subcutaneous factor Xa inhibitor fondaparinux (Arixtra).

Agents not available in the United States include the following.

Dabigatran, an oral direct thrombin inhibitor, is undergoing phase 3 studies of its use for long-term anticoagulation.

Rivaroxaban (a direct factor Xa inhibitor) and dabigatran have been approved in Canada and the European Union to prevent venous thromboembolism after knee and hip arthroplasty, based on prospective comparisons with enoxaparin (Lovenox).34–37

Vitamin K antagonists other than warfarin that are not available in the United States include bishydroxycoumarin (which has limitations including slow absorption and high frequency of gastrointestinal side effects), phenprocoumon, and acenocoumarol. Another is phenindione, which has been associated with serious hypersensitivity reactions, some of which proved fatal and occurred within a few weeks of initiating therapy.

Warfarin (coumadin) differs from most other drugs in that the dosage required to achieve a desired therapeutic effect varies greatly among individuals. This variability can lead to therapeutic failure, potentially resulting in new thrombosis, or, at the other extreme, to life-threatening bleeding.

Further, there is no reliable means to identify patients who require unusually high doses of warfarin, although genetic testing may become available in the future.

See related patient information

Warfarin, a coumarin derivative first synthesized in 1948, is still the only oral anticoagulant available for long-term use in the United States. Indications for its use include the treatment and, to a lesser extent, the prevention of arterial and venous thromboembolism. It is also used for long-term anticoagulation in patients with atrial arrhythmias (atrial fibrillation and atrial flutter) and mechanical heart valves.

In the paragraphs that follow, we review the causes of warfarin resistance and how to recognize and manage it.

WHAT IS WARFARIN RESISTANCE?

Resistance to warfarin has been described as the inability to prolong the prothrombin time or raise the international normalized ratio (INR) into the therapeutic range when the drug is given at normally prescribed doses.1

However, a higher warfarin requirement does not itself establish the diagnosis of warfarin resistance. The prevalence of warfarin resistance varies by patient population and is difficult to determine. The difficulty lies largely in accounting for dietary factors and in defining normal metabolic variations among individuals.

The range of normally recommended daily or weekly warfarin doses to maintain a therapeutic prothrombin time or INR depends on the study population. Nevertheless, patients who need more than 105 mg per week (15 mg/day) should be considered warfarin-resistant. These patients are likely to be in the top 5% for warfarin doses within an anticoagulated cohort.

Warfarin resistance is different than warfarin failure, which is defined as a new thrombotic event despite a therapeutic prothrombin time and INR. This situation is commonly seen in patients with malignant diseases.

An important characteristic of warfarin resistance is that patients need much smaller doses of vitamin K to reverse the effect of warfarin.2 Thijssen3 showed that, in warfarin-resistant rats, warfarin did not irreversibly inhibit vitamin K1 2,3-epoxide reductase (VKORC1) activity. This is consistent with the vitamin K hypersensitivity observed in warfarin-resistant people.2,3

WHAT CAUSES WARFARIN RESISTANCE?

Warfarin resistance can be classified in practical terms as acquired vs hereditary, or in mechanistic terms as pharmacokinetic vs pharmacodynamic.

Acquired vs hereditary resistance

Hulse4 categorizes warfarin resistance as either acquired or hereditary.

Acquired resistance to warfarin may result from:

  • Poor patient compliance (the most common cause)
  • High consumption of vitamin K
  • Decreased absorption of warfarin
  • Increased clearance (see Warfarin is metabolized by P450 enzymes5–11)
  • Drug interactions (Table 1).12,13

Hereditary resistance has been postulated to be caused by genetic factors that result either in faster metabolism of the drug (a form of pharmacokinetic resistance) or in lower activity of the drug (pharmacodynamic resistance). Polymorphisms may play a role, as some VKORC1 and CYP2C9 variant alleles are known to be associated with increased sensitivity to warfarin.14

However, the genetic mechanisms of warfarin resistance are not clearly understood, despite several case reports of hereditary resistance confirmed by similar patterns of resistance in immediate family members.15–19 More than one mechanism is likely. There is ample room for further insight into genetic polymorphisms underlying hereditary warfarin resistance. More on this topic is included in the sections below.

 

 

Pharmacokinetic resistance

Pharmacokinetic resistance can result from diminished absorption or increased elimination of the drug. Causes of diminished absorption include emesis, diarrhea, and malabsorption syndrome.

The mechanism of increased warfarin clearance has not been delineated, although the following have been implicated.

Genetic factors. Duplication or multiplication of cytochrome P450 enzyme genes has been described as contributing to a phenotype of ultrarapid metabolism. Some people may carry multiple copies of the CYP2C9 gene, as has already been reported for cytochrome P450 CYP2D6 and CYP2A6.7,8 It is also plausible that rare allelic variants of CYP2C9 exist that are associated with higher-than-normal activity, given that there are alleles known to predispose to warfarin sensitivity.

Hypoalbuminemia may increase the free fraction of warfarin, leading to enhanced rates of clearance and a shorter plasma half-life.15

Hyperalbuminemia may paradoxically also contribute to warfarin resistance via drug binding.

Hyperlipidemia. Several observers have found that lowering serum lipids, primarily triglycerides, increases the sensitivity to warfarin irrespective of the means used to achieve this decrease.20 This most likely results in a decreased pool of vitamin K, some of which is bound to triglycerides.21 Conversely, patients receiving intravenous lipids with total parenteral nutrition have also been diagnosed clinically with warfarin resistance,22 and rat models have shown an association between a lipidrich diet and increased vitamin K-dependent factor activity.23

Diuretics may decrease the response to warfarin by reducing the plasma volume, with a subsequent increase in clotting factor activity.24

Pharmacodynamic resistance

Potential mechanisms of pharmacodynamic warfarin resistance described in rats and in people include:

  • Increased affinity of vitamin K1, 2,3-epoxide reductase complex (VKOR) for vitamin K25,26 (see How warfarin works2,10,11,27–30)
  • Prolongation of normal clotting factor activity16
  • Production of clotting factors that is not dependent on vitamin K16
  • Decreased VKOR sensitivity to warfarin.26

In rats, these mechanisms are manifested by relatively high doses of warfarin being required to achieve poisoning. In humans, they result in high doses being needed to achieve a therapeutic effect in the setting of normal warfarin pharmacokinetics, normal warfarin concentration, and normal half-lives of blood clotting proteins.

Figure 1.

Genetics of pharmacodynamic resistance. Pharmacodynamic warfarin resistance has also been described with inheritance of a monogenetic dominant trait. An early study by O’Reilly24 traced anticoagulation resistance to a genetically linked abnormality of interaction between warfarin and a putative vitamin K receptor.

In one patient with hereditary resistance and high warfarin requirements, a heterozygous point mutation in the VKORC1 gene was identified.31 This results in a substitution that lies in a conserved (normally constant or unchanging DNA sequence in a genome) region of VKORC1 that contains three of four previously identified amino acid substitutions associated with warfarin resistance (Val29Leu, Val45Ala, and Arg58Gly). Further investigation is required to fully characterize the structure-function relationship for VKORC1 and to determine the relationship between the VKORC1 genotype and other pharmacogenetic determinants of warfarin dose-response.

Separately, Loebstein et al32 reported a new mutation, Asp36Tyr, which was common in Jewish ethnic groups of Ethiopian descent (in whom the prevalence is 5%) and Ashkenazi descent (prevalence 4%). In that study, Asp36Tyr carriers needed doses of more than 70 mg per week, placing them towards the high end of the usual warfarin dosing range.

Daly and Aithal7 discovered that warfarinresistant rats overexpressed a protein known as calumenin. This protein is situated in the endoplasmic reticulum and appears to interact with VKOR, decreasing the binding of warfarin. In mice, the calumenin gene is located on chromosome 7, where the gene for VKORC1 is also located.

 

 

DIAGNOSIS BY HISTORY AND LABORATORY STUDIES

A full drug and diet history is invaluable in diagnosing potential causes of warfarin resistance (Table 1).

Plasma warfarin levels that are subtherapeutic should raise suspicion of intestinal malabsorption or poor compliance. Poor compliance might be more appropriately seen as a mimic of warfarin resistance. Studies in humans suggest that a therapeutic total plasma warfarin level lies between 0.5 μg/mL and 3.0 μg/mL,10 though the range may vary among laboratories and patient populations.

Warfarin absorption and clearance can be evaluated by analyzing plasma levels at specific intervals after administration, eg, every 60 to 180 minutes. The drug’s half-life can be determined on the basis of its concentrations in different time samples. Normally, the S-enantiomer of warfarin is cleared at twice the rate of the R-enantiomer (5.2 vs 2.5 mL/min/70 kg).8 A normal clearance rate confirms that resistance to warfarin is not due to enhanced elimination.

Clotting assays of factors II, VII, IX, and X may be a more precise way to assess the pharmacodynamics of warfarin,10 although there is no strong evidence to support routine use of such assays. Some studies suggest targeting factor II and factor X activity levels of 10% to 30% of normal biologic activity for a therapeutic warfarin effect in patients with an unreliable or prolonged baseline prothrombin time and INR, such as those with lupus anticoagulant.

Figure 2. Algorithm for evaluating suspected warfarin resistance.
An algorithm. Bentley et al33 suggest using the plasma warfarin level in an algorithm to determine the type of resistance pattern. Plasma warfarin levels are typically measured by regional specialized reference laboratories with a turnaround time of 2 to 7 days, as opposed to 24 hours for factor II and X activity. Our suggested algorithm for evaluation of suspected warfarin resistance is shown in Figure 2.

TREAT THE CAUSE

Once the type of warfarin resistance has been determined, treatment should be oriented toward the cause.

Educate the patient

The importance of compliance should be reinforced. Educating the patient about diet and other medications that may interact with warfarin is also important. (See an example of patient education material.)

Increase the warfarin dose

If the patient truly has hereditary resistance, there are two approaches to treatment.

The first is to increase the warfarin dose until the prothrombin time and INR are in the therapeutic ranges. When indicated, the warfarin dose can be safely titrated upward to more than 100 mg per day in patients who are monitored regularly—as all patients on chronic warfarin therapy should be—and whose other medications are otherwise stable. One such example is reported in a warfarinresistant patient who needed 145 mg/day to maintain a therapeutic prothrombin time.22

Try other anticoagulants?

The second approach is to change to another type of anticoagulant. However, there is no strong evidence in favor of this approach over prescribing larger dosages of warfarin.

Other anticoagulant drugs currently available in the United States include subcutaneous heparins (unfractionated and low-molecular-weight heparins) and the subcutaneous factor Xa inhibitor fondaparinux (Arixtra).

Agents not available in the United States include the following.

Dabigatran, an oral direct thrombin inhibitor, is undergoing phase 3 studies of its use for long-term anticoagulation.

Rivaroxaban (a direct factor Xa inhibitor) and dabigatran have been approved in Canada and the European Union to prevent venous thromboembolism after knee and hip arthroplasty, based on prospective comparisons with enoxaparin (Lovenox).34–37

Vitamin K antagonists other than warfarin that are not available in the United States include bishydroxycoumarin (which has limitations including slow absorption and high frequency of gastrointestinal side effects), phenprocoumon, and acenocoumarol. Another is phenindione, which has been associated with serious hypersensitivity reactions, some of which proved fatal and occurred within a few weeks of initiating therapy.

References
  1. Lefrere JJ, Horellou MH, Conard J, Samama M. Proposed classification of resistance to oral anticoagulant therapy. J Clin Pathol 1987; 40:242.
  2. Linder MW. Genetic mechanisms for hypersensitivity and resistance to the anticoagulant warfarin. Clin Chim Acta 2001; 308:915.
  3. Thijssen HH. Warfarin resistance. Vitamin K epoxide reductase of Scottish resistance gene is not irreversibly blocked by warfarin. Biochem Pharmacol 1987; 36:27532757.
  4. Hulse ML. Warfarin resistance: diagnosis and therapeutic alternative. Pharmacotherapy 1996; 16:10091017.
  5. Hirsh J, Dalen JE, Deykin D, Poller L, Bussey H. Oral anticoagulants. Mechanism of action, clinical effectiveness, and optimal therapeutic range. Chest 1995; 108( suppl 4):231S234S.
  6. Daly AK, King BP. Pharmacogenetics of oral anticoagulants. Pharmacogenetics 2003; 13:247252.
  7. Daly AK, Aithal GP. Genetic regulation of warfarin metabolism and response. Semin Vasc Med 2003; 3:231238.
  8. Takahashi H, Echizen H. Pharmacogenetics of warfarin elimination and its clinical implications. Clin Pharmacokinet 2001; 40:587603.
  9. Retti AE, Wienkers LC, Gonzalez FJ, Trager WF, Korezekwa KR. Impaired (S)-warfarin metabolism catalysed by the R144C allele variant of CYP2C9. Pharmacogenetics 1994; 4:3942.
  10. Porter RS, Sawyer WR. Warfarin. In:Evans WE, Shentag JJ, Jusko WJ, editors. Applied Pharmacokinetics. Principles of Therapeutics Drug Monitoring, 3rd ed. Washington, DC: Applied Therapeutics, 1992: 31.131.46.
  11. Warrell DA, Cox TM, Firth JD. Oxford Textbook of Medicine, 4th ed. Oxford University Press, 2003:734.
  12. Holbrook AM, Pereira JA, Labiris R, et al. Systematic overview of warfarin and its drug and food interactions. Arch Intern Med 2005; 165:10951106.
  13. Medical Economics Staff. Physicians’ Desk Reference, 55th Ed. Medical Economics, 2001:11391140.
  14. Schwarz UI, Ritchie MD, Bradford Y, et al. Genetic determinants of response to warfarin during initial anticoagulation. N Engl J Med 2008; 358:9991008.
  15. Diab F, Feffer S. Hereditary warfarin resistance. South Med J 1994; 87:407409.
  16. O’Reilly RA. The second reported kindred with hereditary resistance to oral anticoagulant drugs. N Engl J Med 1970; 282:14481451.
  17. O’Reilly RA, Aggeler PM, Hoag MS, Leong LS, Kropatkin ML. Hereditary transmission of exceptional resistance to coumarin anticoagulant drugs. The first reported kindred. N Engl J Med 1964; 271:809815.
  18. Alving BM, Strickler MP, Knight RD, Barr CF, Berenberg JL, Peek CC. Hereditary warfarin resistance. Investigation of rare phenomenon. Arch Intern Med 1985; 145:499501.
  19. Warrier L, Brennan CA, Lusher JM. Familial warfarin resistance in a black child. Am J Pediatr Hematol Oncol 1986; 8:346347.
  20. Nikkila EA, Pelkonen R. Serum lipid-reducing agents and anticoagulant requirement. Lancet 1963; 1:332.
  21. Robinson A, Liau FO, Routledge PA, Backhouse G, Spragg BP, Bentley DP. Lipids and warfarin requirements. Thromb Haemost 1990; 63:148149.
  22. MacLaren R, Wachsman BA, Swift DK, Kuhl DA. Warfarin resistance associated with intravenous lipid administration: discussion of propofol and review of the literature. Pharmacotherapy 1997; 17:13311337.
  23. DeCurtis A, D’Adamo MC, Amore C, et al. Experimental arterial thrombosis in genetically or diet induced hyperlipidemia in rats—role of vitamin K-dependent clotting factors and prevention by low-intensity oral anticoagulation. Thromb Haemost 2001; 86:14401448.
  24. O’Reilly RA. Drug interaction involving oral anticoagulation. In:Melmon KL, editor. Cardiovascular Drug Therapy, Philadelphia; FA Davis, 1975:2341.
  25. O’ Reilly RA, Pool JG, Aggeler PM. Hereditary resistance to coumarin anticoagulation drugs in man and rat. Ann N Y Acad Sci 1968; 151:913931.
  26. Cain D, Hutson SM, Wallin R. Warfarin resistance is associated with a protein component of the vitamin K 2,3-epoxide reductase enzyme complex in rat liver. Thromb Haemost 1998; 80:128133.
  27. Rodvold KA, Quandt CM, Friedenberg WR. Thromboembolic disorders. In:DiPiro JT, Talbert RL, editors. Pharmacotherapy. A Pathophysiologic Approach, 2nd ed. New York: Elsevier, 1992:312335.
  28. Park BK. Warfarin: metabolism and mode of action. Biochem Pharmacol 1988; 37:1927.
  29. Cain D, Hutson SM, Wallin R. Assembly of the warfarin-sensitive vitamin K 2,3-epoxide reductase enzyme complex in the endoplasmic reticulum membrane. J Biol Chem 1997; 272:2906829075.
  30. Gallop PM, Lian JB, Hauschka PV. Carboxylated calcium binding proteins and vitamin K. N Engl J Med 1980; 302:14601466.
  31. Rost S, Fregin A, Ivaskevicius V, et al. Mutations in VKORC1 cause warfarin resistance and multiple coagulation factor deficiency type 2. Nature 2004; 427:537541.
  32. Loebstein R, Dovskin I, Halkin H, et al. A coding VKORC1 Asp36-Tyr polymorphism predisposes to warfarin resistance. Blood 2007; 109:24772480.
  33. Bentley DP, Backhouse G, Hutchings A, Haddon RL, Spragg B, Routledge PA. Investigation of patients with abnormal response to warfarin. Br J Clin Pharmacol 1986; 22:3741.
  34. Eriksson BI, Borris LC, Friedman RJ, et al. RECORD1 Study Group. Rivaroxaban versus enoxaparin for thromboprophylaxis after hip arthroplasty. N Engl J Med 2008; 358:27652775.
  35. Kakkar AK, Brenner B, Dahl OE, et al; RECORD2 Investigators. Extended duration rivaroxaban versus short-term enoxaparin for the prevention of venous thromboembolism after total hip arthroplasty: a double-blind, randomised controlled trial. Lancet 2008; 372:3139.
  36. Lassen MR, Ageno W, Borris LC, et al; RECORD3 Investigators. Rivaroxaban versus enoxaparin for thromboprophylaxis after total knee arthroplasty. N Engl J Med 2008; 358:27762786.
  37. Wolowacz SE, Roskell NS, Plumb JM, Caprini JA, Eriksson BI. Efficacy and safety of dabigatran etexilate for the prevention of venous thromboembolism following total hip or knee arthroplasty. A meta-analysis. Thromb Haemost 2009; 101:7785.
References
  1. Lefrere JJ, Horellou MH, Conard J, Samama M. Proposed classification of resistance to oral anticoagulant therapy. J Clin Pathol 1987; 40:242.
  2. Linder MW. Genetic mechanisms for hypersensitivity and resistance to the anticoagulant warfarin. Clin Chim Acta 2001; 308:915.
  3. Thijssen HH. Warfarin resistance. Vitamin K epoxide reductase of Scottish resistance gene is not irreversibly blocked by warfarin. Biochem Pharmacol 1987; 36:27532757.
  4. Hulse ML. Warfarin resistance: diagnosis and therapeutic alternative. Pharmacotherapy 1996; 16:10091017.
  5. Hirsh J, Dalen JE, Deykin D, Poller L, Bussey H. Oral anticoagulants. Mechanism of action, clinical effectiveness, and optimal therapeutic range. Chest 1995; 108( suppl 4):231S234S.
  6. Daly AK, King BP. Pharmacogenetics of oral anticoagulants. Pharmacogenetics 2003; 13:247252.
  7. Daly AK, Aithal GP. Genetic regulation of warfarin metabolism and response. Semin Vasc Med 2003; 3:231238.
  8. Takahashi H, Echizen H. Pharmacogenetics of warfarin elimination and its clinical implications. Clin Pharmacokinet 2001; 40:587603.
  9. Retti AE, Wienkers LC, Gonzalez FJ, Trager WF, Korezekwa KR. Impaired (S)-warfarin metabolism catalysed by the R144C allele variant of CYP2C9. Pharmacogenetics 1994; 4:3942.
  10. Porter RS, Sawyer WR. Warfarin. In:Evans WE, Shentag JJ, Jusko WJ, editors. Applied Pharmacokinetics. Principles of Therapeutics Drug Monitoring, 3rd ed. Washington, DC: Applied Therapeutics, 1992: 31.131.46.
  11. Warrell DA, Cox TM, Firth JD. Oxford Textbook of Medicine, 4th ed. Oxford University Press, 2003:734.
  12. Holbrook AM, Pereira JA, Labiris R, et al. Systematic overview of warfarin and its drug and food interactions. Arch Intern Med 2005; 165:10951106.
  13. Medical Economics Staff. Physicians’ Desk Reference, 55th Ed. Medical Economics, 2001:11391140.
  14. Schwarz UI, Ritchie MD, Bradford Y, et al. Genetic determinants of response to warfarin during initial anticoagulation. N Engl J Med 2008; 358:9991008.
  15. Diab F, Feffer S. Hereditary warfarin resistance. South Med J 1994; 87:407409.
  16. O’Reilly RA. The second reported kindred with hereditary resistance to oral anticoagulant drugs. N Engl J Med 1970; 282:14481451.
  17. O’Reilly RA, Aggeler PM, Hoag MS, Leong LS, Kropatkin ML. Hereditary transmission of exceptional resistance to coumarin anticoagulant drugs. The first reported kindred. N Engl J Med 1964; 271:809815.
  18. Alving BM, Strickler MP, Knight RD, Barr CF, Berenberg JL, Peek CC. Hereditary warfarin resistance. Investigation of rare phenomenon. Arch Intern Med 1985; 145:499501.
  19. Warrier L, Brennan CA, Lusher JM. Familial warfarin resistance in a black child. Am J Pediatr Hematol Oncol 1986; 8:346347.
  20. Nikkila EA, Pelkonen R. Serum lipid-reducing agents and anticoagulant requirement. Lancet 1963; 1:332.
  21. Robinson A, Liau FO, Routledge PA, Backhouse G, Spragg BP, Bentley DP. Lipids and warfarin requirements. Thromb Haemost 1990; 63:148149.
  22. MacLaren R, Wachsman BA, Swift DK, Kuhl DA. Warfarin resistance associated with intravenous lipid administration: discussion of propofol and review of the literature. Pharmacotherapy 1997; 17:13311337.
  23. DeCurtis A, D’Adamo MC, Amore C, et al. Experimental arterial thrombosis in genetically or diet induced hyperlipidemia in rats—role of vitamin K-dependent clotting factors and prevention by low-intensity oral anticoagulation. Thromb Haemost 2001; 86:14401448.
  24. O’Reilly RA. Drug interaction involving oral anticoagulation. In:Melmon KL, editor. Cardiovascular Drug Therapy, Philadelphia; FA Davis, 1975:2341.
  25. O’ Reilly RA, Pool JG, Aggeler PM. Hereditary resistance to coumarin anticoagulation drugs in man and rat. Ann N Y Acad Sci 1968; 151:913931.
  26. Cain D, Hutson SM, Wallin R. Warfarin resistance is associated with a protein component of the vitamin K 2,3-epoxide reductase enzyme complex in rat liver. Thromb Haemost 1998; 80:128133.
  27. Rodvold KA, Quandt CM, Friedenberg WR. Thromboembolic disorders. In:DiPiro JT, Talbert RL, editors. Pharmacotherapy. A Pathophysiologic Approach, 2nd ed. New York: Elsevier, 1992:312335.
  28. Park BK. Warfarin: metabolism and mode of action. Biochem Pharmacol 1988; 37:1927.
  29. Cain D, Hutson SM, Wallin R. Assembly of the warfarin-sensitive vitamin K 2,3-epoxide reductase enzyme complex in the endoplasmic reticulum membrane. J Biol Chem 1997; 272:2906829075.
  30. Gallop PM, Lian JB, Hauschka PV. Carboxylated calcium binding proteins and vitamin K. N Engl J Med 1980; 302:14601466.
  31. Rost S, Fregin A, Ivaskevicius V, et al. Mutations in VKORC1 cause warfarin resistance and multiple coagulation factor deficiency type 2. Nature 2004; 427:537541.
  32. Loebstein R, Dovskin I, Halkin H, et al. A coding VKORC1 Asp36-Tyr polymorphism predisposes to warfarin resistance. Blood 2007; 109:24772480.
  33. Bentley DP, Backhouse G, Hutchings A, Haddon RL, Spragg B, Routledge PA. Investigation of patients with abnormal response to warfarin. Br J Clin Pharmacol 1986; 22:3741.
  34. Eriksson BI, Borris LC, Friedman RJ, et al. RECORD1 Study Group. Rivaroxaban versus enoxaparin for thromboprophylaxis after hip arthroplasty. N Engl J Med 2008; 358:27652775.
  35. Kakkar AK, Brenner B, Dahl OE, et al; RECORD2 Investigators. Extended duration rivaroxaban versus short-term enoxaparin for the prevention of venous thromboembolism after total hip arthroplasty: a double-blind, randomised controlled trial. Lancet 2008; 372:3139.
  36. Lassen MR, Ageno W, Borris LC, et al; RECORD3 Investigators. Rivaroxaban versus enoxaparin for thromboprophylaxis after total knee arthroplasty. N Engl J Med 2008; 358:27762786.
  37. Wolowacz SE, Roskell NS, Plumb JM, Caprini JA, Eriksson BI. Efficacy and safety of dabigatran etexilate for the prevention of venous thromboembolism following total hip or knee arthroplasty. A meta-analysis. Thromb Haemost 2009; 101:7785.
Issue
Cleveland Clinic Journal of Medicine - 76(12)
Issue
Cleveland Clinic Journal of Medicine - 76(12)
Page Number
724-730
Page Number
724-730
Publications
Publications
Topics
Article Type
Display Headline
An algorithm for managing warfarin resistance
Display Headline
An algorithm for managing warfarin resistance
Sections
Inside the Article

KEY POINTS

  • The most common cause of warfarin resistance is noncompliance. Others include poor absorption, high vitamin K intake, hypersensitivity to vitamin K, and rapid drug deactivation.
  • Patient education is necessary to improve compliance and to mitigate adverse effects of warfarin therapy, regardless of the dose.
  • In time, it may be possible to individualize anticoagulant dosing on the basis of genetic testing for patients with warfarin resistance, although currently such tests are not routinely advocated and are usually done only in specialized laboratories.
  • In true hereditary warfarin resistance, there are two approaches to treatment: increase the warfarin dosage (perhaps to as high as 100 mg/day or more), or switch to another anticoagulant.
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media

Prasugrel for acute coronary syndromes: Faster, more potent, but higher bleeding risk

Article Type
Changed
Thu, 04/12/2018 - 11:30
Display Headline
Prasugrel for acute coronary syndromes: Faster, more potent, but higher bleeding risk

Prasugrel (Effient) is more potent and consistent in its effects than clopidogrel (Plavix), thus preventing more thrombotic events—but at a price of more bleeding. Therefore, the drugs must be appropriately selected for the individual patient.

Over the last 9 years, the thienopyridines—ticlopidine (Ticlid), clopidogrel, and now prasugrel—have become essential tools for treating acute coronary syndromes.

The usual underlying mechanism of acute coronary syndromes is thrombosis, caused by rupture of atherosclerotic plaque.1 Accordingly, antithrombotic agents—aspirin, heparin, lowmolecular-weight heparin, glycoprotein IIb/IIIa inhibitors, the direct thrombin inhibitor bivalirudin (Angiomax), and thienopyridines—have all been shown to reduce the risk of major adverse cardiac outcomes in this setting.

In this article, we review the pharmacology and evidence of effectiveness of the thienopyridine drugs, focusing on prasugrel, the latest thienopyridine to be approved by the US Food and Drug Administration (FDA).

THIENOPYRIDINES INHIBIT PLATELET ACTIVATION AND AGGREGATION

Thienopyridines are prodrugs that require conversion by hepatic cytochrome P450 enzymes. The active metabolites bind irreversibly to platelet P2Y12 receptors. Consequently, they permanently block signalling mediated by platelet adenosine diphosphate-P2Y12 receptors, thereby inhibiting glycoprotein IIb/IIIa receptor activation and platelet aggregation.

Aspirin, in contrast, inhibits platelets by blocking the thromboxane-mediated pathway. Therefore, the combination of aspirin plus a thienopyridine has an additive effect.2

The effect of thienopyridines on platelets is irreversible. Therefore, although the half-life of prasugrel’s active metabolite is 3.7 hours, its inhibitory effects last for 96 hours, essentially the time for half the body’s circulating platelets to be replaced.

TICLOPIDINE, THE FIRST THIENOPYRIDINE

Ticlopidine was the first thienopyridine to be approved by the FDA. Its initial studies in unstable angina were small, their designs did not call for patients to concurrently receive aspirin, and all they showed was that ticlopidine was about as beneficial as aspirin. Consequently, the studies had little impact on clinical practice.3

In a pivotal trial,4 patients who received coronary stents were randomized to afterward receive either the combination of ticlopidine plus aspirin or anticoagulation therapy with heparin, phenprocoumon (a coumarin derivative available in Europe), and aspirin. At 30 days, an ischemic complication (death, myocardial infarction [MI], repeat intervention) had occurred in 6.2% of the anticoagulation therapy group vs 1.6% of the ticlopidine group, a risk reduction of 75%. Rates of stent occlusion, MI, and revascularization were 80% to 85% lower in the ticlodipine group. This study paved the way for widespread use of thienopyridines.

Ticlopidine’s use was limited, however, by a 2.4% incidence of serious granulocytopenia and rare cases of thrombocytopenic purpura.

BENEFIT OF CLOPIDOGREL

Although prasugrel is the focus of this review, the trials of prasugrel all compared its efficacy with that of clopidogrel. Furthermore, many patients should still receive clopidogrel and not prasugrel, so it is important to be familiar with the evidence of clopidogrel’s benefit.

Once approved for clinical use, clopidogrel was substituted for ticlopidine in patients undergoing coronary stenting on the basis of studies showing it to be at least as effective as ticlopidine and more tolerable. A series of trials of clopidogrel were done in patients across a spectrum of risk groups, from those at high risk of coronary heart disease to those presenting with ST-elevation MI. The time of pretreatment in the studies ranged from 3 hours to 6 days before percutaneous coronary intervention, and the duration of treatment following intervention ranged from 30 days to 1 year.

Clopidogrel in non-ST-elevation acute coronary syndromes

The CURE trial2 (Clopidogrel in Unstable Angina to Prevent Recurrent Events), published in 2001, established clopidogrel as a therapy for unstable ischemic syndromes, whether treated medically or with revascularization. In that trial, 12,562 patients with acute coronary syndromes without ST elevation (ie, unstable angina or non-ST-elevation MI), as defined by electrocardiographic changes or positive cardiac markers, were randomized to receive clopidogrel (a 300-mg loading dose followed by 75-mg maintenance doses) or placebo for a mean duration of 9 months. All patients also received aspirin 75 mg to 325 mg daily.

The composite outcome of death from cardiovascular causes, nonfatal MI, or stroke occurred in 20% fewer patients treated with clopidogrel than with placebo (9.3% vs 11.4%). The benefit was similar in patients undergoing revascularization compared with those treated medically.

Although there were significantly more cases of major bleeding in the clopidogrel group than in the placebo group (3.7% vs 2.7%), the number of episodes of life-threatening bleeding or hemorrhagic strokes was the same.

PCI-CURE5 was a substudy of the CURE trial in patients who underwent a percutaneous coronary intervention. Patients were pretreated with clopidogrel or placebo for a mean of 6 days before the procedure. Afterward, they all received clopidogrel plus aspirin in an unblinded fashion for 2 to 4 weeks, and then the randomized study drug was resumed for a mean of 8 months.

Significantly fewer adverse events occurred in the clopidogrel group as tallied at the time of the intervention, 1 month later, and 8 months later.

 

 

Clopidogrel in ST-elevation acute MI

The CLARITY-TIMI 28 trial6 (Clopidogrel as Adjunctive Reperfusion Therapy—Thrombolysis in Myocardial Infarction 28) showed that adding clopidogrel (a 300-mg loading dose, then 75 mg daily) to aspirin benefitted patients with ST-elevation MI receiving fibrinolytic therapy. At 30 days, cardiovascular death, recurrent MI, or urgent revascularization had occurred in 11.6% of the clopidogrel group vs 14.1% of the placebo group, a statistically significant difference. The rates of major or minor bleeding were no higher in the clopidogrel group than in the placebo group, an especially remarkable finding in patients receiving thrombolytic therapy.

PCI-CLARITY.7 About half of the patients in the CLARITY trial ultimately underwent a percutaneous coronary intervention after fibrinolytic therapy, with results reported as the PCI-CLARITY substudy. Like those in PCI-CURE, these patients were randomized to receive pretreatment with either clopidogrel or placebo before the procedure, in this study for a median of 3 days. Both groups received clopidogrel afterward. At 30 days from randomization, the outcome of cardiovascular death, MI, or stroke had occurred in 7.5% of the clopidogrel group compared with 12.0% of the placebo group, which was statistically significant, without any significant excess in the rates of major or minor bleeding.

COMMIT8 (the Clopidogrel and Metoprolol in Myocardial Infarction Trial) also showed clopidogrel to be beneficial in patients with acute MI. This trial included more than 45,000 patients in China with acute MI, 93% of whom had ST-segment elevation. In contrast to CLARITY, in COMMIT barely more than half of the patients received fibrinolysis, fewer than 5% proceeded to percutaneous interventions, and no loading dose was given: patients in the clopidogrel group received 75 mg/day from the outset.

At 15 days, the incidence of death, reinfarction, or stroke was 9.2% with clopidogrel compared with 10.1% with placebo, a small but statistically significant difference. Again, the rate of major bleeding was not significantly higher, either overall or in patients over age 70.

Of note, patients over age 75 were excluded from CLARITY, and as mentioned, no loading dose was used in COMMIT. Thus, for patients receiving fibrinolysis who are over age 75, there is no evidence to support the safety of a loading dose, and clopidogrel should be started at 75 mg daily.

Clopidogrel in elective percutaneous coronary intervention

The CREDO trial9 (Clopidogrel for the Reduction of Events During Observation) was in patients referred for elective percutaneous coronary intervention. Three to 24 hours before the procedure, the patients received either a 300-mg loading dose of clopidogrel or placebo; afterward, all patients received clopidogrel 75 mg/day for 28 days. All patients also received aspirin.

A clopidogrel loading dose 3 to 24 hours before the intervention did not produce a statistically significant reduction in ischemic events, although a post hoc subgroup analysis suggested that patients who received the loading dose between 6 and 24 hours before did benefit, with a relative risk reduction of 38.6% in the composite end point (P = .051).

After 28 days, the patients who had received the clopidogrel loading dose were continued on clopidogrel, while those in the placebo group were switched back to placebo. At 1 year, the investigators found a significantly lower rate of the composite end point with the prolonged course of clopidogrel (8.5% vs 11.5%).

In summary, these studies found clopidogrel to be beneficial in a broad spectrum of coronary diseases. Subgroup analyses suggest that pretreatment before percutaneous coronary intervention provides additional benefit, particularly if clopidogrel is given at least 6 hours in advance (the time necessary for clopidogrel to cause substantial platelet inhibition).

SOME PATIENTS RESPOND LESS TO CLOPIDOGREL

The level of platelet inhibition induced by clopidogrel varies. In different studies, the frequency of clopidogrel “nonresponsiveness” ranged from 5% to 56% of patients, depending on which test and which cutoff values were used. The distribution of responses to clopidogrel is wide and fits a normal gaussian curve.10

A large fraction of the population carries a gene that may account for some of the interpatient variation in platelet inhibition with clopidogrel. Carriers of a reduced-function CYP2C19 allele—approximately 30% of people in one study—have significantly lower levels of the active metabolite of clopidogrel, less platelet inhibition from clopidogrel therapy, and a 53% higher rate of death from cardiovascular causes, MI, or stroke.11

 

 

PRASUGREL, THE NEWEST THIENOPYRIDINE

Prasugrel, FDA-approved in July 2009 for the treatment of acute coronary syndromes, is given in an oral loading dose of 60 mg followed by an oral maintenance dose of 10 mg daily.

Pharmacology of prasugrel vs clopidogrel

As noted previously, the thienopyridines are prodrugs that require hepatic conversion to exert antiplatelet effects.

Metabolism. Prasugrel’s hepatic activation involves a single step, in contrast to the multiple-step process required for activation of clopidogrel. Clopidogrel is primarily hydrolyzed by intestinal and plasma esterases to an inactive terminal metabolite, with the residual unhydrolized drug undergoing a two-step metabolism that depends on cytochrome P450 enzymes. Prasugrel is also extensively hydrolyzed by these esterases, but the intermediate product is then metabolized in a single step to the active sulfhydryl compound, mainly by CYP3A4 and CYP2B6.

Thus, about 80% of an orally absorbed dose of prasugrel is converted to active drug, compared with only 10% to 20% of absorbed clopidogrel.

Time to peak effect. With clopidogrel, maximal inhibition of platelet aggregation occurs 3 to 5 days after starting therapy with 75 mg daily without a loading dose, but within 4 to 6 hours if a loading dose of 300 to 600 mg is given. In contrast, a prasugrel loading dose produces more than 80% of its platelet inhibitory effects by 30 minutes, and peak activity is observed within 4 hours.12 The platelet inhibition induced by prasugrel at 30 minutes after administration is comparable to the peak effect of clopidogrel at 6 hours.13

Dose-response. Prasugrel’s inhibition of platelet aggregation is dose-related.

Prasugrel is about 10 times more potent than clopidogrel and 100 times more potent than ticlopidine. Thus, treatment with 5 mg of prasugrel results in inhibition of platelet activity (distributed in a gaussian curve) very similar to that produced by 75 mg of clopidogrel. On the other hand, even a maintenance dose of 150 mg of clopidogrel inhibits platelet activity to a lesser degree than 10 mg of prasugrel (46% vs 61%),14 so clopidogrel appears to reach a plateau of platelet inhibition that prasugrel can overcome.

At the approved dose of prasugrel, inhibition of platelet aggregation is significantly greater and there are fewer “nonresponders” than with clopidogrel.

Interactions. Drugs that inhibit CYP3A4 do not inhibit the efficacy of prasugrel, but they can inhibit that of clopidogrel. Some commonly used drugs that have this effect are the statins (eg, atorvastain [Lipitor]) and the macrolide antibiotics (eg, erythromycin). Furthermore, whereas proton pump inhibitors have been shown to diminish the effect of clopidogrel by reducing the formation of its active metabolite, no such effect has been noted with prasugrel.

Prasugrel in phase 2 trials: Finding the optimal dosage

A phase 2 trial compared three prasugrel regimens (loading dose/daily maintenance dose of 40 mg/7.5 mg, 60 mg/10 mg, and 60 mg/15 mg) and standard clopidogrel therapy (300 mg/75 mg) in patients undergoing elective or urgent percutaneous coronary intervention.15 No significant difference in outcomes was seen in the groups receiving the three prasugrel regimens. However, more “minimal bleeding events” (defined by the criteria of the TIMI trial16) occurred with high-dose prasugrel than with lower-dose prasugrel or with clopidogrel, leading to use of the intermediate-dose prasugrel regimen (60-mg loading dose, 10-mg daily maintenance) for later trials.

Another phase 2 trial randomized 201 patients undergoing elective percutaneous coronary intervention to receive prasugrel 60 mg/10 mg or clopidogrel 600 mg/150 mg.14 In all patients, the loading dose was given about 1 hour before cardiac catheterization. As soon as 30 minutes after the loading dose, platelet inhibition was superior with prasugrel (31% vs 5% inhibition of platelet aggregation), and it remained significantly higher at 6 hours (75% vs 32%) and during the maintenance phase (61% vs 46%).

 

 

Phase 3 trial of prasugrel vs clopidogrel: TRITON-TIMI 38

Only one large phase 3 trial of prasugrel has been completed: TRITON-TIMI 38 (the Trial to Assess Improvement in Therapeutic Outcomes by Optimizing Platelet Inhibition With Prasugrel—Thrombolysis in Myocardial Infarction),17 which enrolled adults with moderate-risk to high-risk acute coronary syndromes scheduled to undergo a percutaneous coronary intervention. In this trial, 10,074 patients were enrolled who had moderate-to high-risk unstable angina or non-ST-elevation MI, and 3,534 patients were enrolled who had ST-elevation MI.

Patients were randomized to receive prasugrel (a 60-mg loading dose, then 10 mg daily) or clopidogrel (a 300-mg loading dose, then 75 mg daily) and were treated for 6 to 15 months. All patients also received aspirin.

The primary end point, a composite of death from cardiovascular causes, nonfatal MI, or nonfatal stroke, occurred in significantly fewer patients treated with prasugrel than with clopidogrel (9.9% vs 12.1%, P < .001) (Table 1). Most of the benefit was due to fewer nonfatal MIs during the follow-up period (7.4% vs 9.7%, P < .001). Additionally, the prasugrel group had a significantly lower rate of stent thrombosis compared with the clopidogrel group (1.1% vs 2.4%; P < .001).

These benefits came at a price of more bleeding. Of those patients who did not undergo coronary artery bypass grafting, more experienced bleeding in the prasugrel group than in the clopidogrel group (2.4% vs 1.8%, P = .03), including a higher rate of life-threatening bleeding (1.4% vs 0.89%, P = .01) and fatal bleeding (0.4% vs 0.1%, P = .002). More patients discontinued prasugrel because of hemorrhage (2.5% vs 1.4%, P < .001). In patients who proceeded to coronary artery bypass grafting, the rate of major bleeding was more than four times higher in those who received prasugrel than in those who received clopidogrel (13.4% vs 3.2%, P < .001).

A higher rate of adverse events related to colon cancer was also noted in patients treated with prasugrel, although the authors suggest this may have resulted from the stronger antiplatelet effects of prasugrel bringing more tumors to medical attention due to bleeding.

Overall death rates did not differ significantly between the treatment groups.

In a post hoc analysis,18 prasugrel was superior to clopidogrel in preventing ischemic events both during the first 3 days following randomization (the “loading phase”) and for the remainder of the trial (the “maintenance phase”). Whereas bleeding risk was similar with the two drugs during the loading phase, prasugrel was subsequently associated with more bleeding during the maintenance phase.

Certain patient subgroups had no net benefit or even suffered harm from prasugrel compared with clopidogrel.17 Patients with previous stroke or transient ischemic attack had net harm from prasugrel (hazard ratio 1.54, P = .04) and showed a strong trend toward a greater rate of major bleeding (P = .06). Patients age 75 and older and those weighing less than 60 kg had no net benefit from prasugrel.

Cost of prasugrel

Prasugrel is currently priced at 18% more than clopidogrel, with average wholesale prices per pill of $6.65 for prasugrel 10 mg compared with $5.63 for clopidogrel 75 mg. (Prasugrel 10-mg pills cost $6.33 at drugstore.com or $7.60 at CVS; clopidogrel 75-mg pills cost $5.33 at drugstore.com or $6.43 at CVS.) The patent on clopidogrel expires in November 2011, after which the price differential is expected to become significantly greater.

TICAGRELOR, A REVERSIBLE ORAL AGENT

Ticagrelor, the first reversible oral P2Y12 receptor antagonist, is an alternative to thienopyridine therapy for acute coronary syndromes.

Ticagrelor is quickly absorbed, does not require metabolic activation, and has a rapid antiplatelet effect and offset of effect, which closely follow drug-exposure levels. In a large randomized controlled trial in patients with acute coronary syndromes with or without STsegment elevation, treatment with ticagrelor compared with clopidogrel resulted in a significant reduction in death from vascular causes, MI, or stroke (9.8% vs 11.7%).19

Given its reversible effect on platelet inhibition, ticagrelor may be preferred in patients whose coronary anatomy is unknown and for whom coronary artery bypass grafting is deemed probable. It is still undergoing trials and is not yet approved.

 

 

TAKE-HOME POINTS

Prasugrel is more potent, more rapid in onset, and more consistent in inhibiting platelet aggregation than clopidogrel. A large clinical trial17 found prasugrel to be superior to clopidogrel for patients with moderate-to high-risk acute coronary syndromes with high probability of undergoing a percutaneous coronary intervention.

Who should receive prasugrel, and how?

Prasugrel should be given after angiography to patients with non-ST-elevation acute coronary syndromes or at presentation to patients with ST-elevation MI. When used for planned percutaneous coronary intervention, prasugrel should be given at least 30 minutes before the intervention, as was done in phase 2 trials (although its routine use in this situation is not recommended—see below).

It is given in a one-time loading dose of 60 mg by mouth and then maintained with 10 mg by mouth once daily for at least 1 year. (At least 9 months of treatment with a thienopyridine is indicated for patients with acute coronary syndromes who are medically treated, and at least 1 year is indicated following urgent or elective percutaneous coronary intervention, including balloon angioplasty and placement of a bare-metal or drug-eluting stent.)

Who should not receive prasugrel?

For now, prasugrel should be avoided in favor of clopidogrel in patients at higher risk of bleeding. It is clearly contraindicated in patients with prior transient ischemic attack or stroke, for whom the risk of serious bleeding seems to be prohibitive. It should generally be avoided in patients age 75 and older, although it might be considered in those at particularly high risk of stent thrombosis, such as those with diabetes or prior MI. In patients weighing less than 60 kg, the package insert advises a reduced dose (5 mg), although clinical evidence for this practice is lacking.

As yet, we have no data assuring that prasugrel is safe to use in combination with fibrinolytic agents, so patients on thrombolytic therapy for acute MI should continue to receive clopidogrel starting immediately after lysis. Furthermore, in patients who proceeded to coronary artery bypass grafting, the rate of major bleeding was more than four times higher in the prasugrel group than in the clopidogrel group in the TRITON-TIMI 38 trial.17 No thienopyridine should be given to patients likely to proceed to coronary artery bypass grafting.

Only clopidogrel has evidence supporting its use as an alternative to aspirin for patients with atherosclerotic disease who cannot tolerate aspirin. Neither drug has evidence for use for primary prevention.

Other areas of uncertainty

Prior to angiography. Indications for prasugrel are currently limited by the narrow scope of the trial data. TRITON-TIMI 38,17 the only large trial completed to date, randomized patients to receive prasugrel only after their coronary anatomy was known, except for ST-elevation MI patients. It is unknown whether the benefits of prasugrel will outweigh the higher risk of bleeding in patients with acute coronary syndromes who do not proceed to percutaneous coronary interventions.

A clinical trial is currently under way comparing prasugrel with clopidogrel in 10,000 patients with acute coronary syndromes who will be medically managed without planned revascularization: A Comparison of Prasugrel and Clopidogrel in Acute Coronary Syndrome Subjects (TRILOGY ACS), ClinicalTrials.gov Identifier: NCT00699998. The trial has an estimated completion date of March 2011.

In cases of non-ST-elevation acute coronary syndrome, it is reasonable to wait to give a thienopyridine until after the coronary anatomy has been defined, if angiography will be completed soon after presentation. For example, a 1-hour delay before giving prasugrel still delivers antiplatelet therapy more quickly than giving clopidogrel on presentation. If longer delays are expected before angiography, however, the patient should be given a loading dose of clopidogrel “up front,” in accordance with guidelines published by the American College of Cardiology, American Heart Association, and European Society of Cardiology,20 which recommend starting a thienopyridine early during hospitalization based on trial data with clopidogrel.

Patients undergoing elective percutaneous coronary intervention are at lower risk of stent thrombosis and other ischemic complications, so it is possible that the benefits of prasugrel would not outweigh the risks in these patients. Thus, prasugrel cannot yet be recommended for routine elective percutaneous coronary intervention except in individual cases in which the interventionalist feels that the patient may be at higher risk of thrombosis.

References
  1. Yeghiazarians Y, Braunstein JB, Askari A, Stone PH. Unstable angina pectoris. N Engl J Med 2000; 342:101114.
  2. Yusuf S, Zhao F, Mehta SR, Chrolavicius S, Tognoni G, Fox KK; Clopidogrel in Unstable Angina to Prevent Recurrent Events Trial Investigators. Effects of clopidogrel in addition to aspirin in patients with acute coronary syndromes without ST-segment elevation. N Engl J Med 2001; 345:494502.
  3. Balsano F, Rizzon P, Violi F, et al. Antiplatelet treatment with ticlopidine in unstable angina. A controlled multicenter clinical trial. The Studio della Ticlopidina nell'Angina Instabile Group. Circulation 1990; 82:1726.
  4. Schömig A, Neumann FJ, Kastrati A, et al. A randomized comparison of antiplatelet and anticoagulant therapy after the placement of coronary-artery stents. N Engl J Med 1996; 334:10841089.
  5. Mehta SR, Yusuf S, Peters RJG, et al; Clopidogrel in Unstable Angina to Prevent Recurrent Events Trial (CURE) Investigators. Effects of pretreatment with clopidogrel and aspirin followed by long-term therapy in patients undergoing percutaneous coronary intervention: the PCI-CURE study. Lancet 2001; 358:527533.
  6. Sabatine MS, Cannon CP, Gibson CM, et al; CLA RITY-TIMI 28 Investigators. Addition of clopidogrel to aspirin and fibrinolytic therapy for myocardial infarction with STsegment elevation. N Engl J Med 2005; 352:11791189.
  7. Sabatine MS, Cannon CP, Gibson CM, et al; Clopidogrel as Adjunctive Reperfusion Therapy (CLARITY)-Thrombolysis in Myocardial Infarction (TIMI) 28 Investigators. Effect of clopidogrel pretreatment before percutaneous coronary intervention in patients with ST-elevation myocardial infarction treated with fibrinolytics: the PCI-CLARITY study. JAMA 2005: 294:12241232.
  8. Chen ZM, Jiang LX, Chen YP, et al; COMMIT (ClOpidogrel and Metoprolol in Myocardial Infarction Trial) collaborative group. Addition of clopidogrel to aspirin in 45,852 patients with acute myocardial infarction: randomised placebo-controlled trial. Lancet 2005; 366:16071621.
  9. Steinhubl SR, Berger PB, Mann JT, et al; CREDO Investigators. Clopidogrel for the reduction of events during observation. Early and sustained dual oral antiplatelet therapy following percutaneous coronary intervention: a randomized controlled trial. JAMA 2002; 288:24112420.
  10. Serebruany VL, Steinhubl SR, Berger PB, Malinin AI, Bhatt DL, Topol EJ. Variability in platelet responsiveness to clopidogrel among 544 individuals. J Am Coll Cardiol 2005; 45:246251.
  11. Mega JL, Close SL, Wiviott SD, et al. Cytochrome P-450 polymorphisms and response to clopidogrel. N Engl J Med 2009; 360:354362.
  12. Helft G, Osende JI, Worthley SG, et al. Acute antithrombotic effect of a front-loaded regimen of clopidogrel in patients with atherosclerosis on aspirin. Arterioscler Thromb Vasc Biol 2000; 20:23162321.
  13. Weerakkody GJ, Jakubowski JA, Brandt JT, et al. Comparison of speed of onset of platelet inhibition after loading doses of clopidogrel versus prasugrel in healthy volunteers and correlation with responder status. Am J Cardiol 2007; 100:331336.
  14. Wiviott SD, Trenk D, Frelinger AL, et al; PRINCIPLETIMI 44 Investigators. Prasugrel compared with high loading-and maintenance-dose clopidogrel in patients with planned percutaneous coronary intervention: the Prasugrel in Comparison to Clopidogrel for Inhibition of Platelet Activation and Aggregation-Thrombolysis in Myocardial Infarction 44 trial. Circulation 2007; 116:29232932.
  15. Wiviott SD, Antman EM, Winters KJ, et al; JUMBO-TIMI 26 Investigators. Randomized comparison of prasugrel (CS-747, LY640315), a novel thienopyridine P2Y12 antagonist, with clopidogrel in percutaneous coronary intervention: results of the Joint Utilization of Medications to Block Platelets Optimally (JUMBO)-TIMI 26 Trial. Circulation 2005; 111:33663373.
  16. Bovill EG, Terrin ML, Stump DC, et al. Hemorrhagic events during therapy with recombinant tissue-type plasminogen activator, heparin, and aspirin for acute myocardial infarction. Results of the Thrombolysis in Myocardial Infarction (TIMI) Phase II Trial. Ann Intern Med 1991; 115:256265.
  17. Wiviott SD, Braunwald E, McCabe CH, et al; TRITONTIMI 38 Investigators. Prasugrel versus clopidogrel in patients with acute coronary syndromes. N Engl J Med 2007; 357:20012015.
  18. Antman EM, Wiviott SD, Murphy SA, et al. Early and late benefits of prasugrel in patients with acute coronary syndromes undergoing percutaneous coronary intervention: a TRITON-TIMI 38 (TRial to Assess Improvement in Therapeutic Outcomes by Optimizing Platelet InhibitioN with Prasugrel-Thrombolysis In Myocardial Infarction) analysis. J Am Coll Cardiol 2008; 51:20282033.
  19. Wallentin L, Becker RC, Budaj A, Freij A, Thorsén M, et al; PLATO Investigators. Ticagrelor versus clopidogrel in patients with acute coronary syndromes. N Engl J Med 2009; 361:10451057.
  20. Braunwald E, Antman EM, Beasley JW, et al. ACC/AHA 2002 guideline update for the management of patients with unstable angina and non–ST-segment elevation myocardial infarction—summary article*1: A report of the American College of Cardiology/American Heart Association task force on practice guidelines (Committee on the Management of Patients With Unstable Angina). J Am Coll Cardiol 2002; 40:13661374.
Article PDF
Author and Disclosure Information

Lawrence D. Lazar, MD
Department of Cardiovascular Medicine, Cleveland Clinic

A. Michael Lincoff, MD
Professor of Medicine; Vice Chairman, Department of Cardiovascular Medicine; Director, Center for Clinical Research, Lerner Research Institute; Director, Cleveland Clinic Coordinating Center for Clinical Research, Department of Cardiovascular Medicine, Cleveland Clinic

Address: Lawrence D. Lazar, MD, Department of Cardiovascular Medicine, Cleveland Clinic, J2-3, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail [email protected]

Issue
Cleveland Clinic Journal of Medicine - 76(12)
Publications
Topics
Page Number
707-714
Sections
Author and Disclosure Information

Lawrence D. Lazar, MD
Department of Cardiovascular Medicine, Cleveland Clinic

A. Michael Lincoff, MD
Professor of Medicine; Vice Chairman, Department of Cardiovascular Medicine; Director, Center for Clinical Research, Lerner Research Institute; Director, Cleveland Clinic Coordinating Center for Clinical Research, Department of Cardiovascular Medicine, Cleveland Clinic

Address: Lawrence D. Lazar, MD, Department of Cardiovascular Medicine, Cleveland Clinic, J2-3, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail [email protected]

Author and Disclosure Information

Lawrence D. Lazar, MD
Department of Cardiovascular Medicine, Cleveland Clinic

A. Michael Lincoff, MD
Professor of Medicine; Vice Chairman, Department of Cardiovascular Medicine; Director, Center for Clinical Research, Lerner Research Institute; Director, Cleveland Clinic Coordinating Center for Clinical Research, Department of Cardiovascular Medicine, Cleveland Clinic

Address: Lawrence D. Lazar, MD, Department of Cardiovascular Medicine, Cleveland Clinic, J2-3, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail [email protected]

Article PDF
Article PDF

Prasugrel (Effient) is more potent and consistent in its effects than clopidogrel (Plavix), thus preventing more thrombotic events—but at a price of more bleeding. Therefore, the drugs must be appropriately selected for the individual patient.

Over the last 9 years, the thienopyridines—ticlopidine (Ticlid), clopidogrel, and now prasugrel—have become essential tools for treating acute coronary syndromes.

The usual underlying mechanism of acute coronary syndromes is thrombosis, caused by rupture of atherosclerotic plaque.1 Accordingly, antithrombotic agents—aspirin, heparin, lowmolecular-weight heparin, glycoprotein IIb/IIIa inhibitors, the direct thrombin inhibitor bivalirudin (Angiomax), and thienopyridines—have all been shown to reduce the risk of major adverse cardiac outcomes in this setting.

In this article, we review the pharmacology and evidence of effectiveness of the thienopyridine drugs, focusing on prasugrel, the latest thienopyridine to be approved by the US Food and Drug Administration (FDA).

THIENOPYRIDINES INHIBIT PLATELET ACTIVATION AND AGGREGATION

Thienopyridines are prodrugs that require conversion by hepatic cytochrome P450 enzymes. The active metabolites bind irreversibly to platelet P2Y12 receptors. Consequently, they permanently block signalling mediated by platelet adenosine diphosphate-P2Y12 receptors, thereby inhibiting glycoprotein IIb/IIIa receptor activation and platelet aggregation.

Aspirin, in contrast, inhibits platelets by blocking the thromboxane-mediated pathway. Therefore, the combination of aspirin plus a thienopyridine has an additive effect.2

The effect of thienopyridines on platelets is irreversible. Therefore, although the half-life of prasugrel’s active metabolite is 3.7 hours, its inhibitory effects last for 96 hours, essentially the time for half the body’s circulating platelets to be replaced.

TICLOPIDINE, THE FIRST THIENOPYRIDINE

Ticlopidine was the first thienopyridine to be approved by the FDA. Its initial studies in unstable angina were small, their designs did not call for patients to concurrently receive aspirin, and all they showed was that ticlopidine was about as beneficial as aspirin. Consequently, the studies had little impact on clinical practice.3

In a pivotal trial,4 patients who received coronary stents were randomized to afterward receive either the combination of ticlopidine plus aspirin or anticoagulation therapy with heparin, phenprocoumon (a coumarin derivative available in Europe), and aspirin. At 30 days, an ischemic complication (death, myocardial infarction [MI], repeat intervention) had occurred in 6.2% of the anticoagulation therapy group vs 1.6% of the ticlopidine group, a risk reduction of 75%. Rates of stent occlusion, MI, and revascularization were 80% to 85% lower in the ticlodipine group. This study paved the way for widespread use of thienopyridines.

Ticlopidine’s use was limited, however, by a 2.4% incidence of serious granulocytopenia and rare cases of thrombocytopenic purpura.

BENEFIT OF CLOPIDOGREL

Although prasugrel is the focus of this review, the trials of prasugrel all compared its efficacy with that of clopidogrel. Furthermore, many patients should still receive clopidogrel and not prasugrel, so it is important to be familiar with the evidence of clopidogrel’s benefit.

Once approved for clinical use, clopidogrel was substituted for ticlopidine in patients undergoing coronary stenting on the basis of studies showing it to be at least as effective as ticlopidine and more tolerable. A series of trials of clopidogrel were done in patients across a spectrum of risk groups, from those at high risk of coronary heart disease to those presenting with ST-elevation MI. The time of pretreatment in the studies ranged from 3 hours to 6 days before percutaneous coronary intervention, and the duration of treatment following intervention ranged from 30 days to 1 year.

Clopidogrel in non-ST-elevation acute coronary syndromes

The CURE trial2 (Clopidogrel in Unstable Angina to Prevent Recurrent Events), published in 2001, established clopidogrel as a therapy for unstable ischemic syndromes, whether treated medically or with revascularization. In that trial, 12,562 patients with acute coronary syndromes without ST elevation (ie, unstable angina or non-ST-elevation MI), as defined by electrocardiographic changes or positive cardiac markers, were randomized to receive clopidogrel (a 300-mg loading dose followed by 75-mg maintenance doses) or placebo for a mean duration of 9 months. All patients also received aspirin 75 mg to 325 mg daily.

The composite outcome of death from cardiovascular causes, nonfatal MI, or stroke occurred in 20% fewer patients treated with clopidogrel than with placebo (9.3% vs 11.4%). The benefit was similar in patients undergoing revascularization compared with those treated medically.

Although there were significantly more cases of major bleeding in the clopidogrel group than in the placebo group (3.7% vs 2.7%), the number of episodes of life-threatening bleeding or hemorrhagic strokes was the same.

PCI-CURE5 was a substudy of the CURE trial in patients who underwent a percutaneous coronary intervention. Patients were pretreated with clopidogrel or placebo for a mean of 6 days before the procedure. Afterward, they all received clopidogrel plus aspirin in an unblinded fashion for 2 to 4 weeks, and then the randomized study drug was resumed for a mean of 8 months.

Significantly fewer adverse events occurred in the clopidogrel group as tallied at the time of the intervention, 1 month later, and 8 months later.

 

 

Clopidogrel in ST-elevation acute MI

The CLARITY-TIMI 28 trial6 (Clopidogrel as Adjunctive Reperfusion Therapy—Thrombolysis in Myocardial Infarction 28) showed that adding clopidogrel (a 300-mg loading dose, then 75 mg daily) to aspirin benefitted patients with ST-elevation MI receiving fibrinolytic therapy. At 30 days, cardiovascular death, recurrent MI, or urgent revascularization had occurred in 11.6% of the clopidogrel group vs 14.1% of the placebo group, a statistically significant difference. The rates of major or minor bleeding were no higher in the clopidogrel group than in the placebo group, an especially remarkable finding in patients receiving thrombolytic therapy.

PCI-CLARITY.7 About half of the patients in the CLARITY trial ultimately underwent a percutaneous coronary intervention after fibrinolytic therapy, with results reported as the PCI-CLARITY substudy. Like those in PCI-CURE, these patients were randomized to receive pretreatment with either clopidogrel or placebo before the procedure, in this study for a median of 3 days. Both groups received clopidogrel afterward. At 30 days from randomization, the outcome of cardiovascular death, MI, or stroke had occurred in 7.5% of the clopidogrel group compared with 12.0% of the placebo group, which was statistically significant, without any significant excess in the rates of major or minor bleeding.

COMMIT8 (the Clopidogrel and Metoprolol in Myocardial Infarction Trial) also showed clopidogrel to be beneficial in patients with acute MI. This trial included more than 45,000 patients in China with acute MI, 93% of whom had ST-segment elevation. In contrast to CLARITY, in COMMIT barely more than half of the patients received fibrinolysis, fewer than 5% proceeded to percutaneous interventions, and no loading dose was given: patients in the clopidogrel group received 75 mg/day from the outset.

At 15 days, the incidence of death, reinfarction, or stroke was 9.2% with clopidogrel compared with 10.1% with placebo, a small but statistically significant difference. Again, the rate of major bleeding was not significantly higher, either overall or in patients over age 70.

Of note, patients over age 75 were excluded from CLARITY, and as mentioned, no loading dose was used in COMMIT. Thus, for patients receiving fibrinolysis who are over age 75, there is no evidence to support the safety of a loading dose, and clopidogrel should be started at 75 mg daily.

Clopidogrel in elective percutaneous coronary intervention

The CREDO trial9 (Clopidogrel for the Reduction of Events During Observation) was in patients referred for elective percutaneous coronary intervention. Three to 24 hours before the procedure, the patients received either a 300-mg loading dose of clopidogrel or placebo; afterward, all patients received clopidogrel 75 mg/day for 28 days. All patients also received aspirin.

A clopidogrel loading dose 3 to 24 hours before the intervention did not produce a statistically significant reduction in ischemic events, although a post hoc subgroup analysis suggested that patients who received the loading dose between 6 and 24 hours before did benefit, with a relative risk reduction of 38.6% in the composite end point (P = .051).

After 28 days, the patients who had received the clopidogrel loading dose were continued on clopidogrel, while those in the placebo group were switched back to placebo. At 1 year, the investigators found a significantly lower rate of the composite end point with the prolonged course of clopidogrel (8.5% vs 11.5%).

In summary, these studies found clopidogrel to be beneficial in a broad spectrum of coronary diseases. Subgroup analyses suggest that pretreatment before percutaneous coronary intervention provides additional benefit, particularly if clopidogrel is given at least 6 hours in advance (the time necessary for clopidogrel to cause substantial platelet inhibition).

SOME PATIENTS RESPOND LESS TO CLOPIDOGREL

The level of platelet inhibition induced by clopidogrel varies. In different studies, the frequency of clopidogrel “nonresponsiveness” ranged from 5% to 56% of patients, depending on which test and which cutoff values were used. The distribution of responses to clopidogrel is wide and fits a normal gaussian curve.10

A large fraction of the population carries a gene that may account for some of the interpatient variation in platelet inhibition with clopidogrel. Carriers of a reduced-function CYP2C19 allele—approximately 30% of people in one study—have significantly lower levels of the active metabolite of clopidogrel, less platelet inhibition from clopidogrel therapy, and a 53% higher rate of death from cardiovascular causes, MI, or stroke.11

 

 

PRASUGREL, THE NEWEST THIENOPYRIDINE

Prasugrel, FDA-approved in July 2009 for the treatment of acute coronary syndromes, is given in an oral loading dose of 60 mg followed by an oral maintenance dose of 10 mg daily.

Pharmacology of prasugrel vs clopidogrel

As noted previously, the thienopyridines are prodrugs that require hepatic conversion to exert antiplatelet effects.

Metabolism. Prasugrel’s hepatic activation involves a single step, in contrast to the multiple-step process required for activation of clopidogrel. Clopidogrel is primarily hydrolyzed by intestinal and plasma esterases to an inactive terminal metabolite, with the residual unhydrolized drug undergoing a two-step metabolism that depends on cytochrome P450 enzymes. Prasugrel is also extensively hydrolyzed by these esterases, but the intermediate product is then metabolized in a single step to the active sulfhydryl compound, mainly by CYP3A4 and CYP2B6.

Thus, about 80% of an orally absorbed dose of prasugrel is converted to active drug, compared with only 10% to 20% of absorbed clopidogrel.

Time to peak effect. With clopidogrel, maximal inhibition of platelet aggregation occurs 3 to 5 days after starting therapy with 75 mg daily without a loading dose, but within 4 to 6 hours if a loading dose of 300 to 600 mg is given. In contrast, a prasugrel loading dose produces more than 80% of its platelet inhibitory effects by 30 minutes, and peak activity is observed within 4 hours.12 The platelet inhibition induced by prasugrel at 30 minutes after administration is comparable to the peak effect of clopidogrel at 6 hours.13

Dose-response. Prasugrel’s inhibition of platelet aggregation is dose-related.

Prasugrel is about 10 times more potent than clopidogrel and 100 times more potent than ticlopidine. Thus, treatment with 5 mg of prasugrel results in inhibition of platelet activity (distributed in a gaussian curve) very similar to that produced by 75 mg of clopidogrel. On the other hand, even a maintenance dose of 150 mg of clopidogrel inhibits platelet activity to a lesser degree than 10 mg of prasugrel (46% vs 61%),14 so clopidogrel appears to reach a plateau of platelet inhibition that prasugrel can overcome.

At the approved dose of prasugrel, inhibition of platelet aggregation is significantly greater and there are fewer “nonresponders” than with clopidogrel.

Interactions. Drugs that inhibit CYP3A4 do not inhibit the efficacy of prasugrel, but they can inhibit that of clopidogrel. Some commonly used drugs that have this effect are the statins (eg, atorvastain [Lipitor]) and the macrolide antibiotics (eg, erythromycin). Furthermore, whereas proton pump inhibitors have been shown to diminish the effect of clopidogrel by reducing the formation of its active metabolite, no such effect has been noted with prasugrel.

Prasugrel in phase 2 trials: Finding the optimal dosage

A phase 2 trial compared three prasugrel regimens (loading dose/daily maintenance dose of 40 mg/7.5 mg, 60 mg/10 mg, and 60 mg/15 mg) and standard clopidogrel therapy (300 mg/75 mg) in patients undergoing elective or urgent percutaneous coronary intervention.15 No significant difference in outcomes was seen in the groups receiving the three prasugrel regimens. However, more “minimal bleeding events” (defined by the criteria of the TIMI trial16) occurred with high-dose prasugrel than with lower-dose prasugrel or with clopidogrel, leading to use of the intermediate-dose prasugrel regimen (60-mg loading dose, 10-mg daily maintenance) for later trials.

Another phase 2 trial randomized 201 patients undergoing elective percutaneous coronary intervention to receive prasugrel 60 mg/10 mg or clopidogrel 600 mg/150 mg.14 In all patients, the loading dose was given about 1 hour before cardiac catheterization. As soon as 30 minutes after the loading dose, platelet inhibition was superior with prasugrel (31% vs 5% inhibition of platelet aggregation), and it remained significantly higher at 6 hours (75% vs 32%) and during the maintenance phase (61% vs 46%).

 

 

Phase 3 trial of prasugrel vs clopidogrel: TRITON-TIMI 38

Only one large phase 3 trial of prasugrel has been completed: TRITON-TIMI 38 (the Trial to Assess Improvement in Therapeutic Outcomes by Optimizing Platelet Inhibition With Prasugrel—Thrombolysis in Myocardial Infarction),17 which enrolled adults with moderate-risk to high-risk acute coronary syndromes scheduled to undergo a percutaneous coronary intervention. In this trial, 10,074 patients were enrolled who had moderate-to high-risk unstable angina or non-ST-elevation MI, and 3,534 patients were enrolled who had ST-elevation MI.

Patients were randomized to receive prasugrel (a 60-mg loading dose, then 10 mg daily) or clopidogrel (a 300-mg loading dose, then 75 mg daily) and were treated for 6 to 15 months. All patients also received aspirin.

The primary end point, a composite of death from cardiovascular causes, nonfatal MI, or nonfatal stroke, occurred in significantly fewer patients treated with prasugrel than with clopidogrel (9.9% vs 12.1%, P < .001) (Table 1). Most of the benefit was due to fewer nonfatal MIs during the follow-up period (7.4% vs 9.7%, P < .001). Additionally, the prasugrel group had a significantly lower rate of stent thrombosis compared with the clopidogrel group (1.1% vs 2.4%; P < .001).

These benefits came at a price of more bleeding. Of those patients who did not undergo coronary artery bypass grafting, more experienced bleeding in the prasugrel group than in the clopidogrel group (2.4% vs 1.8%, P = .03), including a higher rate of life-threatening bleeding (1.4% vs 0.89%, P = .01) and fatal bleeding (0.4% vs 0.1%, P = .002). More patients discontinued prasugrel because of hemorrhage (2.5% vs 1.4%, P < .001). In patients who proceeded to coronary artery bypass grafting, the rate of major bleeding was more than four times higher in those who received prasugrel than in those who received clopidogrel (13.4% vs 3.2%, P < .001).

A higher rate of adverse events related to colon cancer was also noted in patients treated with prasugrel, although the authors suggest this may have resulted from the stronger antiplatelet effects of prasugrel bringing more tumors to medical attention due to bleeding.

Overall death rates did not differ significantly between the treatment groups.

In a post hoc analysis,18 prasugrel was superior to clopidogrel in preventing ischemic events both during the first 3 days following randomization (the “loading phase”) and for the remainder of the trial (the “maintenance phase”). Whereas bleeding risk was similar with the two drugs during the loading phase, prasugrel was subsequently associated with more bleeding during the maintenance phase.

Certain patient subgroups had no net benefit or even suffered harm from prasugrel compared with clopidogrel.17 Patients with previous stroke or transient ischemic attack had net harm from prasugrel (hazard ratio 1.54, P = .04) and showed a strong trend toward a greater rate of major bleeding (P = .06). Patients age 75 and older and those weighing less than 60 kg had no net benefit from prasugrel.

Cost of prasugrel

Prasugrel is currently priced at 18% more than clopidogrel, with average wholesale prices per pill of $6.65 for prasugrel 10 mg compared with $5.63 for clopidogrel 75 mg. (Prasugrel 10-mg pills cost $6.33 at drugstore.com or $7.60 at CVS; clopidogrel 75-mg pills cost $5.33 at drugstore.com or $6.43 at CVS.) The patent on clopidogrel expires in November 2011, after which the price differential is expected to become significantly greater.

TICAGRELOR, A REVERSIBLE ORAL AGENT

Ticagrelor, the first reversible oral P2Y12 receptor antagonist, is an alternative to thienopyridine therapy for acute coronary syndromes.

Ticagrelor is quickly absorbed, does not require metabolic activation, and has a rapid antiplatelet effect and offset of effect, which closely follow drug-exposure levels. In a large randomized controlled trial in patients with acute coronary syndromes with or without STsegment elevation, treatment with ticagrelor compared with clopidogrel resulted in a significant reduction in death from vascular causes, MI, or stroke (9.8% vs 11.7%).19

Given its reversible effect on platelet inhibition, ticagrelor may be preferred in patients whose coronary anatomy is unknown and for whom coronary artery bypass grafting is deemed probable. It is still undergoing trials and is not yet approved.

 

 

TAKE-HOME POINTS

Prasugrel is more potent, more rapid in onset, and more consistent in inhibiting platelet aggregation than clopidogrel. A large clinical trial17 found prasugrel to be superior to clopidogrel for patients with moderate-to high-risk acute coronary syndromes with high probability of undergoing a percutaneous coronary intervention.

Who should receive prasugrel, and how?

Prasugrel should be given after angiography to patients with non-ST-elevation acute coronary syndromes or at presentation to patients with ST-elevation MI. When used for planned percutaneous coronary intervention, prasugrel should be given at least 30 minutes before the intervention, as was done in phase 2 trials (although its routine use in this situation is not recommended—see below).

It is given in a one-time loading dose of 60 mg by mouth and then maintained with 10 mg by mouth once daily for at least 1 year. (At least 9 months of treatment with a thienopyridine is indicated for patients with acute coronary syndromes who are medically treated, and at least 1 year is indicated following urgent or elective percutaneous coronary intervention, including balloon angioplasty and placement of a bare-metal or drug-eluting stent.)

Who should not receive prasugrel?

For now, prasugrel should be avoided in favor of clopidogrel in patients at higher risk of bleeding. It is clearly contraindicated in patients with prior transient ischemic attack or stroke, for whom the risk of serious bleeding seems to be prohibitive. It should generally be avoided in patients age 75 and older, although it might be considered in those at particularly high risk of stent thrombosis, such as those with diabetes or prior MI. In patients weighing less than 60 kg, the package insert advises a reduced dose (5 mg), although clinical evidence for this practice is lacking.

As yet, we have no data assuring that prasugrel is safe to use in combination with fibrinolytic agents, so patients on thrombolytic therapy for acute MI should continue to receive clopidogrel starting immediately after lysis. Furthermore, in patients who proceeded to coronary artery bypass grafting, the rate of major bleeding was more than four times higher in the prasugrel group than in the clopidogrel group in the TRITON-TIMI 38 trial.17 No thienopyridine should be given to patients likely to proceed to coronary artery bypass grafting.

Only clopidogrel has evidence supporting its use as an alternative to aspirin for patients with atherosclerotic disease who cannot tolerate aspirin. Neither drug has evidence for use for primary prevention.

Other areas of uncertainty

Prior to angiography. Indications for prasugrel are currently limited by the narrow scope of the trial data. TRITON-TIMI 38,17 the only large trial completed to date, randomized patients to receive prasugrel only after their coronary anatomy was known, except for ST-elevation MI patients. It is unknown whether the benefits of prasugrel will outweigh the higher risk of bleeding in patients with acute coronary syndromes who do not proceed to percutaneous coronary interventions.

A clinical trial is currently under way comparing prasugrel with clopidogrel in 10,000 patients with acute coronary syndromes who will be medically managed without planned revascularization: A Comparison of Prasugrel and Clopidogrel in Acute Coronary Syndrome Subjects (TRILOGY ACS), ClinicalTrials.gov Identifier: NCT00699998. The trial has an estimated completion date of March 2011.

In cases of non-ST-elevation acute coronary syndrome, it is reasonable to wait to give a thienopyridine until after the coronary anatomy has been defined, if angiography will be completed soon after presentation. For example, a 1-hour delay before giving prasugrel still delivers antiplatelet therapy more quickly than giving clopidogrel on presentation. If longer delays are expected before angiography, however, the patient should be given a loading dose of clopidogrel “up front,” in accordance with guidelines published by the American College of Cardiology, American Heart Association, and European Society of Cardiology,20 which recommend starting a thienopyridine early during hospitalization based on trial data with clopidogrel.

Patients undergoing elective percutaneous coronary intervention are at lower risk of stent thrombosis and other ischemic complications, so it is possible that the benefits of prasugrel would not outweigh the risks in these patients. Thus, prasugrel cannot yet be recommended for routine elective percutaneous coronary intervention except in individual cases in which the interventionalist feels that the patient may be at higher risk of thrombosis.

Prasugrel (Effient) is more potent and consistent in its effects than clopidogrel (Plavix), thus preventing more thrombotic events—but at a price of more bleeding. Therefore, the drugs must be appropriately selected for the individual patient.

Over the last 9 years, the thienopyridines—ticlopidine (Ticlid), clopidogrel, and now prasugrel—have become essential tools for treating acute coronary syndromes.

The usual underlying mechanism of acute coronary syndromes is thrombosis, caused by rupture of atherosclerotic plaque.1 Accordingly, antithrombotic agents—aspirin, heparin, lowmolecular-weight heparin, glycoprotein IIb/IIIa inhibitors, the direct thrombin inhibitor bivalirudin (Angiomax), and thienopyridines—have all been shown to reduce the risk of major adverse cardiac outcomes in this setting.

In this article, we review the pharmacology and evidence of effectiveness of the thienopyridine drugs, focusing on prasugrel, the latest thienopyridine to be approved by the US Food and Drug Administration (FDA).

THIENOPYRIDINES INHIBIT PLATELET ACTIVATION AND AGGREGATION

Thienopyridines are prodrugs that require conversion by hepatic cytochrome P450 enzymes. The active metabolites bind irreversibly to platelet P2Y12 receptors. Consequently, they permanently block signalling mediated by platelet adenosine diphosphate-P2Y12 receptors, thereby inhibiting glycoprotein IIb/IIIa receptor activation and platelet aggregation.

Aspirin, in contrast, inhibits platelets by blocking the thromboxane-mediated pathway. Therefore, the combination of aspirin plus a thienopyridine has an additive effect.2

The effect of thienopyridines on platelets is irreversible. Therefore, although the half-life of prasugrel’s active metabolite is 3.7 hours, its inhibitory effects last for 96 hours, essentially the time for half the body’s circulating platelets to be replaced.

TICLOPIDINE, THE FIRST THIENOPYRIDINE

Ticlopidine was the first thienopyridine to be approved by the FDA. Its initial studies in unstable angina were small, their designs did not call for patients to concurrently receive aspirin, and all they showed was that ticlopidine was about as beneficial as aspirin. Consequently, the studies had little impact on clinical practice.3

In a pivotal trial,4 patients who received coronary stents were randomized to afterward receive either the combination of ticlopidine plus aspirin or anticoagulation therapy with heparin, phenprocoumon (a coumarin derivative available in Europe), and aspirin. At 30 days, an ischemic complication (death, myocardial infarction [MI], repeat intervention) had occurred in 6.2% of the anticoagulation therapy group vs 1.6% of the ticlopidine group, a risk reduction of 75%. Rates of stent occlusion, MI, and revascularization were 80% to 85% lower in the ticlodipine group. This study paved the way for widespread use of thienopyridines.

Ticlopidine’s use was limited, however, by a 2.4% incidence of serious granulocytopenia and rare cases of thrombocytopenic purpura.

BENEFIT OF CLOPIDOGREL

Although prasugrel is the focus of this review, the trials of prasugrel all compared its efficacy with that of clopidogrel. Furthermore, many patients should still receive clopidogrel and not prasugrel, so it is important to be familiar with the evidence of clopidogrel’s benefit.

Once approved for clinical use, clopidogrel was substituted for ticlopidine in patients undergoing coronary stenting on the basis of studies showing it to be at least as effective as ticlopidine and more tolerable. A series of trials of clopidogrel were done in patients across a spectrum of risk groups, from those at high risk of coronary heart disease to those presenting with ST-elevation MI. The time of pretreatment in the studies ranged from 3 hours to 6 days before percutaneous coronary intervention, and the duration of treatment following intervention ranged from 30 days to 1 year.

Clopidogrel in non-ST-elevation acute coronary syndromes

The CURE trial2 (Clopidogrel in Unstable Angina to Prevent Recurrent Events), published in 2001, established clopidogrel as a therapy for unstable ischemic syndromes, whether treated medically or with revascularization. In that trial, 12,562 patients with acute coronary syndromes without ST elevation (ie, unstable angina or non-ST-elevation MI), as defined by electrocardiographic changes or positive cardiac markers, were randomized to receive clopidogrel (a 300-mg loading dose followed by 75-mg maintenance doses) or placebo for a mean duration of 9 months. All patients also received aspirin 75 mg to 325 mg daily.

The composite outcome of death from cardiovascular causes, nonfatal MI, or stroke occurred in 20% fewer patients treated with clopidogrel than with placebo (9.3% vs 11.4%). The benefit was similar in patients undergoing revascularization compared with those treated medically.

Although there were significantly more cases of major bleeding in the clopidogrel group than in the placebo group (3.7% vs 2.7%), the number of episodes of life-threatening bleeding or hemorrhagic strokes was the same.

PCI-CURE5 was a substudy of the CURE trial in patients who underwent a percutaneous coronary intervention. Patients were pretreated with clopidogrel or placebo for a mean of 6 days before the procedure. Afterward, they all received clopidogrel plus aspirin in an unblinded fashion for 2 to 4 weeks, and then the randomized study drug was resumed for a mean of 8 months.

Significantly fewer adverse events occurred in the clopidogrel group as tallied at the time of the intervention, 1 month later, and 8 months later.

 

 

Clopidogrel in ST-elevation acute MI

The CLARITY-TIMI 28 trial6 (Clopidogrel as Adjunctive Reperfusion Therapy—Thrombolysis in Myocardial Infarction 28) showed that adding clopidogrel (a 300-mg loading dose, then 75 mg daily) to aspirin benefitted patients with ST-elevation MI receiving fibrinolytic therapy. At 30 days, cardiovascular death, recurrent MI, or urgent revascularization had occurred in 11.6% of the clopidogrel group vs 14.1% of the placebo group, a statistically significant difference. The rates of major or minor bleeding were no higher in the clopidogrel group than in the placebo group, an especially remarkable finding in patients receiving thrombolytic therapy.

PCI-CLARITY.7 About half of the patients in the CLARITY trial ultimately underwent a percutaneous coronary intervention after fibrinolytic therapy, with results reported as the PCI-CLARITY substudy. Like those in PCI-CURE, these patients were randomized to receive pretreatment with either clopidogrel or placebo before the procedure, in this study for a median of 3 days. Both groups received clopidogrel afterward. At 30 days from randomization, the outcome of cardiovascular death, MI, or stroke had occurred in 7.5% of the clopidogrel group compared with 12.0% of the placebo group, which was statistically significant, without any significant excess in the rates of major or minor bleeding.

COMMIT8 (the Clopidogrel and Metoprolol in Myocardial Infarction Trial) also showed clopidogrel to be beneficial in patients with acute MI. This trial included more than 45,000 patients in China with acute MI, 93% of whom had ST-segment elevation. In contrast to CLARITY, in COMMIT barely more than half of the patients received fibrinolysis, fewer than 5% proceeded to percutaneous interventions, and no loading dose was given: patients in the clopidogrel group received 75 mg/day from the outset.

At 15 days, the incidence of death, reinfarction, or stroke was 9.2% with clopidogrel compared with 10.1% with placebo, a small but statistically significant difference. Again, the rate of major bleeding was not significantly higher, either overall or in patients over age 70.

Of note, patients over age 75 were excluded from CLARITY, and as mentioned, no loading dose was used in COMMIT. Thus, for patients receiving fibrinolysis who are over age 75, there is no evidence to support the safety of a loading dose, and clopidogrel should be started at 75 mg daily.

Clopidogrel in elective percutaneous coronary intervention

The CREDO trial9 (Clopidogrel for the Reduction of Events During Observation) was in patients referred for elective percutaneous coronary intervention. Three to 24 hours before the procedure, the patients received either a 300-mg loading dose of clopidogrel or placebo; afterward, all patients received clopidogrel 75 mg/day for 28 days. All patients also received aspirin.

A clopidogrel loading dose 3 to 24 hours before the intervention did not produce a statistically significant reduction in ischemic events, although a post hoc subgroup analysis suggested that patients who received the loading dose between 6 and 24 hours before did benefit, with a relative risk reduction of 38.6% in the composite end point (P = .051).

After 28 days, the patients who had received the clopidogrel loading dose were continued on clopidogrel, while those in the placebo group were switched back to placebo. At 1 year, the investigators found a significantly lower rate of the composite end point with the prolonged course of clopidogrel (8.5% vs 11.5%).

In summary, these studies found clopidogrel to be beneficial in a broad spectrum of coronary diseases. Subgroup analyses suggest that pretreatment before percutaneous coronary intervention provides additional benefit, particularly if clopidogrel is given at least 6 hours in advance (the time necessary for clopidogrel to cause substantial platelet inhibition).

SOME PATIENTS RESPOND LESS TO CLOPIDOGREL

The level of platelet inhibition induced by clopidogrel varies. In different studies, the frequency of clopidogrel “nonresponsiveness” ranged from 5% to 56% of patients, depending on which test and which cutoff values were used. The distribution of responses to clopidogrel is wide and fits a normal gaussian curve.10

A large fraction of the population carries a gene that may account for some of the interpatient variation in platelet inhibition with clopidogrel. Carriers of a reduced-function CYP2C19 allele—approximately 30% of people in one study—have significantly lower levels of the active metabolite of clopidogrel, less platelet inhibition from clopidogrel therapy, and a 53% higher rate of death from cardiovascular causes, MI, or stroke.11

 

 

PRASUGREL, THE NEWEST THIENOPYRIDINE

Prasugrel, FDA-approved in July 2009 for the treatment of acute coronary syndromes, is given in an oral loading dose of 60 mg followed by an oral maintenance dose of 10 mg daily.

Pharmacology of prasugrel vs clopidogrel

As noted previously, the thienopyridines are prodrugs that require hepatic conversion to exert antiplatelet effects.

Metabolism. Prasugrel’s hepatic activation involves a single step, in contrast to the multiple-step process required for activation of clopidogrel. Clopidogrel is primarily hydrolyzed by intestinal and plasma esterases to an inactive terminal metabolite, with the residual unhydrolized drug undergoing a two-step metabolism that depends on cytochrome P450 enzymes. Prasugrel is also extensively hydrolyzed by these esterases, but the intermediate product is then metabolized in a single step to the active sulfhydryl compound, mainly by CYP3A4 and CYP2B6.

Thus, about 80% of an orally absorbed dose of prasugrel is converted to active drug, compared with only 10% to 20% of absorbed clopidogrel.

Time to peak effect. With clopidogrel, maximal inhibition of platelet aggregation occurs 3 to 5 days after starting therapy with 75 mg daily without a loading dose, but within 4 to 6 hours if a loading dose of 300 to 600 mg is given. In contrast, a prasugrel loading dose produces more than 80% of its platelet inhibitory effects by 30 minutes, and peak activity is observed within 4 hours.12 The platelet inhibition induced by prasugrel at 30 minutes after administration is comparable to the peak effect of clopidogrel at 6 hours.13

Dose-response. Prasugrel’s inhibition of platelet aggregation is dose-related.

Prasugrel is about 10 times more potent than clopidogrel and 100 times more potent than ticlopidine. Thus, treatment with 5 mg of prasugrel results in inhibition of platelet activity (distributed in a gaussian curve) very similar to that produced by 75 mg of clopidogrel. On the other hand, even a maintenance dose of 150 mg of clopidogrel inhibits platelet activity to a lesser degree than 10 mg of prasugrel (46% vs 61%),14 so clopidogrel appears to reach a plateau of platelet inhibition that prasugrel can overcome.

At the approved dose of prasugrel, inhibition of platelet aggregation is significantly greater and there are fewer “nonresponders” than with clopidogrel.

Interactions. Drugs that inhibit CYP3A4 do not inhibit the efficacy of prasugrel, but they can inhibit that of clopidogrel. Some commonly used drugs that have this effect are the statins (eg, atorvastain [Lipitor]) and the macrolide antibiotics (eg, erythromycin). Furthermore, whereas proton pump inhibitors have been shown to diminish the effect of clopidogrel by reducing the formation of its active metabolite, no such effect has been noted with prasugrel.

Prasugrel in phase 2 trials: Finding the optimal dosage

A phase 2 trial compared three prasugrel regimens (loading dose/daily maintenance dose of 40 mg/7.5 mg, 60 mg/10 mg, and 60 mg/15 mg) and standard clopidogrel therapy (300 mg/75 mg) in patients undergoing elective or urgent percutaneous coronary intervention.15 No significant difference in outcomes was seen in the groups receiving the three prasugrel regimens. However, more “minimal bleeding events” (defined by the criteria of the TIMI trial16) occurred with high-dose prasugrel than with lower-dose prasugrel or with clopidogrel, leading to use of the intermediate-dose prasugrel regimen (60-mg loading dose, 10-mg daily maintenance) for later trials.

Another phase 2 trial randomized 201 patients undergoing elective percutaneous coronary intervention to receive prasugrel 60 mg/10 mg or clopidogrel 600 mg/150 mg.14 In all patients, the loading dose was given about 1 hour before cardiac catheterization. As soon as 30 minutes after the loading dose, platelet inhibition was superior with prasugrel (31% vs 5% inhibition of platelet aggregation), and it remained significantly higher at 6 hours (75% vs 32%) and during the maintenance phase (61% vs 46%).

 

 

Phase 3 trial of prasugrel vs clopidogrel: TRITON-TIMI 38

Only one large phase 3 trial of prasugrel has been completed: TRITON-TIMI 38 (the Trial to Assess Improvement in Therapeutic Outcomes by Optimizing Platelet Inhibition With Prasugrel—Thrombolysis in Myocardial Infarction),17 which enrolled adults with moderate-risk to high-risk acute coronary syndromes scheduled to undergo a percutaneous coronary intervention. In this trial, 10,074 patients were enrolled who had moderate-to high-risk unstable angina or non-ST-elevation MI, and 3,534 patients were enrolled who had ST-elevation MI.

Patients were randomized to receive prasugrel (a 60-mg loading dose, then 10 mg daily) or clopidogrel (a 300-mg loading dose, then 75 mg daily) and were treated for 6 to 15 months. All patients also received aspirin.

The primary end point, a composite of death from cardiovascular causes, nonfatal MI, or nonfatal stroke, occurred in significantly fewer patients treated with prasugrel than with clopidogrel (9.9% vs 12.1%, P < .001) (Table 1). Most of the benefit was due to fewer nonfatal MIs during the follow-up period (7.4% vs 9.7%, P < .001). Additionally, the prasugrel group had a significantly lower rate of stent thrombosis compared with the clopidogrel group (1.1% vs 2.4%; P < .001).

These benefits came at a price of more bleeding. Of those patients who did not undergo coronary artery bypass grafting, more experienced bleeding in the prasugrel group than in the clopidogrel group (2.4% vs 1.8%, P = .03), including a higher rate of life-threatening bleeding (1.4% vs 0.89%, P = .01) and fatal bleeding (0.4% vs 0.1%, P = .002). More patients discontinued prasugrel because of hemorrhage (2.5% vs 1.4%, P < .001). In patients who proceeded to coronary artery bypass grafting, the rate of major bleeding was more than four times higher in those who received prasugrel than in those who received clopidogrel (13.4% vs 3.2%, P < .001).

A higher rate of adverse events related to colon cancer was also noted in patients treated with prasugrel, although the authors suggest this may have resulted from the stronger antiplatelet effects of prasugrel bringing more tumors to medical attention due to bleeding.

Overall death rates did not differ significantly between the treatment groups.

In a post hoc analysis,18 prasugrel was superior to clopidogrel in preventing ischemic events both during the first 3 days following randomization (the “loading phase”) and for the remainder of the trial (the “maintenance phase”). Whereas bleeding risk was similar with the two drugs during the loading phase, prasugrel was subsequently associated with more bleeding during the maintenance phase.

Certain patient subgroups had no net benefit or even suffered harm from prasugrel compared with clopidogrel.17 Patients with previous stroke or transient ischemic attack had net harm from prasugrel (hazard ratio 1.54, P = .04) and showed a strong trend toward a greater rate of major bleeding (P = .06). Patients age 75 and older and those weighing less than 60 kg had no net benefit from prasugrel.

Cost of prasugrel

Prasugrel is currently priced at 18% more than clopidogrel, with average wholesale prices per pill of $6.65 for prasugrel 10 mg compared with $5.63 for clopidogrel 75 mg. (Prasugrel 10-mg pills cost $6.33 at drugstore.com or $7.60 at CVS; clopidogrel 75-mg pills cost $5.33 at drugstore.com or $6.43 at CVS.) The patent on clopidogrel expires in November 2011, after which the price differential is expected to become significantly greater.

TICAGRELOR, A REVERSIBLE ORAL AGENT

Ticagrelor, the first reversible oral P2Y12 receptor antagonist, is an alternative to thienopyridine therapy for acute coronary syndromes.

Ticagrelor is quickly absorbed, does not require metabolic activation, and has a rapid antiplatelet effect and offset of effect, which closely follow drug-exposure levels. In a large randomized controlled trial in patients with acute coronary syndromes with or without STsegment elevation, treatment with ticagrelor compared with clopidogrel resulted in a significant reduction in death from vascular causes, MI, or stroke (9.8% vs 11.7%).19

Given its reversible effect on platelet inhibition, ticagrelor may be preferred in patients whose coronary anatomy is unknown and for whom coronary artery bypass grafting is deemed probable. It is still undergoing trials and is not yet approved.

 

 

TAKE-HOME POINTS

Prasugrel is more potent, more rapid in onset, and more consistent in inhibiting platelet aggregation than clopidogrel. A large clinical trial17 found prasugrel to be superior to clopidogrel for patients with moderate-to high-risk acute coronary syndromes with high probability of undergoing a percutaneous coronary intervention.

Who should receive prasugrel, and how?

Prasugrel should be given after angiography to patients with non-ST-elevation acute coronary syndromes or at presentation to patients with ST-elevation MI. When used for planned percutaneous coronary intervention, prasugrel should be given at least 30 minutes before the intervention, as was done in phase 2 trials (although its routine use in this situation is not recommended—see below).

It is given in a one-time loading dose of 60 mg by mouth and then maintained with 10 mg by mouth once daily for at least 1 year. (At least 9 months of treatment with a thienopyridine is indicated for patients with acute coronary syndromes who are medically treated, and at least 1 year is indicated following urgent or elective percutaneous coronary intervention, including balloon angioplasty and placement of a bare-metal or drug-eluting stent.)

Who should not receive prasugrel?

For now, prasugrel should be avoided in favor of clopidogrel in patients at higher risk of bleeding. It is clearly contraindicated in patients with prior transient ischemic attack or stroke, for whom the risk of serious bleeding seems to be prohibitive. It should generally be avoided in patients age 75 and older, although it might be considered in those at particularly high risk of stent thrombosis, such as those with diabetes or prior MI. In patients weighing less than 60 kg, the package insert advises a reduced dose (5 mg), although clinical evidence for this practice is lacking.

As yet, we have no data assuring that prasugrel is safe to use in combination with fibrinolytic agents, so patients on thrombolytic therapy for acute MI should continue to receive clopidogrel starting immediately after lysis. Furthermore, in patients who proceeded to coronary artery bypass grafting, the rate of major bleeding was more than four times higher in the prasugrel group than in the clopidogrel group in the TRITON-TIMI 38 trial.17 No thienopyridine should be given to patients likely to proceed to coronary artery bypass grafting.

Only clopidogrel has evidence supporting its use as an alternative to aspirin for patients with atherosclerotic disease who cannot tolerate aspirin. Neither drug has evidence for use for primary prevention.

Other areas of uncertainty

Prior to angiography. Indications for prasugrel are currently limited by the narrow scope of the trial data. TRITON-TIMI 38,17 the only large trial completed to date, randomized patients to receive prasugrel only after their coronary anatomy was known, except for ST-elevation MI patients. It is unknown whether the benefits of prasugrel will outweigh the higher risk of bleeding in patients with acute coronary syndromes who do not proceed to percutaneous coronary interventions.

A clinical trial is currently under way comparing prasugrel with clopidogrel in 10,000 patients with acute coronary syndromes who will be medically managed without planned revascularization: A Comparison of Prasugrel and Clopidogrel in Acute Coronary Syndrome Subjects (TRILOGY ACS), ClinicalTrials.gov Identifier: NCT00699998. The trial has an estimated completion date of March 2011.

In cases of non-ST-elevation acute coronary syndrome, it is reasonable to wait to give a thienopyridine until after the coronary anatomy has been defined, if angiography will be completed soon after presentation. For example, a 1-hour delay before giving prasugrel still delivers antiplatelet therapy more quickly than giving clopidogrel on presentation. If longer delays are expected before angiography, however, the patient should be given a loading dose of clopidogrel “up front,” in accordance with guidelines published by the American College of Cardiology, American Heart Association, and European Society of Cardiology,20 which recommend starting a thienopyridine early during hospitalization based on trial data with clopidogrel.

Patients undergoing elective percutaneous coronary intervention are at lower risk of stent thrombosis and other ischemic complications, so it is possible that the benefits of prasugrel would not outweigh the risks in these patients. Thus, prasugrel cannot yet be recommended for routine elective percutaneous coronary intervention except in individual cases in which the interventionalist feels that the patient may be at higher risk of thrombosis.

References
  1. Yeghiazarians Y, Braunstein JB, Askari A, Stone PH. Unstable angina pectoris. N Engl J Med 2000; 342:101114.
  2. Yusuf S, Zhao F, Mehta SR, Chrolavicius S, Tognoni G, Fox KK; Clopidogrel in Unstable Angina to Prevent Recurrent Events Trial Investigators. Effects of clopidogrel in addition to aspirin in patients with acute coronary syndromes without ST-segment elevation. N Engl J Med 2001; 345:494502.
  3. Balsano F, Rizzon P, Violi F, et al. Antiplatelet treatment with ticlopidine in unstable angina. A controlled multicenter clinical trial. The Studio della Ticlopidina nell'Angina Instabile Group. Circulation 1990; 82:1726.
  4. Schömig A, Neumann FJ, Kastrati A, et al. A randomized comparison of antiplatelet and anticoagulant therapy after the placement of coronary-artery stents. N Engl J Med 1996; 334:10841089.
  5. Mehta SR, Yusuf S, Peters RJG, et al; Clopidogrel in Unstable Angina to Prevent Recurrent Events Trial (CURE) Investigators. Effects of pretreatment with clopidogrel and aspirin followed by long-term therapy in patients undergoing percutaneous coronary intervention: the PCI-CURE study. Lancet 2001; 358:527533.
  6. Sabatine MS, Cannon CP, Gibson CM, et al; CLA RITY-TIMI 28 Investigators. Addition of clopidogrel to aspirin and fibrinolytic therapy for myocardial infarction with STsegment elevation. N Engl J Med 2005; 352:11791189.
  7. Sabatine MS, Cannon CP, Gibson CM, et al; Clopidogrel as Adjunctive Reperfusion Therapy (CLARITY)-Thrombolysis in Myocardial Infarction (TIMI) 28 Investigators. Effect of clopidogrel pretreatment before percutaneous coronary intervention in patients with ST-elevation myocardial infarction treated with fibrinolytics: the PCI-CLARITY study. JAMA 2005: 294:12241232.
  8. Chen ZM, Jiang LX, Chen YP, et al; COMMIT (ClOpidogrel and Metoprolol in Myocardial Infarction Trial) collaborative group. Addition of clopidogrel to aspirin in 45,852 patients with acute myocardial infarction: randomised placebo-controlled trial. Lancet 2005; 366:16071621.
  9. Steinhubl SR, Berger PB, Mann JT, et al; CREDO Investigators. Clopidogrel for the reduction of events during observation. Early and sustained dual oral antiplatelet therapy following percutaneous coronary intervention: a randomized controlled trial. JAMA 2002; 288:24112420.
  10. Serebruany VL, Steinhubl SR, Berger PB, Malinin AI, Bhatt DL, Topol EJ. Variability in platelet responsiveness to clopidogrel among 544 individuals. J Am Coll Cardiol 2005; 45:246251.
  11. Mega JL, Close SL, Wiviott SD, et al. Cytochrome P-450 polymorphisms and response to clopidogrel. N Engl J Med 2009; 360:354362.
  12. Helft G, Osende JI, Worthley SG, et al. Acute antithrombotic effect of a front-loaded regimen of clopidogrel in patients with atherosclerosis on aspirin. Arterioscler Thromb Vasc Biol 2000; 20:23162321.
  13. Weerakkody GJ, Jakubowski JA, Brandt JT, et al. Comparison of speed of onset of platelet inhibition after loading doses of clopidogrel versus prasugrel in healthy volunteers and correlation with responder status. Am J Cardiol 2007; 100:331336.
  14. Wiviott SD, Trenk D, Frelinger AL, et al; PRINCIPLETIMI 44 Investigators. Prasugrel compared with high loading-and maintenance-dose clopidogrel in patients with planned percutaneous coronary intervention: the Prasugrel in Comparison to Clopidogrel for Inhibition of Platelet Activation and Aggregation-Thrombolysis in Myocardial Infarction 44 trial. Circulation 2007; 116:29232932.
  15. Wiviott SD, Antman EM, Winters KJ, et al; JUMBO-TIMI 26 Investigators. Randomized comparison of prasugrel (CS-747, LY640315), a novel thienopyridine P2Y12 antagonist, with clopidogrel in percutaneous coronary intervention: results of the Joint Utilization of Medications to Block Platelets Optimally (JUMBO)-TIMI 26 Trial. Circulation 2005; 111:33663373.
  16. Bovill EG, Terrin ML, Stump DC, et al. Hemorrhagic events during therapy with recombinant tissue-type plasminogen activator, heparin, and aspirin for acute myocardial infarction. Results of the Thrombolysis in Myocardial Infarction (TIMI) Phase II Trial. Ann Intern Med 1991; 115:256265.
  17. Wiviott SD, Braunwald E, McCabe CH, et al; TRITONTIMI 38 Investigators. Prasugrel versus clopidogrel in patients with acute coronary syndromes. N Engl J Med 2007; 357:20012015.
  18. Antman EM, Wiviott SD, Murphy SA, et al. Early and late benefits of prasugrel in patients with acute coronary syndromes undergoing percutaneous coronary intervention: a TRITON-TIMI 38 (TRial to Assess Improvement in Therapeutic Outcomes by Optimizing Platelet InhibitioN with Prasugrel-Thrombolysis In Myocardial Infarction) analysis. J Am Coll Cardiol 2008; 51:20282033.
  19. Wallentin L, Becker RC, Budaj A, Freij A, Thorsén M, et al; PLATO Investigators. Ticagrelor versus clopidogrel in patients with acute coronary syndromes. N Engl J Med 2009; 361:10451057.
  20. Braunwald E, Antman EM, Beasley JW, et al. ACC/AHA 2002 guideline update for the management of patients with unstable angina and non–ST-segment elevation myocardial infarction—summary article*1: A report of the American College of Cardiology/American Heart Association task force on practice guidelines (Committee on the Management of Patients With Unstable Angina). J Am Coll Cardiol 2002; 40:13661374.
References
  1. Yeghiazarians Y, Braunstein JB, Askari A, Stone PH. Unstable angina pectoris. N Engl J Med 2000; 342:101114.
  2. Yusuf S, Zhao F, Mehta SR, Chrolavicius S, Tognoni G, Fox KK; Clopidogrel in Unstable Angina to Prevent Recurrent Events Trial Investigators. Effects of clopidogrel in addition to aspirin in patients with acute coronary syndromes without ST-segment elevation. N Engl J Med 2001; 345:494502.
  3. Balsano F, Rizzon P, Violi F, et al. Antiplatelet treatment with ticlopidine in unstable angina. A controlled multicenter clinical trial. The Studio della Ticlopidina nell'Angina Instabile Group. Circulation 1990; 82:1726.
  4. Schömig A, Neumann FJ, Kastrati A, et al. A randomized comparison of antiplatelet and anticoagulant therapy after the placement of coronary-artery stents. N Engl J Med 1996; 334:10841089.
  5. Mehta SR, Yusuf S, Peters RJG, et al; Clopidogrel in Unstable Angina to Prevent Recurrent Events Trial (CURE) Investigators. Effects of pretreatment with clopidogrel and aspirin followed by long-term therapy in patients undergoing percutaneous coronary intervention: the PCI-CURE study. Lancet 2001; 358:527533.
  6. Sabatine MS, Cannon CP, Gibson CM, et al; CLA RITY-TIMI 28 Investigators. Addition of clopidogrel to aspirin and fibrinolytic therapy for myocardial infarction with STsegment elevation. N Engl J Med 2005; 352:11791189.
  7. Sabatine MS, Cannon CP, Gibson CM, et al; Clopidogrel as Adjunctive Reperfusion Therapy (CLARITY)-Thrombolysis in Myocardial Infarction (TIMI) 28 Investigators. Effect of clopidogrel pretreatment before percutaneous coronary intervention in patients with ST-elevation myocardial infarction treated with fibrinolytics: the PCI-CLARITY study. JAMA 2005: 294:12241232.
  8. Chen ZM, Jiang LX, Chen YP, et al; COMMIT (ClOpidogrel and Metoprolol in Myocardial Infarction Trial) collaborative group. Addition of clopidogrel to aspirin in 45,852 patients with acute myocardial infarction: randomised placebo-controlled trial. Lancet 2005; 366:16071621.
  9. Steinhubl SR, Berger PB, Mann JT, et al; CREDO Investigators. Clopidogrel for the reduction of events during observation. Early and sustained dual oral antiplatelet therapy following percutaneous coronary intervention: a randomized controlled trial. JAMA 2002; 288:24112420.
  10. Serebruany VL, Steinhubl SR, Berger PB, Malinin AI, Bhatt DL, Topol EJ. Variability in platelet responsiveness to clopidogrel among 544 individuals. J Am Coll Cardiol 2005; 45:246251.
  11. Mega JL, Close SL, Wiviott SD, et al. Cytochrome P-450 polymorphisms and response to clopidogrel. N Engl J Med 2009; 360:354362.
  12. Helft G, Osende JI, Worthley SG, et al. Acute antithrombotic effect of a front-loaded regimen of clopidogrel in patients with atherosclerosis on aspirin. Arterioscler Thromb Vasc Biol 2000; 20:23162321.
  13. Weerakkody GJ, Jakubowski JA, Brandt JT, et al. Comparison of speed of onset of platelet inhibition after loading doses of clopidogrel versus prasugrel in healthy volunteers and correlation with responder status. Am J Cardiol 2007; 100:331336.
  14. Wiviott SD, Trenk D, Frelinger AL, et al; PRINCIPLETIMI 44 Investigators. Prasugrel compared with high loading-and maintenance-dose clopidogrel in patients with planned percutaneous coronary intervention: the Prasugrel in Comparison to Clopidogrel for Inhibition of Platelet Activation and Aggregation-Thrombolysis in Myocardial Infarction 44 trial. Circulation 2007; 116:29232932.
  15. Wiviott SD, Antman EM, Winters KJ, et al; JUMBO-TIMI 26 Investigators. Randomized comparison of prasugrel (CS-747, LY640315), a novel thienopyridine P2Y12 antagonist, with clopidogrel in percutaneous coronary intervention: results of the Joint Utilization of Medications to Block Platelets Optimally (JUMBO)-TIMI 26 Trial. Circulation 2005; 111:33663373.
  16. Bovill EG, Terrin ML, Stump DC, et al. Hemorrhagic events during therapy with recombinant tissue-type plasminogen activator, heparin, and aspirin for acute myocardial infarction. Results of the Thrombolysis in Myocardial Infarction (TIMI) Phase II Trial. Ann Intern Med 1991; 115:256265.
  17. Wiviott SD, Braunwald E, McCabe CH, et al; TRITONTIMI 38 Investigators. Prasugrel versus clopidogrel in patients with acute coronary syndromes. N Engl J Med 2007; 357:20012015.
  18. Antman EM, Wiviott SD, Murphy SA, et al. Early and late benefits of prasugrel in patients with acute coronary syndromes undergoing percutaneous coronary intervention: a TRITON-TIMI 38 (TRial to Assess Improvement in Therapeutic Outcomes by Optimizing Platelet InhibitioN with Prasugrel-Thrombolysis In Myocardial Infarction) analysis. J Am Coll Cardiol 2008; 51:20282033.
  19. Wallentin L, Becker RC, Budaj A, Freij A, Thorsén M, et al; PLATO Investigators. Ticagrelor versus clopidogrel in patients with acute coronary syndromes. N Engl J Med 2009; 361:10451057.
  20. Braunwald E, Antman EM, Beasley JW, et al. ACC/AHA 2002 guideline update for the management of patients with unstable angina and non–ST-segment elevation myocardial infarction—summary article*1: A report of the American College of Cardiology/American Heart Association task force on practice guidelines (Committee on the Management of Patients With Unstable Angina). J Am Coll Cardiol 2002; 40:13661374.
Issue
Cleveland Clinic Journal of Medicine - 76(12)
Issue
Cleveland Clinic Journal of Medicine - 76(12)
Page Number
707-714
Page Number
707-714
Publications
Publications
Topics
Article Type
Display Headline
Prasugrel for acute coronary syndromes: Faster, more potent, but higher bleeding risk
Display Headline
Prasugrel for acute coronary syndromes: Faster, more potent, but higher bleeding risk
Sections
Inside the Article

KEY POINTS

  • The thienopyridines—ticlopidine (Ticlid), clopidogrel (Plavix), and now prasugrel—reduce the risk of death from and serious complications of acute coronary syndromes by inhibiting platelet aggregation.
  • Compared with clopidogrel, prasugrel is more potent, faster in onset, and more consistent in inhibiting platelets.
  • Prasugrel should be avoided in patients at higher risk of bleeding, including those with a history of stroke or transient ischemic attack, those age 75 or older, or those who weigh less than 60 kg.
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media

Acute pancreatitis: Problems in adherence to guidelines

Article Type
Changed
Thu, 04/12/2018 - 11:19
Display Headline
Acute pancreatitis: Problems in adherence to guidelines

Several major gastroenterological and surgical societies have issued guidelines on how to manage acute pancreatitis, based on evidence from high-quality randomized trials and nonrandomized studies as well as on expert opinion.1–3 Information is limited on how well physicians in the United States comply with these guidelines, but compliance is suboptimal in other developed countries, according to several studies,4–8 and we suspect that many US physicians are not following the guidelines either.

Acute pancreatitis is a frequent inpatient diagnosis that internists, gastroenterologists, and surgeons all confront. The most common causes are gallstones and heavy alcohol intake. Its management is typically straightforward: intravenous fluids, analgesia, and nothing by mouth. However, treatment of severe cases can be quite complex, particularly if multiple organ systems are involved or if there are local complications.

The primary aim of this article is to raise awareness of recognized deviations from established recommendations that may lead to adverse patient outcomes.

MEASURING ENZYME LEVELS DAILY ADDS COST BUT LITTLE BENEFIT

Problem: Serum amylase and lipase levels are often needlessly measured every day.

Measuring the serum amylase and lipase levels is useful in diagnosing acute pancreatitis, which requires two of the following three features1:

  • Characteristic abdominal pain
  • Levels of serum amylase or serum lipase, or both, that are three or more times the upper limit of normal
  • Findings of acute pancreatitis on computed tomography (CT).

However, the magnitude or duration of the serum enzyme elevation does not correlate with the severity of the attack. Further, we have noticed that physicians at our hospital often order daily serum amylase and lipase levels in patients admitted with acute pancreatitis.

The American College of Gastroenterology (ACG) guidelines1 state that daily monitoring of amylase and lipase has limited value in managing acute pancreatitis. Rechecking these concentrations may be reasonable if pain fails to resolve or worsens during a prolonged hospitalization, as this may suggest a recurrent attack of acute pancreatitis or a developing pseudocyst. But in most cases of acute pancreatitis, daily serum enzyme measurements add cost but little benefit.

REGULAR ASSESSMENT IS IMPORTANT

Problem: Often, severity assessments are not performed regularly or acted on.

Most cases of acute pancreatitis are mild, with rapid recovery and excellent prognosis. However, 15% to 20% are severe and may result in a prolonged hospitalization, systemic inflammatory response syndrome (SIRS), multiorgan system failure, and death.

In severe acute pancreatitis, as pancreatic enzymes and inflammatory cytokines damage the blood vessels, a vast amount of fluid leaks out into the interstitial (“third”) space. This fluid extravasation leads to decreased effective circulating volume, local pancreatic necrosis, hemodynamic instability, and end-organ failure.

It is important to recognize severe acute pancreatitis early because the patient needs to be transferred to a step-down unit or intensive care unit to receive optimal fluid resuscitation and supportive care for organ dysfunction. After 48 to 72 hours, a prediction of severe acute pancreatitis should also prompt the physician to order CT to detect pancreatic necrosis, and also to initiate nutritional support.

Assessment of severity begins in the emergency room or on admission to the hospital. Older age, obesity, organ failure, and pulmonary infiltrates or pleural effusions are initial indicators of poor prognosis. Signs of SIRS (high or low core body temperature, tachycardia, tachypnea, low or high peripheral white blood cell count) or organ failure (eg, elevated serum creatinine) are present on admission in 21% of patients with acute pancreatitis.9

Hemoconcentration is a marker of decreased effective circulating volume in severe acute pancreatitis. A hematocrit higher than 44% at admission or that rises in the first 24 to 48 hours of admission predicts necrosis.10,11 However, a more robust marker of organ failure may be the blood urea nitrogen level.12

 

 

Clinical scoring systems

Several clinical scoring systems have been studied for assessing severity.

The Ranson score is based on 11 clinical factors, 5 checked at admission and 6 checked at 48 hours (Table 1). Patients are at higher risk of death or “serious illness” (needing 7 or more days of intensive care) if they have 3 or more of these factors.13 In a meta-analysis of 12 studies, a Ranson score of 3 or higher had a sensitivity of 75% and a specificity of 77% for predicting severe acute pancreatitis.14

Limitations of the Ranson score are that it can only be completed after 48 hours, all the data points are not always obtained, and it cannot be repeated on a daily basis. Owing to these limitations and its less-than-optimal predictive value, the Ranson score has fallen into disuse.

The APACHE II (Acute Physiology and Chronic Health Evaluation II) score is more versatile. It is based on multiple clinical and laboratory values, and it correlates very well with the risk of death in acute pancreatitis. Death rates are less than 4% when the APACHE II score is less than 8, and 11% to 18% when it is 8 or higher.1 The trajectory of the APACHE II score in the first 48 hours is also an accurate prognostic indicator.

Previous limitations of the APACHE II score were that it was complicated and timeconsuming to calculate and required arterial blood gas measurements. Easy-to-use online calculators are now available (eg, www.globalrph.com/apacheii.htm), and the venous bicarbonate level and the oxygen saturation can be substituted for the arterial pH and oxygen partial pressure.

BISAP, a new five-point scoring system,15 was recently prospectively validated.12 “BISAP” is an acronym for the five markers it is based on, each of which has been shown to predict severe illness in acute pancreatitis:

  • Blood urea nitrogen level > 25 mg/dL
  • Impaired mental status
  • SIRS
  • Age > 60 years
  • Pleural effusion.

The presence of three or more of these factors correlates with higher risk of death, organ failure, and pancreatic necrosis.12

Compared with APACHE II, BISAP has similar accuracy and is easier to calculate. Also, BISAP was specifically developed for acute pancreatitis, whereas APACHE II is a generic score for all critically ill patients.

The Atlanta criteria16 define severe acute pancreatitis as one or more of the following:

  • A Ranson score of 3 or higher during the first 48 hours
  • An APACHE II score of 8 or higher at any time
  • Failure of one or more organs
  • One or more local complications (eg, necrosis, pseudocysts, abscesses).

Recommendation: Assess severity at least daily

A severity assessment should be performed at admission and at least every day thereafter. Clinical guidelines recognize the importance of severity assessment but vary in their specific recommendations.

The ACG advises calculating the APACHE II score within 3 days of admission and measuring the hematocrit at admission, at 12 hours, and at 24 hours. The level of evidence is III, ie, “from published well-designed trials without randomization, single group prepost, cohort, time series, or matched case controlled studies”.1

The American Gastroenterological Association (AGA) provides a more generalized recommendation, that “clinical judgment” should take into account the presence of risk factors (eg, age, obesity), presence or absence of SIRS, routine laboratory values (eg, hematocrit, serum creatinine), and APACHE II score when assessing severity and making decisions.2

In a German survey, only 32% of gastroenterologists used the APACHE II score for assessing risk in acute pancreatitis, in spite of national guidelines emphasizing its importance.7 Also, not all patients with severe acute pancreatitis are transferred to a step-down unit or intensive care unit as recommended. In a British study,4 only 8 (17%) of 46 patients with predicted severe acute pancreatitis were transferred, and 8 of the 38 patients who were not transferred died.

FLUID MUST BE AGGRESSIVELY REPLACED AND MONITORED

Problem: Often, not enough fluid is replaced, or fluid status is not adequately monitored.

Fluid must be aggressively replaced to balance the massive third-space fluid losses that occur in the early inflammatory phase of acute pancreatitis. Intravascular volume depletion can develop rapidly and result in tachycardia, hypotension, and renal failure. It may also impair the blood flow to the pancreas and worsen necrosis.

Animal studies show that aggressive fluid replacement supports the pancreatic microcirculation and prevents necrosis.17 It may also support the intestinal microcirculation and gut barrier, preventing bacterial translocation.

In humans, no controlled trials have been done to test the efficacy of aggressive fluid resuscitation in acute pancreatitis. However, the notion that intravascular fluid loss contributes to poor outcomes is inferred from human studies showing more necrosis and deaths in patients with hemoconcentration. In one study, patients who received inadequate fluid replacement (evidenced by a rise in hematocrit at 24 hours) were more likely to develop necrotizing pancreatitis.18

 

 

Recommendation: Early, aggressive fluid replacement

Experts have suggested initially infusing 500 to 1,000 mL of fluid per hour in those who are volume-depleted, initially infusing 250 to 350 mL per hour in those who are not volumedepleted, and adjusting the fluid rate every 1 to 4 hours on the basis of clinical variables.19 The sufficiency of fluid replacement should be carefully monitored by vital signs, urine output, and serum hematocrit.

On the other hand, overly aggressive fluid resuscitation can be detrimental in patients at risk of volume overload or pulmonary edema. Fluid replacement should be tempered in elderly patients and those with cardiac or renal comorbidities, and may require monitoring of central venous pressure.

The ACG and AGA guidelines both recognize the need for early aggressive volume replacement in acute pancreatitis (level of evidence III), but they do not specify the exact amounts and rates. Young and healthy patients should receive a rapid bolus of isotonic saline or Ringer’s lactate solution followed by an infusion at a high initial maintenance rate.

Few studies have been done to assess physicians’ compliance with recommendations for aggressive volume replacement. In an Italian multicenter study, patients with mild or severe acute pancreatitis received an average of only 2.5 L of fluid per day (about 100 mL/hour).20 Gardner et al21 recently summarized the available evidence for fluid support in acute pancreatitis.

NUTRITIONAL SUPPORT

Problem: In many severe cases, enteral or parenteral feeding is not started soon enough.

Nutritional support entails enteral or parenteral feeding when an oral diet is contraindicated. Enteral feeding is usually via a nasojejunal tube, which may need to be placed under endoscopic or radiographic guidance. Neither parenteral nor nasojejunal feeding stimulates pancreatic secretion, and both are safe in acute pancreatitis.

Severe acute pancreatitis is an intensely catabolic state characterized by increased energy expenditure, protein breakdown, and substrate utilization. Patients may not be able to resume an oral diet for weeks or even months, particularly if local complications develop. Early nutritional support has been shown to improve outcomes in severe acute pancreatitis.22 Therefore, nutritional support should be started as soon as possible in severe acute pancreatitis based on initial clinical and radiographic indicators of severity, optimally within the first 2 or 3 days.

Enteral nutrition is preferred to parenteral nutrition in pancreatitis: it is less expensive and does not pose a risk of catheter-related infection or thrombosis or hepatic complications. Also, there is experimental evidence that enteral nutrition may preserve the gut barrier, decreasing mucosal permeability and bacterial translocation.

A number of small randomized trials compared enteral and parenteral nutrition in acute pancreatitis, but they yielded mixed results. A meta-analysis of six trials showed a lower rate of infectious complications with enteral than with parenteral nutrition. 23 However, no significant difference was found in the rates of death or noninfectious complications.

Recommendation: Enteral feeding, when possible

Nutritional support is unnecessary in most cases of mild acute pancreatitis. Pancreatic inflammation typically resolves within a few days, allowing patients to resume eating. Occasionally, patients in whom pain resolves slowly and who fast for more than 5 to 7 days need nutritional support to prevent proteincalorie malnutrition.

The ACG guidelines1 and most others suggest that, whenever possible, enteral rather than parenteral feeding should be given to those who require nutritional support. The level of evidence is II (“strong evidence from at least one published properly designed randomized controlled trial of appropriate size and in an appropriate clinical setting”).

However, not all physicians recognize the benefit of enteral feeding. In a cohort of German gastroenterologists, only 73% favored enteral over parenteral feeding in acute pancreatitis.7

COMPUTED TOMOGRAPHY

Problem: CT is not done in many patients with severe acute pancreatitis, or it is done too soon during the admission.

Dual-phase, contrast-enhanced, pancreatic-protocol CT provides a sensitive structural evaluation of the pancreas and is useful to diagnose necrotizing pancreatitis. Pancreatic necrosis is correlated with a severe clinical course, the development of single or multiorgan dysfunction, and death.

Necrosis is diagnosed when more than 30% of the pancreas does not enhance (ie, perfuse) after intravenous contrast is given. The Balthazar-Ranson CT severity index includes the degree of pancreatic enlargement and inflammation, presence and number of fluid collections, and degree of necrosis (Table 2).24

Recommendation: CT in severe cases

Not every patient with acute pancreatitis needs to undergo CT. Most mild cases do not require routine CT, since necrosis and other local complications are infrequent in this group.

Also, CT is often ordered too soon during the hospitalization. Indicators of severity on CT are not usually evident until 2 to 3 days after admission.25 CT should be considered about 3 days after the onset of symptoms rather than immediately upon admission.

On the other hand, CT at the time of admission may be warranted to rule out other life-threatening causes of abdominal pain and hyperamylasemia (eg, bowel obstruction, viscus perforation). CT may also be useful in the late phase of acute pancreatitis (weeks after admission) to diagnose or monitor complications (eg, pseudocysts, abscesses, splenic vein thrombosis, splenic artery pseudoaneurysms). Magnetic resonance imaging with gadolinium contrast is a reasonable alternative to CT for detecting pancreatic necrosis and other local complications.

In patients who have severe acute pancreatitis and compromised renal function (serum creatinine > 1.5 mg/dL), CT can be performed without contrast to assess severity based on a limited Balthazar score (ie, without a necrosis score). Studies in rats suggest that iodinated contrast may decrease pancreatic microcirculation and worsen or precipitate necrosis,26 although published human studies do not support this contention.27,28

Guidelines uniformly recommend CT for patients with severe acute pancreatitis (the ACG guideline gives it a level of evidence of III), but this recommendation is not always followed. A study from Australia showed that CT was done in only 27% to 67% of patients with severe acute pancreatitis.5 In a British study, only 8 of 46 patients with clinically predicted severe pancreatitis underwent CT within the first 10 days of admission.4

 

 

SUSPECTED INFECTED NECROSIS

Problem: Fine-needle aspiration is not done in many cases of suspected infected necrosis.

Approximately one-third of patients with necrotizing pancreatitis develop infected necrosis. The death rate for patients with infected pancreatic necrosis is high—30%, compared with 12% in those with sterile necrosis.1 Differentiating sterile and infected necrosis is therefore essential.

Clinical signs such as fever are poor predictors of infection. Signs of SIRS can be present in both sterile and infected necrotizing pancreatitis.

Recommendation: Fine-needle aspiration of necrosis

For the reasons given above, the findings of necrosis on CT and persistent SIRS should prompt consideration of fine-needle aspiration with Gram stain and culture to differentiate sterile and infected necrosis (ACG guideline, level of evidence III).1 If infection is confirmed, surgical debridement should be strongly considered. Other less-invasive approaches such as endoscopic debridement can be used in selected cases.

Fine-needle aspiration of necrosis is too often neglected. In a cohort of German surgeons, only 55% complied with International Association of Pancreatology recommendations to perform biopsy to differentiate sterile from infected necrosis in patients with signs of sepsis.29

BROAD-SPECTRUM ANTIBIOTICS

Problem: Broad-spectrum antibiotics are often used inappropriately in patients with mild acute pancreatitis and in patients with sterile necrotizing pancreatitis who are clinically stable and have no signs of sepsis.

Antibiotics are not indicated in mild acute pancreatitis. A limited course of antibiotics is typically indicated in severe cases with suspected or proven infected necrosis (in conjunction with surgical necrosectomy). However, the use of antibiotics in sterile necrosis has been very controversial.

At least six small, nonblinded, randomized trials have evaluated the benefit of giving antibiotics prophylactically for presumed sterile necrosis. A recent Cochrane analysis of five of these trials (294 patients) suggested that patients who got antibiotics had a lower risk of death (odds ratio 0.37, 95% confidence interval [CI] 0.17–0.83) but no difference in the rates of pancreatic infection or surgery.30 These paradoxical results suggest that antibiotics may prevent death by preventing nonpancreatic infections (eg, pneumonia, line infections) rather than by preventing infection of necrotic pancreatic tissue. The five trials in the meta-analysis are limited by significant methodologic heterogeneity and by lack of double-blinding.

In spite of the overall lower death rate observed in the meta-analysis, the prophylactic use of antibiotics in sterile necrosis remains controversial. One concern is that patients given long prophylactic courses of antibiotics may develop resistant bacterial or fungal infections. However, the Cochrane and other meta-analyses have not shown a higher rate of fungal infections in those given antibiotics.31

Recommendation: No routine antibiotics for mild cases

The AGA guidelines recommend against routinely giving antibiotics in mild acute pancreatitis and do not provide strict recommendations for prophylactic antibiotic use in necrotizing acute pancreatitis.2 The guidelines state that antibiotics can be used “on demand” based on clinical signs of infection (eg, high fevers, rising leukocytosis, hypotension) or worsening organ failure.

If a purely prophylactic strategy is used, only patients at high risk of developing infection (eg, those with necrosis in more than 30% of the pancreas) should receive antibiotics. Antibiotics with high tissue-penetration should be used, such as imipenem-cilastin (Primaxin IV) or ciprofloxacin (Cipro) plus metronidazole (Flagyl).

Adherence to these guidelines is not optimal. For example, in an Italian multicenter study, 9% of patients with mild acute pancreatitis were treated with antibiotics.19 Moreover, many patients with proven infected necrosis received antibiotics that do not penetrate the pancreatic tissue very well.

 

 

ERCP IN SEVERE BILIARY ACUTE PANCREATITIS

Problem: Endoscopic retrograde cholangiopancreatography (ERCP) often is performed inappropriately in mild biliary acute pancreatitis or is not performed urgently in severe cases.

In most cases of mild biliary pancreatitis, the stones pass spontaneously, as verified by cholangiography done during laparoscopic cholecystectomy. Ongoing ampullary obstruction by impacted biliary stones can perpetuate pancreatic inflammation and delay recovery.

Two early randomized trials showed a benefit from early ERCP (within 72 hours) with sphincterotomy and stone extraction, primarily in those with severe biliary acute pancreatitis or ascending cholangitis,32,33 but a third trial failed to reveal a benefit.34 A Cochrane metaanalysis of these three trials failed to show a lower death rate with ERCP in mild or severe biliary pancreatitis.35 However, early ERCP did prevent complications in severe biliary pancreatitis (odds ratio 0.27, 95% CI 0.14–0.53).

Later, a fourth randomized trial was restricted to patients with suspected biliary pancreatitis, evidence of biliary obstruction, and no signs of cholangitis36: 103 patients were randomized to undergo either ERCP within 72 hours or conservative management. No difference was observed in rates of death or organ failure or in the CT severity index.

Recommendation: ER CP for suspected retained stones

ERCP has a limited role in patients with biliary pancreatitis, being used to clear retained bile duct stones or to relieve ongoing biliary obstruction.

The decision to perform ERCP before surgery should be based on how strongly one suspects retained stones. ERCP is most appropriate if the suspicion of retained stones and the likelihood of therapeutic intervention are high (eg, if the serum bilirubin and alkaline phosphatase levels are rising and ultrasonography shows a dilated bile duct). If there is moderate suspicion, a safer and less-invasive imaging study such as magnetic resonance cholangiopancreatography (MRCP) or endoscopic ultrasonography can be done to screen for bile duct stones before proceeding to ERCP.

The ACG guidelines suggest urgent ERCP (preferably within 24 hours) for those with severe biliary pancreatitis complicated by organ failure or those with suspicion of cholangitis. The level of evidence is I, ie, “strong evidence from at least one published systematic review of multiple well-designed randomized controlled trials.”1

Elective ERCP is recommended for those who are poor surgical candidates. ERCP is also recommended for those with rising liver enzyme values or imaging findings suggesting a retained common bile duct stone (including intraoperative cholangiography). Endoscopic ultrasonography or MRCP is recommended for those with slow clinical resolution, who are pregnant, or in whom uncertainty exists regarding the biliary etiology of pancreatitis.

Compliance rates with these and similar guidelines are not adequate. In an audit of adherence to the British Society of Gastroenterology guidelines, early ERCP was performed in only 25% of patients with severe biliary acute pancreatitis.6

LAPAROSCOPIC CHOLECYSTECTOMY FOR MILD BILIARY PANCREATITIS

Problem: Laparoscopic cholecystectomy is not done at admission or within 2 weeks in many patients with mild biliary pancreatitis.

If the gallbladder is not removed, biliary pancreatitis may recur in up to 61% of patients within 6 weeks of hospital discharge.37 This is the basis for guideline recommendations for surgery (or a confirmation of a surgery date) prior to hospital discharge.

The International Association of Pancreatology recommends early cholecystectomy (preferably during the same hospitalization) for patients with mild gallstone-associated acute pancreatitis.38 In severe gallstone-associated acute pancreatitis, cholecystectomy should be delayed until there is sufficient resolution of the inflammatory response and clinical recovery. The AGA guidelines advocate cholecystectomy as soon as possible and in no case later than 4 weeks after discharge to prevent relapse. ERCP with biliary sphinc-terotomy may also protect against relapse in those who are not fit to undergo surgery.

Recommendations for definitive management of gallstones (laparoscopic cholecystectomy or ERCP, or both) are not always followed. For example, a British study showed 70% compliance with this recommendation.4 A similar compliance audit in Germany revealed that cholecystectomy was performed during the initial hospital stay in only 23% of cases.7 In a New Zealand study, a regular compliance audit with feedback to surgeons resulted in an increase in the early cholecystectomy rate from 54% to 80%.8

References
  1. Banks PA, Freeman ML; Practice Parameters Committee of the American College of Gastroenterology. Practice guidelines in acute pancreatitis. Am J Gastroenterol 2006; 101:23792400.
  2. Forsmark CE, Baillie J; AGA Institute Clinical Practice and Economics Committee. AGA Institute technical review on acute pancreatitis. Gastroenterology 2007; 132:20222044.
  3. United Kingdom guidelines for the management of acute pancreatitis. British Society of Gastroenterology. Gut 1998; 42(suppl 2):S1S13.
  4. Norton SA, Cheruvu CV, Collins J, Dix FP, Eyre-Brook IA. An assessment of clinical guidelines for the management of acute pancreatitis. Ann R Coll Surg Engl 2001; 83:399405.
  5. Chiang DT, Anozie A, Fleming WR, Kiroff GK. Comparative study on acute pancreatitis management. ANZ J Surg 2004; 74:218221.
  6. Barnard J, Siriwardena AK. Variations in implementation of current national guidelines for the treatment of acute pancreatitis: implications for acute surgical service provision. Ann R Coll Surg Engl 2002; 84:7981.
  7. Lankisch PG, Weber-Dany B, Lerch MM. Clinical perspectives in pancreatology: compliance with acute pancreatitis in Germany [letter]. Pancreatology 2005; 5:591593.
  8. Connor SJ, Lienert AR, Brown LA, Bagshaw PF. Closing the audit loop is necessary to achieve compliance with evidence-based guidelines in the management of acute pancreatitis. N Z Med J 2008; 121:1925.
  9. Mofidi R, Duff MD, Wigmore SJ, Madhavan KK, Garden OJ, Parks RW. Association between early systemic inflammatory response, severity of multiorgan dysfunction, and death in acute pancreatitis. Br J Surg 2006; 93:738744.
  10. Brown A, Orav J, Banks PA. Hemoconcentration is an early marker for organ failure and necrotizing pancreatitis. Pancreas 2000; 20:367372.
  11. Lankisch PG, Mahlke R, Blum T, et al. Hemoconcentration: an early marker of severe and/or necrotizing pancreatitis? A critical appraisal. Am J Gastroenterol 2001; 96:20812085.
  12. Singh VK, Wu BU, Bollen TL, et al. A prospective evaluation of the bedside index for severity in acute pancreatitis score in assessing mortality and intermediate markers of severity in acute pancreatitis. Am J Gastroenterol 2009; 104:966971.
  13. Ranson JH, Rifkind KM, Roses DF, Fink SD, Eng K, Spencer FC. Prognostic signs and the role of operative management in acute pancreatitis. Surg Gynecol Obstet 1974; 139:6981.
  14. Larvin M. Assessment of clinical severity and prognosis. In:Beger HG, Warshaw AL, Buchler MW, et al, editors. The Pancreas. Blackwell Science: New York, 1998:489502.
  15. Wu BU, Johannes RS, Sun X, Tabak Y, Conwell DL, Banks PA. The early prediction of mortality in acute pancreatitis: a large population-based study. Gut 2008; 57:16981703.
  16. Bradley EL. A clinically based classification system for acute pancreatitis. Summary of the International Symposium on Acute Pancreatitis, Atlanta, GA, September 11 through 13, 1992. Arch Surg 1993, 128:586590.
  17. Forgacs B, Eible G, Faulhaber J, Kahrau S, Buhr H, Foitzik T. Effect of fluid resuscitation with and without endothelin A receptor blockade on hemoconcentration and organ function in experimental pancreatitis. Eur Surg Res 2000; 32:162168.
  18. Brown A, Baillargeon JD, Hughes MD, Banks PA. Can fluid resuscitation prevent pancreatic necrosis in severe acute pancreatitis? Pancreatology 2002; 2:104107.
  19. Pandol SJ, Saluja AK, Imrie CW, Banks PA. Acute pancreatitis: bench to the bedside. Gastroenterology 2007; 132:11271151.
  20. Pezzilli R, Uomo G, Gabbrielli A, et al; ProInf-AISP Study Group. A prospective multicenter survey on the treatment of acute pancreatitis in Italy. Dig Liver Dis 2007; 39:838846.
  21. Gardner TB, Vege SS, Pearson RK, Chari ST. Fluid resuscitation in acute pancreatitis. Clin Gastroenterol Hepatol 2008; 6:10701076.
  22. Petrov MS, Pylypchuk RD, Emelyanov NV. Systematic review: nutritional support in acute pancreatitis. Aliment Pharmacol Ther 2008; 28:704712.
  23. Marik PE, Zaloga GP. Meta-analysis of parenteral nutrition versus enteral nutrition in patients with acute pancreatitis. BMJ 2004; 328:1407.
  24. Balthazar EJ, Robinson DL, Megibow AJ, Ranson JH. Acute pancreatitis: value of CT in establishing prognosis. Radiology 1990; 174:331336.
  25. Balthazar EJ. Acute pancreatitis: assessment of severity with clinical and CT evaluation. Radiology 2002; 223:603613.
  26. Foitzik T, Bassi DG, Schmidt J, et al. Intravenous contrast medium accentuates the severity of acute necrotizing pancreatitis in the rat. Gastroenterology 1994; 106:207214.
  27. Carmona-Sanchez R, Uscanga L, Bezaury-Rivas P, Robles-Díaz G, Suazo-Barahona J, Vargas-Vorácková F. Potential harmful effect of iodinated intravenous contrast medium on the clinical course of mild acute pancreatitis. Arch Surg 2000; 135:12801284.
  28. Uhl W, Roggo A, Kirschstein T, et al. Influence of contrast-enhanced computed tomography on couse and outcome in patients with acute pancreatitis. Pancreas 2002; 24:191197.
  29. Foitzik T, Klar E. Non-compliance with guidelines for the management of severe acute pancreatitis among German surgeons. Pancreatology 2007; 7:8085.
  30. Villatoro E, Bassi C, Larvin M. Antibiotic therapy for prophylaxis against infection of pancreatic necrosis in acute pancreatitis. Cochrane Database Syst Rev 2006;CD002941.
  31. Heinrich S, Schafer M, Rousson V, Clavien PA. Evidence-based treatment of acute pancreatitis: a look at established paradigms. Ann Surg 2006; 243:154168.
  32. Neoptolemos JP, Carr-Locke DL, London NJ, Bailey IA, James D, Fossard DP. Controlled trial of urgent endoscopic retrograde cholangiopancreatography and endoscopic sphincterotomy versus conservative treatment for acute pancreatitis due to gallstones. Lancet 1988; 2:979983.
  33. Fan ST, Lai EC, Mok FP, Lo CM, Zheng SS, Wong J. Early treatment of acute biliary pancreatitis by endoscopic papillotomy. N Engl J Med 1993; 328:228232.
  34. Folsch UR, Nitsche R, Ludtke R, Hilgers RA, Creutzfeldt W. Early ERCP and papillotomy compared with conservative treatment for acute biliary pancreatitis. The German Study Group on Acute Biliary Pancreatitis. N Engl J Med 1997; 336:237242.
  35. Ayub K, Imada R, Slavin J. Endoscopic retrograde cholangiopancreatography in gallstone associated pancreatitis. Cochrane Database Syst Rev 2004;CD003630
  36. Oria A, Cimmino D, Ocampo C, et al. Early endoscopic intervention versus early conservative management in patients with acute gallstone pancreatitis and biliopancreatic obstruction. A randomized clinical trial. Ann Surg 2007; 245:1017.
  37. Frei GJ, Frei VT, Thirlby RC, McClelland RN. Biliary pancreatitis: clinical presentation and surgical management. Am J Surg 1986; 151:170175.
  38. Uhl W, Warshaw A, Imrie C, et al; International Association of Pancreatology. IAP guidelines on the surgical management of acute pancreatitis. Pancreatology 2002; 2:565573.
Article PDF
Author and Disclosure Information

Tyler Stevens, MD
Digestive Disease Institute, Cleveland Clinic

Mansour A. Parsi, MD
Digestive Disease Institute, Cleveland Clinic

R. Matthew Walsh, MD
The Rich Family Distinguished Chair in Digestive Diseases, Department of Hepatic-Pancreatic-Biliary and Transplant Surgery, Digestive Disease Institute, Cleveland Clinic

Adddress: Tyler Stevens, MD, Department of Gastroenterology and Hepatology, A31, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail [email protected]

Issue
Cleveland Clinic Journal of Medicine - 76(12)
Publications
Topics
Page Number
697-704
Sections
Author and Disclosure Information

Tyler Stevens, MD
Digestive Disease Institute, Cleveland Clinic

Mansour A. Parsi, MD
Digestive Disease Institute, Cleveland Clinic

R. Matthew Walsh, MD
The Rich Family Distinguished Chair in Digestive Diseases, Department of Hepatic-Pancreatic-Biliary and Transplant Surgery, Digestive Disease Institute, Cleveland Clinic

Adddress: Tyler Stevens, MD, Department of Gastroenterology and Hepatology, A31, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail [email protected]

Author and Disclosure Information

Tyler Stevens, MD
Digestive Disease Institute, Cleveland Clinic

Mansour A. Parsi, MD
Digestive Disease Institute, Cleveland Clinic

R. Matthew Walsh, MD
The Rich Family Distinguished Chair in Digestive Diseases, Department of Hepatic-Pancreatic-Biliary and Transplant Surgery, Digestive Disease Institute, Cleveland Clinic

Adddress: Tyler Stevens, MD, Department of Gastroenterology and Hepatology, A31, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail [email protected]

Article PDF
Article PDF

Several major gastroenterological and surgical societies have issued guidelines on how to manage acute pancreatitis, based on evidence from high-quality randomized trials and nonrandomized studies as well as on expert opinion.1–3 Information is limited on how well physicians in the United States comply with these guidelines, but compliance is suboptimal in other developed countries, according to several studies,4–8 and we suspect that many US physicians are not following the guidelines either.

Acute pancreatitis is a frequent inpatient diagnosis that internists, gastroenterologists, and surgeons all confront. The most common causes are gallstones and heavy alcohol intake. Its management is typically straightforward: intravenous fluids, analgesia, and nothing by mouth. However, treatment of severe cases can be quite complex, particularly if multiple organ systems are involved or if there are local complications.

The primary aim of this article is to raise awareness of recognized deviations from established recommendations that may lead to adverse patient outcomes.

MEASURING ENZYME LEVELS DAILY ADDS COST BUT LITTLE BENEFIT

Problem: Serum amylase and lipase levels are often needlessly measured every day.

Measuring the serum amylase and lipase levels is useful in diagnosing acute pancreatitis, which requires two of the following three features1:

  • Characteristic abdominal pain
  • Levels of serum amylase or serum lipase, or both, that are three or more times the upper limit of normal
  • Findings of acute pancreatitis on computed tomography (CT).

However, the magnitude or duration of the serum enzyme elevation does not correlate with the severity of the attack. Further, we have noticed that physicians at our hospital often order daily serum amylase and lipase levels in patients admitted with acute pancreatitis.

The American College of Gastroenterology (ACG) guidelines1 state that daily monitoring of amylase and lipase has limited value in managing acute pancreatitis. Rechecking these concentrations may be reasonable if pain fails to resolve or worsens during a prolonged hospitalization, as this may suggest a recurrent attack of acute pancreatitis or a developing pseudocyst. But in most cases of acute pancreatitis, daily serum enzyme measurements add cost but little benefit.

REGULAR ASSESSMENT IS IMPORTANT

Problem: Often, severity assessments are not performed regularly or acted on.

Most cases of acute pancreatitis are mild, with rapid recovery and excellent prognosis. However, 15% to 20% are severe and may result in a prolonged hospitalization, systemic inflammatory response syndrome (SIRS), multiorgan system failure, and death.

In severe acute pancreatitis, as pancreatic enzymes and inflammatory cytokines damage the blood vessels, a vast amount of fluid leaks out into the interstitial (“third”) space. This fluid extravasation leads to decreased effective circulating volume, local pancreatic necrosis, hemodynamic instability, and end-organ failure.

It is important to recognize severe acute pancreatitis early because the patient needs to be transferred to a step-down unit or intensive care unit to receive optimal fluid resuscitation and supportive care for organ dysfunction. After 48 to 72 hours, a prediction of severe acute pancreatitis should also prompt the physician to order CT to detect pancreatic necrosis, and also to initiate nutritional support.

Assessment of severity begins in the emergency room or on admission to the hospital. Older age, obesity, organ failure, and pulmonary infiltrates or pleural effusions are initial indicators of poor prognosis. Signs of SIRS (high or low core body temperature, tachycardia, tachypnea, low or high peripheral white blood cell count) or organ failure (eg, elevated serum creatinine) are present on admission in 21% of patients with acute pancreatitis.9

Hemoconcentration is a marker of decreased effective circulating volume in severe acute pancreatitis. A hematocrit higher than 44% at admission or that rises in the first 24 to 48 hours of admission predicts necrosis.10,11 However, a more robust marker of organ failure may be the blood urea nitrogen level.12

 

 

Clinical scoring systems

Several clinical scoring systems have been studied for assessing severity.

The Ranson score is based on 11 clinical factors, 5 checked at admission and 6 checked at 48 hours (Table 1). Patients are at higher risk of death or “serious illness” (needing 7 or more days of intensive care) if they have 3 or more of these factors.13 In a meta-analysis of 12 studies, a Ranson score of 3 or higher had a sensitivity of 75% and a specificity of 77% for predicting severe acute pancreatitis.14

Limitations of the Ranson score are that it can only be completed after 48 hours, all the data points are not always obtained, and it cannot be repeated on a daily basis. Owing to these limitations and its less-than-optimal predictive value, the Ranson score has fallen into disuse.

The APACHE II (Acute Physiology and Chronic Health Evaluation II) score is more versatile. It is based on multiple clinical and laboratory values, and it correlates very well with the risk of death in acute pancreatitis. Death rates are less than 4% when the APACHE II score is less than 8, and 11% to 18% when it is 8 or higher.1 The trajectory of the APACHE II score in the first 48 hours is also an accurate prognostic indicator.

Previous limitations of the APACHE II score were that it was complicated and timeconsuming to calculate and required arterial blood gas measurements. Easy-to-use online calculators are now available (eg, www.globalrph.com/apacheii.htm), and the venous bicarbonate level and the oxygen saturation can be substituted for the arterial pH and oxygen partial pressure.

BISAP, a new five-point scoring system,15 was recently prospectively validated.12 “BISAP” is an acronym for the five markers it is based on, each of which has been shown to predict severe illness in acute pancreatitis:

  • Blood urea nitrogen level > 25 mg/dL
  • Impaired mental status
  • SIRS
  • Age > 60 years
  • Pleural effusion.

The presence of three or more of these factors correlates with higher risk of death, organ failure, and pancreatic necrosis.12

Compared with APACHE II, BISAP has similar accuracy and is easier to calculate. Also, BISAP was specifically developed for acute pancreatitis, whereas APACHE II is a generic score for all critically ill patients.

The Atlanta criteria16 define severe acute pancreatitis as one or more of the following:

  • A Ranson score of 3 or higher during the first 48 hours
  • An APACHE II score of 8 or higher at any time
  • Failure of one or more organs
  • One or more local complications (eg, necrosis, pseudocysts, abscesses).

Recommendation: Assess severity at least daily

A severity assessment should be performed at admission and at least every day thereafter. Clinical guidelines recognize the importance of severity assessment but vary in their specific recommendations.

The ACG advises calculating the APACHE II score within 3 days of admission and measuring the hematocrit at admission, at 12 hours, and at 24 hours. The level of evidence is III, ie, “from published well-designed trials without randomization, single group prepost, cohort, time series, or matched case controlled studies”.1

The American Gastroenterological Association (AGA) provides a more generalized recommendation, that “clinical judgment” should take into account the presence of risk factors (eg, age, obesity), presence or absence of SIRS, routine laboratory values (eg, hematocrit, serum creatinine), and APACHE II score when assessing severity and making decisions.2

In a German survey, only 32% of gastroenterologists used the APACHE II score for assessing risk in acute pancreatitis, in spite of national guidelines emphasizing its importance.7 Also, not all patients with severe acute pancreatitis are transferred to a step-down unit or intensive care unit as recommended. In a British study,4 only 8 (17%) of 46 patients with predicted severe acute pancreatitis were transferred, and 8 of the 38 patients who were not transferred died.

FLUID MUST BE AGGRESSIVELY REPLACED AND MONITORED

Problem: Often, not enough fluid is replaced, or fluid status is not adequately monitored.

Fluid must be aggressively replaced to balance the massive third-space fluid losses that occur in the early inflammatory phase of acute pancreatitis. Intravascular volume depletion can develop rapidly and result in tachycardia, hypotension, and renal failure. It may also impair the blood flow to the pancreas and worsen necrosis.

Animal studies show that aggressive fluid replacement supports the pancreatic microcirculation and prevents necrosis.17 It may also support the intestinal microcirculation and gut barrier, preventing bacterial translocation.

In humans, no controlled trials have been done to test the efficacy of aggressive fluid resuscitation in acute pancreatitis. However, the notion that intravascular fluid loss contributes to poor outcomes is inferred from human studies showing more necrosis and deaths in patients with hemoconcentration. In one study, patients who received inadequate fluid replacement (evidenced by a rise in hematocrit at 24 hours) were more likely to develop necrotizing pancreatitis.18

 

 

Recommendation: Early, aggressive fluid replacement

Experts have suggested initially infusing 500 to 1,000 mL of fluid per hour in those who are volume-depleted, initially infusing 250 to 350 mL per hour in those who are not volumedepleted, and adjusting the fluid rate every 1 to 4 hours on the basis of clinical variables.19 The sufficiency of fluid replacement should be carefully monitored by vital signs, urine output, and serum hematocrit.

On the other hand, overly aggressive fluid resuscitation can be detrimental in patients at risk of volume overload or pulmonary edema. Fluid replacement should be tempered in elderly patients and those with cardiac or renal comorbidities, and may require monitoring of central venous pressure.

The ACG and AGA guidelines both recognize the need for early aggressive volume replacement in acute pancreatitis (level of evidence III), but they do not specify the exact amounts and rates. Young and healthy patients should receive a rapid bolus of isotonic saline or Ringer’s lactate solution followed by an infusion at a high initial maintenance rate.

Few studies have been done to assess physicians’ compliance with recommendations for aggressive volume replacement. In an Italian multicenter study, patients with mild or severe acute pancreatitis received an average of only 2.5 L of fluid per day (about 100 mL/hour).20 Gardner et al21 recently summarized the available evidence for fluid support in acute pancreatitis.

NUTRITIONAL SUPPORT

Problem: In many severe cases, enteral or parenteral feeding is not started soon enough.

Nutritional support entails enteral or parenteral feeding when an oral diet is contraindicated. Enteral feeding is usually via a nasojejunal tube, which may need to be placed under endoscopic or radiographic guidance. Neither parenteral nor nasojejunal feeding stimulates pancreatic secretion, and both are safe in acute pancreatitis.

Severe acute pancreatitis is an intensely catabolic state characterized by increased energy expenditure, protein breakdown, and substrate utilization. Patients may not be able to resume an oral diet for weeks or even months, particularly if local complications develop. Early nutritional support has been shown to improve outcomes in severe acute pancreatitis.22 Therefore, nutritional support should be started as soon as possible in severe acute pancreatitis based on initial clinical and radiographic indicators of severity, optimally within the first 2 or 3 days.

Enteral nutrition is preferred to parenteral nutrition in pancreatitis: it is less expensive and does not pose a risk of catheter-related infection or thrombosis or hepatic complications. Also, there is experimental evidence that enteral nutrition may preserve the gut barrier, decreasing mucosal permeability and bacterial translocation.

A number of small randomized trials compared enteral and parenteral nutrition in acute pancreatitis, but they yielded mixed results. A meta-analysis of six trials showed a lower rate of infectious complications with enteral than with parenteral nutrition. 23 However, no significant difference was found in the rates of death or noninfectious complications.

Recommendation: Enteral feeding, when possible

Nutritional support is unnecessary in most cases of mild acute pancreatitis. Pancreatic inflammation typically resolves within a few days, allowing patients to resume eating. Occasionally, patients in whom pain resolves slowly and who fast for more than 5 to 7 days need nutritional support to prevent proteincalorie malnutrition.

The ACG guidelines1 and most others suggest that, whenever possible, enteral rather than parenteral feeding should be given to those who require nutritional support. The level of evidence is II (“strong evidence from at least one published properly designed randomized controlled trial of appropriate size and in an appropriate clinical setting”).

However, not all physicians recognize the benefit of enteral feeding. In a cohort of German gastroenterologists, only 73% favored enteral over parenteral feeding in acute pancreatitis.7

COMPUTED TOMOGRAPHY

Problem: CT is not done in many patients with severe acute pancreatitis, or it is done too soon during the admission.

Dual-phase, contrast-enhanced, pancreatic-protocol CT provides a sensitive structural evaluation of the pancreas and is useful to diagnose necrotizing pancreatitis. Pancreatic necrosis is correlated with a severe clinical course, the development of single or multiorgan dysfunction, and death.

Necrosis is diagnosed when more than 30% of the pancreas does not enhance (ie, perfuse) after intravenous contrast is given. The Balthazar-Ranson CT severity index includes the degree of pancreatic enlargement and inflammation, presence and number of fluid collections, and degree of necrosis (Table 2).24

Recommendation: CT in severe cases

Not every patient with acute pancreatitis needs to undergo CT. Most mild cases do not require routine CT, since necrosis and other local complications are infrequent in this group.

Also, CT is often ordered too soon during the hospitalization. Indicators of severity on CT are not usually evident until 2 to 3 days after admission.25 CT should be considered about 3 days after the onset of symptoms rather than immediately upon admission.

On the other hand, CT at the time of admission may be warranted to rule out other life-threatening causes of abdominal pain and hyperamylasemia (eg, bowel obstruction, viscus perforation). CT may also be useful in the late phase of acute pancreatitis (weeks after admission) to diagnose or monitor complications (eg, pseudocysts, abscesses, splenic vein thrombosis, splenic artery pseudoaneurysms). Magnetic resonance imaging with gadolinium contrast is a reasonable alternative to CT for detecting pancreatic necrosis and other local complications.

In patients who have severe acute pancreatitis and compromised renal function (serum creatinine > 1.5 mg/dL), CT can be performed without contrast to assess severity based on a limited Balthazar score (ie, without a necrosis score). Studies in rats suggest that iodinated contrast may decrease pancreatic microcirculation and worsen or precipitate necrosis,26 although published human studies do not support this contention.27,28

Guidelines uniformly recommend CT for patients with severe acute pancreatitis (the ACG guideline gives it a level of evidence of III), but this recommendation is not always followed. A study from Australia showed that CT was done in only 27% to 67% of patients with severe acute pancreatitis.5 In a British study, only 8 of 46 patients with clinically predicted severe pancreatitis underwent CT within the first 10 days of admission.4

 

 

SUSPECTED INFECTED NECROSIS

Problem: Fine-needle aspiration is not done in many cases of suspected infected necrosis.

Approximately one-third of patients with necrotizing pancreatitis develop infected necrosis. The death rate for patients with infected pancreatic necrosis is high—30%, compared with 12% in those with sterile necrosis.1 Differentiating sterile and infected necrosis is therefore essential.

Clinical signs such as fever are poor predictors of infection. Signs of SIRS can be present in both sterile and infected necrotizing pancreatitis.

Recommendation: Fine-needle aspiration of necrosis

For the reasons given above, the findings of necrosis on CT and persistent SIRS should prompt consideration of fine-needle aspiration with Gram stain and culture to differentiate sterile and infected necrosis (ACG guideline, level of evidence III).1 If infection is confirmed, surgical debridement should be strongly considered. Other less-invasive approaches such as endoscopic debridement can be used in selected cases.

Fine-needle aspiration of necrosis is too often neglected. In a cohort of German surgeons, only 55% complied with International Association of Pancreatology recommendations to perform biopsy to differentiate sterile from infected necrosis in patients with signs of sepsis.29

BROAD-SPECTRUM ANTIBIOTICS

Problem: Broad-spectrum antibiotics are often used inappropriately in patients with mild acute pancreatitis and in patients with sterile necrotizing pancreatitis who are clinically stable and have no signs of sepsis.

Antibiotics are not indicated in mild acute pancreatitis. A limited course of antibiotics is typically indicated in severe cases with suspected or proven infected necrosis (in conjunction with surgical necrosectomy). However, the use of antibiotics in sterile necrosis has been very controversial.

At least six small, nonblinded, randomized trials have evaluated the benefit of giving antibiotics prophylactically for presumed sterile necrosis. A recent Cochrane analysis of five of these trials (294 patients) suggested that patients who got antibiotics had a lower risk of death (odds ratio 0.37, 95% confidence interval [CI] 0.17–0.83) but no difference in the rates of pancreatic infection or surgery.30 These paradoxical results suggest that antibiotics may prevent death by preventing nonpancreatic infections (eg, pneumonia, line infections) rather than by preventing infection of necrotic pancreatic tissue. The five trials in the meta-analysis are limited by significant methodologic heterogeneity and by lack of double-blinding.

In spite of the overall lower death rate observed in the meta-analysis, the prophylactic use of antibiotics in sterile necrosis remains controversial. One concern is that patients given long prophylactic courses of antibiotics may develop resistant bacterial or fungal infections. However, the Cochrane and other meta-analyses have not shown a higher rate of fungal infections in those given antibiotics.31

Recommendation: No routine antibiotics for mild cases

The AGA guidelines recommend against routinely giving antibiotics in mild acute pancreatitis and do not provide strict recommendations for prophylactic antibiotic use in necrotizing acute pancreatitis.2 The guidelines state that antibiotics can be used “on demand” based on clinical signs of infection (eg, high fevers, rising leukocytosis, hypotension) or worsening organ failure.

If a purely prophylactic strategy is used, only patients at high risk of developing infection (eg, those with necrosis in more than 30% of the pancreas) should receive antibiotics. Antibiotics with high tissue-penetration should be used, such as imipenem-cilastin (Primaxin IV) or ciprofloxacin (Cipro) plus metronidazole (Flagyl).

Adherence to these guidelines is not optimal. For example, in an Italian multicenter study, 9% of patients with mild acute pancreatitis were treated with antibiotics.19 Moreover, many patients with proven infected necrosis received antibiotics that do not penetrate the pancreatic tissue very well.

 

 

ERCP IN SEVERE BILIARY ACUTE PANCREATITIS

Problem: Endoscopic retrograde cholangiopancreatography (ERCP) often is performed inappropriately in mild biliary acute pancreatitis or is not performed urgently in severe cases.

In most cases of mild biliary pancreatitis, the stones pass spontaneously, as verified by cholangiography done during laparoscopic cholecystectomy. Ongoing ampullary obstruction by impacted biliary stones can perpetuate pancreatic inflammation and delay recovery.

Two early randomized trials showed a benefit from early ERCP (within 72 hours) with sphincterotomy and stone extraction, primarily in those with severe biliary acute pancreatitis or ascending cholangitis,32,33 but a third trial failed to reveal a benefit.34 A Cochrane metaanalysis of these three trials failed to show a lower death rate with ERCP in mild or severe biliary pancreatitis.35 However, early ERCP did prevent complications in severe biliary pancreatitis (odds ratio 0.27, 95% CI 0.14–0.53).

Later, a fourth randomized trial was restricted to patients with suspected biliary pancreatitis, evidence of biliary obstruction, and no signs of cholangitis36: 103 patients were randomized to undergo either ERCP within 72 hours or conservative management. No difference was observed in rates of death or organ failure or in the CT severity index.

Recommendation: ER CP for suspected retained stones

ERCP has a limited role in patients with biliary pancreatitis, being used to clear retained bile duct stones or to relieve ongoing biliary obstruction.

The decision to perform ERCP before surgery should be based on how strongly one suspects retained stones. ERCP is most appropriate if the suspicion of retained stones and the likelihood of therapeutic intervention are high (eg, if the serum bilirubin and alkaline phosphatase levels are rising and ultrasonography shows a dilated bile duct). If there is moderate suspicion, a safer and less-invasive imaging study such as magnetic resonance cholangiopancreatography (MRCP) or endoscopic ultrasonography can be done to screen for bile duct stones before proceeding to ERCP.

The ACG guidelines suggest urgent ERCP (preferably within 24 hours) for those with severe biliary pancreatitis complicated by organ failure or those with suspicion of cholangitis. The level of evidence is I, ie, “strong evidence from at least one published systematic review of multiple well-designed randomized controlled trials.”1

Elective ERCP is recommended for those who are poor surgical candidates. ERCP is also recommended for those with rising liver enzyme values or imaging findings suggesting a retained common bile duct stone (including intraoperative cholangiography). Endoscopic ultrasonography or MRCP is recommended for those with slow clinical resolution, who are pregnant, or in whom uncertainty exists regarding the biliary etiology of pancreatitis.

Compliance rates with these and similar guidelines are not adequate. In an audit of adherence to the British Society of Gastroenterology guidelines, early ERCP was performed in only 25% of patients with severe biliary acute pancreatitis.6

LAPAROSCOPIC CHOLECYSTECTOMY FOR MILD BILIARY PANCREATITIS

Problem: Laparoscopic cholecystectomy is not done at admission or within 2 weeks in many patients with mild biliary pancreatitis.

If the gallbladder is not removed, biliary pancreatitis may recur in up to 61% of patients within 6 weeks of hospital discharge.37 This is the basis for guideline recommendations for surgery (or a confirmation of a surgery date) prior to hospital discharge.

The International Association of Pancreatology recommends early cholecystectomy (preferably during the same hospitalization) for patients with mild gallstone-associated acute pancreatitis.38 In severe gallstone-associated acute pancreatitis, cholecystectomy should be delayed until there is sufficient resolution of the inflammatory response and clinical recovery. The AGA guidelines advocate cholecystectomy as soon as possible and in no case later than 4 weeks after discharge to prevent relapse. ERCP with biliary sphinc-terotomy may also protect against relapse in those who are not fit to undergo surgery.

Recommendations for definitive management of gallstones (laparoscopic cholecystectomy or ERCP, or both) are not always followed. For example, a British study showed 70% compliance with this recommendation.4 A similar compliance audit in Germany revealed that cholecystectomy was performed during the initial hospital stay in only 23% of cases.7 In a New Zealand study, a regular compliance audit with feedback to surgeons resulted in an increase in the early cholecystectomy rate from 54% to 80%.8

Several major gastroenterological and surgical societies have issued guidelines on how to manage acute pancreatitis, based on evidence from high-quality randomized trials and nonrandomized studies as well as on expert opinion.1–3 Information is limited on how well physicians in the United States comply with these guidelines, but compliance is suboptimal in other developed countries, according to several studies,4–8 and we suspect that many US physicians are not following the guidelines either.

Acute pancreatitis is a frequent inpatient diagnosis that internists, gastroenterologists, and surgeons all confront. The most common causes are gallstones and heavy alcohol intake. Its management is typically straightforward: intravenous fluids, analgesia, and nothing by mouth. However, treatment of severe cases can be quite complex, particularly if multiple organ systems are involved or if there are local complications.

The primary aim of this article is to raise awareness of recognized deviations from established recommendations that may lead to adverse patient outcomes.

MEASURING ENZYME LEVELS DAILY ADDS COST BUT LITTLE BENEFIT

Problem: Serum amylase and lipase levels are often needlessly measured every day.

Measuring the serum amylase and lipase levels is useful in diagnosing acute pancreatitis, which requires two of the following three features1:

  • Characteristic abdominal pain
  • Levels of serum amylase or serum lipase, or both, that are three or more times the upper limit of normal
  • Findings of acute pancreatitis on computed tomography (CT).

However, the magnitude or duration of the serum enzyme elevation does not correlate with the severity of the attack. Further, we have noticed that physicians at our hospital often order daily serum amylase and lipase levels in patients admitted with acute pancreatitis.

The American College of Gastroenterology (ACG) guidelines1 state that daily monitoring of amylase and lipase has limited value in managing acute pancreatitis. Rechecking these concentrations may be reasonable if pain fails to resolve or worsens during a prolonged hospitalization, as this may suggest a recurrent attack of acute pancreatitis or a developing pseudocyst. But in most cases of acute pancreatitis, daily serum enzyme measurements add cost but little benefit.

REGULAR ASSESSMENT IS IMPORTANT

Problem: Often, severity assessments are not performed regularly or acted on.

Most cases of acute pancreatitis are mild, with rapid recovery and excellent prognosis. However, 15% to 20% are severe and may result in a prolonged hospitalization, systemic inflammatory response syndrome (SIRS), multiorgan system failure, and death.

In severe acute pancreatitis, as pancreatic enzymes and inflammatory cytokines damage the blood vessels, a vast amount of fluid leaks out into the interstitial (“third”) space. This fluid extravasation leads to decreased effective circulating volume, local pancreatic necrosis, hemodynamic instability, and end-organ failure.

It is important to recognize severe acute pancreatitis early because the patient needs to be transferred to a step-down unit or intensive care unit to receive optimal fluid resuscitation and supportive care for organ dysfunction. After 48 to 72 hours, a prediction of severe acute pancreatitis should also prompt the physician to order CT to detect pancreatic necrosis, and also to initiate nutritional support.

Assessment of severity begins in the emergency room or on admission to the hospital. Older age, obesity, organ failure, and pulmonary infiltrates or pleural effusions are initial indicators of poor prognosis. Signs of SIRS (high or low core body temperature, tachycardia, tachypnea, low or high peripheral white blood cell count) or organ failure (eg, elevated serum creatinine) are present on admission in 21% of patients with acute pancreatitis.9

Hemoconcentration is a marker of decreased effective circulating volume in severe acute pancreatitis. A hematocrit higher than 44% at admission or that rises in the first 24 to 48 hours of admission predicts necrosis.10,11 However, a more robust marker of organ failure may be the blood urea nitrogen level.12

 

 

Clinical scoring systems

Several clinical scoring systems have been studied for assessing severity.

The Ranson score is based on 11 clinical factors, 5 checked at admission and 6 checked at 48 hours (Table 1). Patients are at higher risk of death or “serious illness” (needing 7 or more days of intensive care) if they have 3 or more of these factors.13 In a meta-analysis of 12 studies, a Ranson score of 3 or higher had a sensitivity of 75% and a specificity of 77% for predicting severe acute pancreatitis.14

Limitations of the Ranson score are that it can only be completed after 48 hours, all the data points are not always obtained, and it cannot be repeated on a daily basis. Owing to these limitations and its less-than-optimal predictive value, the Ranson score has fallen into disuse.

The APACHE II (Acute Physiology and Chronic Health Evaluation II) score is more versatile. It is based on multiple clinical and laboratory values, and it correlates very well with the risk of death in acute pancreatitis. Death rates are less than 4% when the APACHE II score is less than 8, and 11% to 18% when it is 8 or higher.1 The trajectory of the APACHE II score in the first 48 hours is also an accurate prognostic indicator.

Previous limitations of the APACHE II score were that it was complicated and timeconsuming to calculate and required arterial blood gas measurements. Easy-to-use online calculators are now available (eg, www.globalrph.com/apacheii.htm), and the venous bicarbonate level and the oxygen saturation can be substituted for the arterial pH and oxygen partial pressure.

BISAP, a new five-point scoring system,15 was recently prospectively validated.12 “BISAP” is an acronym for the five markers it is based on, each of which has been shown to predict severe illness in acute pancreatitis:

  • Blood urea nitrogen level > 25 mg/dL
  • Impaired mental status
  • SIRS
  • Age > 60 years
  • Pleural effusion.

The presence of three or more of these factors correlates with higher risk of death, organ failure, and pancreatic necrosis.12

Compared with APACHE II, BISAP has similar accuracy and is easier to calculate. Also, BISAP was specifically developed for acute pancreatitis, whereas APACHE II is a generic score for all critically ill patients.

The Atlanta criteria16 define severe acute pancreatitis as one or more of the following:

  • A Ranson score of 3 or higher during the first 48 hours
  • An APACHE II score of 8 or higher at any time
  • Failure of one or more organs
  • One or more local complications (eg, necrosis, pseudocysts, abscesses).

Recommendation: Assess severity at least daily

A severity assessment should be performed at admission and at least every day thereafter. Clinical guidelines recognize the importance of severity assessment but vary in their specific recommendations.

The ACG advises calculating the APACHE II score within 3 days of admission and measuring the hematocrit at admission, at 12 hours, and at 24 hours. The level of evidence is III, ie, “from published well-designed trials without randomization, single group prepost, cohort, time series, or matched case controlled studies”.1

The American Gastroenterological Association (AGA) provides a more generalized recommendation, that “clinical judgment” should take into account the presence of risk factors (eg, age, obesity), presence or absence of SIRS, routine laboratory values (eg, hematocrit, serum creatinine), and APACHE II score when assessing severity and making decisions.2

In a German survey, only 32% of gastroenterologists used the APACHE II score for assessing risk in acute pancreatitis, in spite of national guidelines emphasizing its importance.7 Also, not all patients with severe acute pancreatitis are transferred to a step-down unit or intensive care unit as recommended. In a British study,4 only 8 (17%) of 46 patients with predicted severe acute pancreatitis were transferred, and 8 of the 38 patients who were not transferred died.

FLUID MUST BE AGGRESSIVELY REPLACED AND MONITORED

Problem: Often, not enough fluid is replaced, or fluid status is not adequately monitored.

Fluid must be aggressively replaced to balance the massive third-space fluid losses that occur in the early inflammatory phase of acute pancreatitis. Intravascular volume depletion can develop rapidly and result in tachycardia, hypotension, and renal failure. It may also impair the blood flow to the pancreas and worsen necrosis.

Animal studies show that aggressive fluid replacement supports the pancreatic microcirculation and prevents necrosis.17 It may also support the intestinal microcirculation and gut barrier, preventing bacterial translocation.

In humans, no controlled trials have been done to test the efficacy of aggressive fluid resuscitation in acute pancreatitis. However, the notion that intravascular fluid loss contributes to poor outcomes is inferred from human studies showing more necrosis and deaths in patients with hemoconcentration. In one study, patients who received inadequate fluid replacement (evidenced by a rise in hematocrit at 24 hours) were more likely to develop necrotizing pancreatitis.18

 

 

Recommendation: Early, aggressive fluid replacement

Experts have suggested initially infusing 500 to 1,000 mL of fluid per hour in those who are volume-depleted, initially infusing 250 to 350 mL per hour in those who are not volumedepleted, and adjusting the fluid rate every 1 to 4 hours on the basis of clinical variables.19 The sufficiency of fluid replacement should be carefully monitored by vital signs, urine output, and serum hematocrit.

On the other hand, overly aggressive fluid resuscitation can be detrimental in patients at risk of volume overload or pulmonary edema. Fluid replacement should be tempered in elderly patients and those with cardiac or renal comorbidities, and may require monitoring of central venous pressure.

The ACG and AGA guidelines both recognize the need for early aggressive volume replacement in acute pancreatitis (level of evidence III), but they do not specify the exact amounts and rates. Young and healthy patients should receive a rapid bolus of isotonic saline or Ringer’s lactate solution followed by an infusion at a high initial maintenance rate.

Few studies have been done to assess physicians’ compliance with recommendations for aggressive volume replacement. In an Italian multicenter study, patients with mild or severe acute pancreatitis received an average of only 2.5 L of fluid per day (about 100 mL/hour).20 Gardner et al21 recently summarized the available evidence for fluid support in acute pancreatitis.

NUTRITIONAL SUPPORT

Problem: In many severe cases, enteral or parenteral feeding is not started soon enough.

Nutritional support entails enteral or parenteral feeding when an oral diet is contraindicated. Enteral feeding is usually via a nasojejunal tube, which may need to be placed under endoscopic or radiographic guidance. Neither parenteral nor nasojejunal feeding stimulates pancreatic secretion, and both are safe in acute pancreatitis.

Severe acute pancreatitis is an intensely catabolic state characterized by increased energy expenditure, protein breakdown, and substrate utilization. Patients may not be able to resume an oral diet for weeks or even months, particularly if local complications develop. Early nutritional support has been shown to improve outcomes in severe acute pancreatitis.22 Therefore, nutritional support should be started as soon as possible in severe acute pancreatitis based on initial clinical and radiographic indicators of severity, optimally within the first 2 or 3 days.

Enteral nutrition is preferred to parenteral nutrition in pancreatitis: it is less expensive and does not pose a risk of catheter-related infection or thrombosis or hepatic complications. Also, there is experimental evidence that enteral nutrition may preserve the gut barrier, decreasing mucosal permeability and bacterial translocation.

A number of small randomized trials compared enteral and parenteral nutrition in acute pancreatitis, but they yielded mixed results. A meta-analysis of six trials showed a lower rate of infectious complications with enteral than with parenteral nutrition. 23 However, no significant difference was found in the rates of death or noninfectious complications.

Recommendation: Enteral feeding, when possible

Nutritional support is unnecessary in most cases of mild acute pancreatitis. Pancreatic inflammation typically resolves within a few days, allowing patients to resume eating. Occasionally, patients in whom pain resolves slowly and who fast for more than 5 to 7 days need nutritional support to prevent proteincalorie malnutrition.

The ACG guidelines1 and most others suggest that, whenever possible, enteral rather than parenteral feeding should be given to those who require nutritional support. The level of evidence is II (“strong evidence from at least one published properly designed randomized controlled trial of appropriate size and in an appropriate clinical setting”).

However, not all physicians recognize the benefit of enteral feeding. In a cohort of German gastroenterologists, only 73% favored enteral over parenteral feeding in acute pancreatitis.7

COMPUTED TOMOGRAPHY

Problem: CT is not done in many patients with severe acute pancreatitis, or it is done too soon during the admission.

Dual-phase, contrast-enhanced, pancreatic-protocol CT provides a sensitive structural evaluation of the pancreas and is useful to diagnose necrotizing pancreatitis. Pancreatic necrosis is correlated with a severe clinical course, the development of single or multiorgan dysfunction, and death.

Necrosis is diagnosed when more than 30% of the pancreas does not enhance (ie, perfuse) after intravenous contrast is given. The Balthazar-Ranson CT severity index includes the degree of pancreatic enlargement and inflammation, presence and number of fluid collections, and degree of necrosis (Table 2).24

Recommendation: CT in severe cases

Not every patient with acute pancreatitis needs to undergo CT. Most mild cases do not require routine CT, since necrosis and other local complications are infrequent in this group.

Also, CT is often ordered too soon during the hospitalization. Indicators of severity on CT are not usually evident until 2 to 3 days after admission.25 CT should be considered about 3 days after the onset of symptoms rather than immediately upon admission.

On the other hand, CT at the time of admission may be warranted to rule out other life-threatening causes of abdominal pain and hyperamylasemia (eg, bowel obstruction, viscus perforation). CT may also be useful in the late phase of acute pancreatitis (weeks after admission) to diagnose or monitor complications (eg, pseudocysts, abscesses, splenic vein thrombosis, splenic artery pseudoaneurysms). Magnetic resonance imaging with gadolinium contrast is a reasonable alternative to CT for detecting pancreatic necrosis and other local complications.

In patients who have severe acute pancreatitis and compromised renal function (serum creatinine > 1.5 mg/dL), CT can be performed without contrast to assess severity based on a limited Balthazar score (ie, without a necrosis score). Studies in rats suggest that iodinated contrast may decrease pancreatic microcirculation and worsen or precipitate necrosis,26 although published human studies do not support this contention.27,28

Guidelines uniformly recommend CT for patients with severe acute pancreatitis (the ACG guideline gives it a level of evidence of III), but this recommendation is not always followed. A study from Australia showed that CT was done in only 27% to 67% of patients with severe acute pancreatitis.5 In a British study, only 8 of 46 patients with clinically predicted severe pancreatitis underwent CT within the first 10 days of admission.4

 

 

SUSPECTED INFECTED NECROSIS

Problem: Fine-needle aspiration is not done in many cases of suspected infected necrosis.

Approximately one-third of patients with necrotizing pancreatitis develop infected necrosis. The death rate for patients with infected pancreatic necrosis is high—30%, compared with 12% in those with sterile necrosis.1 Differentiating sterile and infected necrosis is therefore essential.

Clinical signs such as fever are poor predictors of infection. Signs of SIRS can be present in both sterile and infected necrotizing pancreatitis.

Recommendation: Fine-needle aspiration of necrosis

For the reasons given above, the findings of necrosis on CT and persistent SIRS should prompt consideration of fine-needle aspiration with Gram stain and culture to differentiate sterile and infected necrosis (ACG guideline, level of evidence III).1 If infection is confirmed, surgical debridement should be strongly considered. Other less-invasive approaches such as endoscopic debridement can be used in selected cases.

Fine-needle aspiration of necrosis is too often neglected. In a cohort of German surgeons, only 55% complied with International Association of Pancreatology recommendations to perform biopsy to differentiate sterile from infected necrosis in patients with signs of sepsis.29

BROAD-SPECTRUM ANTIBIOTICS

Problem: Broad-spectrum antibiotics are often used inappropriately in patients with mild acute pancreatitis and in patients with sterile necrotizing pancreatitis who are clinically stable and have no signs of sepsis.

Antibiotics are not indicated in mild acute pancreatitis. A limited course of antibiotics is typically indicated in severe cases with suspected or proven infected necrosis (in conjunction with surgical necrosectomy). However, the use of antibiotics in sterile necrosis has been very controversial.

At least six small, nonblinded, randomized trials have evaluated the benefit of giving antibiotics prophylactically for presumed sterile necrosis. A recent Cochrane analysis of five of these trials (294 patients) suggested that patients who got antibiotics had a lower risk of death (odds ratio 0.37, 95% confidence interval [CI] 0.17–0.83) but no difference in the rates of pancreatic infection or surgery.30 These paradoxical results suggest that antibiotics may prevent death by preventing nonpancreatic infections (eg, pneumonia, line infections) rather than by preventing infection of necrotic pancreatic tissue. The five trials in the meta-analysis are limited by significant methodologic heterogeneity and by lack of double-blinding.

In spite of the overall lower death rate observed in the meta-analysis, the prophylactic use of antibiotics in sterile necrosis remains controversial. One concern is that patients given long prophylactic courses of antibiotics may develop resistant bacterial or fungal infections. However, the Cochrane and other meta-analyses have not shown a higher rate of fungal infections in those given antibiotics.31

Recommendation: No routine antibiotics for mild cases

The AGA guidelines recommend against routinely giving antibiotics in mild acute pancreatitis and do not provide strict recommendations for prophylactic antibiotic use in necrotizing acute pancreatitis.2 The guidelines state that antibiotics can be used “on demand” based on clinical signs of infection (eg, high fevers, rising leukocytosis, hypotension) or worsening organ failure.

If a purely prophylactic strategy is used, only patients at high risk of developing infection (eg, those with necrosis in more than 30% of the pancreas) should receive antibiotics. Antibiotics with high tissue-penetration should be used, such as imipenem-cilastin (Primaxin IV) or ciprofloxacin (Cipro) plus metronidazole (Flagyl).

Adherence to these guidelines is not optimal. For example, in an Italian multicenter study, 9% of patients with mild acute pancreatitis were treated with antibiotics.19 Moreover, many patients with proven infected necrosis received antibiotics that do not penetrate the pancreatic tissue very well.

 

 

ERCP IN SEVERE BILIARY ACUTE PANCREATITIS

Problem: Endoscopic retrograde cholangiopancreatography (ERCP) often is performed inappropriately in mild biliary acute pancreatitis or is not performed urgently in severe cases.

In most cases of mild biliary pancreatitis, the stones pass spontaneously, as verified by cholangiography done during laparoscopic cholecystectomy. Ongoing ampullary obstruction by impacted biliary stones can perpetuate pancreatic inflammation and delay recovery.

Two early randomized trials showed a benefit from early ERCP (within 72 hours) with sphincterotomy and stone extraction, primarily in those with severe biliary acute pancreatitis or ascending cholangitis,32,33 but a third trial failed to reveal a benefit.34 A Cochrane metaanalysis of these three trials failed to show a lower death rate with ERCP in mild or severe biliary pancreatitis.35 However, early ERCP did prevent complications in severe biliary pancreatitis (odds ratio 0.27, 95% CI 0.14–0.53).

Later, a fourth randomized trial was restricted to patients with suspected biliary pancreatitis, evidence of biliary obstruction, and no signs of cholangitis36: 103 patients were randomized to undergo either ERCP within 72 hours or conservative management. No difference was observed in rates of death or organ failure or in the CT severity index.

Recommendation: ER CP for suspected retained stones

ERCP has a limited role in patients with biliary pancreatitis, being used to clear retained bile duct stones or to relieve ongoing biliary obstruction.

The decision to perform ERCP before surgery should be based on how strongly one suspects retained stones. ERCP is most appropriate if the suspicion of retained stones and the likelihood of therapeutic intervention are high (eg, if the serum bilirubin and alkaline phosphatase levels are rising and ultrasonography shows a dilated bile duct). If there is moderate suspicion, a safer and less-invasive imaging study such as magnetic resonance cholangiopancreatography (MRCP) or endoscopic ultrasonography can be done to screen for bile duct stones before proceeding to ERCP.

The ACG guidelines suggest urgent ERCP (preferably within 24 hours) for those with severe biliary pancreatitis complicated by organ failure or those with suspicion of cholangitis. The level of evidence is I, ie, “strong evidence from at least one published systematic review of multiple well-designed randomized controlled trials.”1

Elective ERCP is recommended for those who are poor surgical candidates. ERCP is also recommended for those with rising liver enzyme values or imaging findings suggesting a retained common bile duct stone (including intraoperative cholangiography). Endoscopic ultrasonography or MRCP is recommended for those with slow clinical resolution, who are pregnant, or in whom uncertainty exists regarding the biliary etiology of pancreatitis.

Compliance rates with these and similar guidelines are not adequate. In an audit of adherence to the British Society of Gastroenterology guidelines, early ERCP was performed in only 25% of patients with severe biliary acute pancreatitis.6

LAPAROSCOPIC CHOLECYSTECTOMY FOR MILD BILIARY PANCREATITIS

Problem: Laparoscopic cholecystectomy is not done at admission or within 2 weeks in many patients with mild biliary pancreatitis.

If the gallbladder is not removed, biliary pancreatitis may recur in up to 61% of patients within 6 weeks of hospital discharge.37 This is the basis for guideline recommendations for surgery (or a confirmation of a surgery date) prior to hospital discharge.

The International Association of Pancreatology recommends early cholecystectomy (preferably during the same hospitalization) for patients with mild gallstone-associated acute pancreatitis.38 In severe gallstone-associated acute pancreatitis, cholecystectomy should be delayed until there is sufficient resolution of the inflammatory response and clinical recovery. The AGA guidelines advocate cholecystectomy as soon as possible and in no case later than 4 weeks after discharge to prevent relapse. ERCP with biliary sphinc-terotomy may also protect against relapse in those who are not fit to undergo surgery.

Recommendations for definitive management of gallstones (laparoscopic cholecystectomy or ERCP, or both) are not always followed. For example, a British study showed 70% compliance with this recommendation.4 A similar compliance audit in Germany revealed that cholecystectomy was performed during the initial hospital stay in only 23% of cases.7 In a New Zealand study, a regular compliance audit with feedback to surgeons resulted in an increase in the early cholecystectomy rate from 54% to 80%.8

References
  1. Banks PA, Freeman ML; Practice Parameters Committee of the American College of Gastroenterology. Practice guidelines in acute pancreatitis. Am J Gastroenterol 2006; 101:23792400.
  2. Forsmark CE, Baillie J; AGA Institute Clinical Practice and Economics Committee. AGA Institute technical review on acute pancreatitis. Gastroenterology 2007; 132:20222044.
  3. United Kingdom guidelines for the management of acute pancreatitis. British Society of Gastroenterology. Gut 1998; 42(suppl 2):S1S13.
  4. Norton SA, Cheruvu CV, Collins J, Dix FP, Eyre-Brook IA. An assessment of clinical guidelines for the management of acute pancreatitis. Ann R Coll Surg Engl 2001; 83:399405.
  5. Chiang DT, Anozie A, Fleming WR, Kiroff GK. Comparative study on acute pancreatitis management. ANZ J Surg 2004; 74:218221.
  6. Barnard J, Siriwardena AK. Variations in implementation of current national guidelines for the treatment of acute pancreatitis: implications for acute surgical service provision. Ann R Coll Surg Engl 2002; 84:7981.
  7. Lankisch PG, Weber-Dany B, Lerch MM. Clinical perspectives in pancreatology: compliance with acute pancreatitis in Germany [letter]. Pancreatology 2005; 5:591593.
  8. Connor SJ, Lienert AR, Brown LA, Bagshaw PF. Closing the audit loop is necessary to achieve compliance with evidence-based guidelines in the management of acute pancreatitis. N Z Med J 2008; 121:1925.
  9. Mofidi R, Duff MD, Wigmore SJ, Madhavan KK, Garden OJ, Parks RW. Association between early systemic inflammatory response, severity of multiorgan dysfunction, and death in acute pancreatitis. Br J Surg 2006; 93:738744.
  10. Brown A, Orav J, Banks PA. Hemoconcentration is an early marker for organ failure and necrotizing pancreatitis. Pancreas 2000; 20:367372.
  11. Lankisch PG, Mahlke R, Blum T, et al. Hemoconcentration: an early marker of severe and/or necrotizing pancreatitis? A critical appraisal. Am J Gastroenterol 2001; 96:20812085.
  12. Singh VK, Wu BU, Bollen TL, et al. A prospective evaluation of the bedside index for severity in acute pancreatitis score in assessing mortality and intermediate markers of severity in acute pancreatitis. Am J Gastroenterol 2009; 104:966971.
  13. Ranson JH, Rifkind KM, Roses DF, Fink SD, Eng K, Spencer FC. Prognostic signs and the role of operative management in acute pancreatitis. Surg Gynecol Obstet 1974; 139:6981.
  14. Larvin M. Assessment of clinical severity and prognosis. In:Beger HG, Warshaw AL, Buchler MW, et al, editors. The Pancreas. Blackwell Science: New York, 1998:489502.
  15. Wu BU, Johannes RS, Sun X, Tabak Y, Conwell DL, Banks PA. The early prediction of mortality in acute pancreatitis: a large population-based study. Gut 2008; 57:16981703.
  16. Bradley EL. A clinically based classification system for acute pancreatitis. Summary of the International Symposium on Acute Pancreatitis, Atlanta, GA, September 11 through 13, 1992. Arch Surg 1993, 128:586590.
  17. Forgacs B, Eible G, Faulhaber J, Kahrau S, Buhr H, Foitzik T. Effect of fluid resuscitation with and without endothelin A receptor blockade on hemoconcentration and organ function in experimental pancreatitis. Eur Surg Res 2000; 32:162168.
  18. Brown A, Baillargeon JD, Hughes MD, Banks PA. Can fluid resuscitation prevent pancreatic necrosis in severe acute pancreatitis? Pancreatology 2002; 2:104107.
  19. Pandol SJ, Saluja AK, Imrie CW, Banks PA. Acute pancreatitis: bench to the bedside. Gastroenterology 2007; 132:11271151.
  20. Pezzilli R, Uomo G, Gabbrielli A, et al; ProInf-AISP Study Group. A prospective multicenter survey on the treatment of acute pancreatitis in Italy. Dig Liver Dis 2007; 39:838846.
  21. Gardner TB, Vege SS, Pearson RK, Chari ST. Fluid resuscitation in acute pancreatitis. Clin Gastroenterol Hepatol 2008; 6:10701076.
  22. Petrov MS, Pylypchuk RD, Emelyanov NV. Systematic review: nutritional support in acute pancreatitis. Aliment Pharmacol Ther 2008; 28:704712.
  23. Marik PE, Zaloga GP. Meta-analysis of parenteral nutrition versus enteral nutrition in patients with acute pancreatitis. BMJ 2004; 328:1407.
  24. Balthazar EJ, Robinson DL, Megibow AJ, Ranson JH. Acute pancreatitis: value of CT in establishing prognosis. Radiology 1990; 174:331336.
  25. Balthazar EJ. Acute pancreatitis: assessment of severity with clinical and CT evaluation. Radiology 2002; 223:603613.
  26. Foitzik T, Bassi DG, Schmidt J, et al. Intravenous contrast medium accentuates the severity of acute necrotizing pancreatitis in the rat. Gastroenterology 1994; 106:207214.
  27. Carmona-Sanchez R, Uscanga L, Bezaury-Rivas P, Robles-Díaz G, Suazo-Barahona J, Vargas-Vorácková F. Potential harmful effect of iodinated intravenous contrast medium on the clinical course of mild acute pancreatitis. Arch Surg 2000; 135:12801284.
  28. Uhl W, Roggo A, Kirschstein T, et al. Influence of contrast-enhanced computed tomography on couse and outcome in patients with acute pancreatitis. Pancreas 2002; 24:191197.
  29. Foitzik T, Klar E. Non-compliance with guidelines for the management of severe acute pancreatitis among German surgeons. Pancreatology 2007; 7:8085.
  30. Villatoro E, Bassi C, Larvin M. Antibiotic therapy for prophylaxis against infection of pancreatic necrosis in acute pancreatitis. Cochrane Database Syst Rev 2006;CD002941.
  31. Heinrich S, Schafer M, Rousson V, Clavien PA. Evidence-based treatment of acute pancreatitis: a look at established paradigms. Ann Surg 2006; 243:154168.
  32. Neoptolemos JP, Carr-Locke DL, London NJ, Bailey IA, James D, Fossard DP. Controlled trial of urgent endoscopic retrograde cholangiopancreatography and endoscopic sphincterotomy versus conservative treatment for acute pancreatitis due to gallstones. Lancet 1988; 2:979983.
  33. Fan ST, Lai EC, Mok FP, Lo CM, Zheng SS, Wong J. Early treatment of acute biliary pancreatitis by endoscopic papillotomy. N Engl J Med 1993; 328:228232.
  34. Folsch UR, Nitsche R, Ludtke R, Hilgers RA, Creutzfeldt W. Early ERCP and papillotomy compared with conservative treatment for acute biliary pancreatitis. The German Study Group on Acute Biliary Pancreatitis. N Engl J Med 1997; 336:237242.
  35. Ayub K, Imada R, Slavin J. Endoscopic retrograde cholangiopancreatography in gallstone associated pancreatitis. Cochrane Database Syst Rev 2004;CD003630
  36. Oria A, Cimmino D, Ocampo C, et al. Early endoscopic intervention versus early conservative management in patients with acute gallstone pancreatitis and biliopancreatic obstruction. A randomized clinical trial. Ann Surg 2007; 245:1017.
  37. Frei GJ, Frei VT, Thirlby RC, McClelland RN. Biliary pancreatitis: clinical presentation and surgical management. Am J Surg 1986; 151:170175.
  38. Uhl W, Warshaw A, Imrie C, et al; International Association of Pancreatology. IAP guidelines on the surgical management of acute pancreatitis. Pancreatology 2002; 2:565573.
References
  1. Banks PA, Freeman ML; Practice Parameters Committee of the American College of Gastroenterology. Practice guidelines in acute pancreatitis. Am J Gastroenterol 2006; 101:23792400.
  2. Forsmark CE, Baillie J; AGA Institute Clinical Practice and Economics Committee. AGA Institute technical review on acute pancreatitis. Gastroenterology 2007; 132:20222044.
  3. United Kingdom guidelines for the management of acute pancreatitis. British Society of Gastroenterology. Gut 1998; 42(suppl 2):S1S13.
  4. Norton SA, Cheruvu CV, Collins J, Dix FP, Eyre-Brook IA. An assessment of clinical guidelines for the management of acute pancreatitis. Ann R Coll Surg Engl 2001; 83:399405.
  5. Chiang DT, Anozie A, Fleming WR, Kiroff GK. Comparative study on acute pancreatitis management. ANZ J Surg 2004; 74:218221.
  6. Barnard J, Siriwardena AK. Variations in implementation of current national guidelines for the treatment of acute pancreatitis: implications for acute surgical service provision. Ann R Coll Surg Engl 2002; 84:7981.
  7. Lankisch PG, Weber-Dany B, Lerch MM. Clinical perspectives in pancreatology: compliance with acute pancreatitis in Germany [letter]. Pancreatology 2005; 5:591593.
  8. Connor SJ, Lienert AR, Brown LA, Bagshaw PF. Closing the audit loop is necessary to achieve compliance with evidence-based guidelines in the management of acute pancreatitis. N Z Med J 2008; 121:1925.
  9. Mofidi R, Duff MD, Wigmore SJ, Madhavan KK, Garden OJ, Parks RW. Association between early systemic inflammatory response, severity of multiorgan dysfunction, and death in acute pancreatitis. Br J Surg 2006; 93:738744.
  10. Brown A, Orav J, Banks PA. Hemoconcentration is an early marker for organ failure and necrotizing pancreatitis. Pancreas 2000; 20:367372.
  11. Lankisch PG, Mahlke R, Blum T, et al. Hemoconcentration: an early marker of severe and/or necrotizing pancreatitis? A critical appraisal. Am J Gastroenterol 2001; 96:20812085.
  12. Singh VK, Wu BU, Bollen TL, et al. A prospective evaluation of the bedside index for severity in acute pancreatitis score in assessing mortality and intermediate markers of severity in acute pancreatitis. Am J Gastroenterol 2009; 104:966971.
  13. Ranson JH, Rifkind KM, Roses DF, Fink SD, Eng K, Spencer FC. Prognostic signs and the role of operative management in acute pancreatitis. Surg Gynecol Obstet 1974; 139:6981.
  14. Larvin M. Assessment of clinical severity and prognosis. In:Beger HG, Warshaw AL, Buchler MW, et al, editors. The Pancreas. Blackwell Science: New York, 1998:489502.
  15. Wu BU, Johannes RS, Sun X, Tabak Y, Conwell DL, Banks PA. The early prediction of mortality in acute pancreatitis: a large population-based study. Gut 2008; 57:16981703.
  16. Bradley EL. A clinically based classification system for acute pancreatitis. Summary of the International Symposium on Acute Pancreatitis, Atlanta, GA, September 11 through 13, 1992. Arch Surg 1993, 128:586590.
  17. Forgacs B, Eible G, Faulhaber J, Kahrau S, Buhr H, Foitzik T. Effect of fluid resuscitation with and without endothelin A receptor blockade on hemoconcentration and organ function in experimental pancreatitis. Eur Surg Res 2000; 32:162168.
  18. Brown A, Baillargeon JD, Hughes MD, Banks PA. Can fluid resuscitation prevent pancreatic necrosis in severe acute pancreatitis? Pancreatology 2002; 2:104107.
  19. Pandol SJ, Saluja AK, Imrie CW, Banks PA. Acute pancreatitis: bench to the bedside. Gastroenterology 2007; 132:11271151.
  20. Pezzilli R, Uomo G, Gabbrielli A, et al; ProInf-AISP Study Group. A prospective multicenter survey on the treatment of acute pancreatitis in Italy. Dig Liver Dis 2007; 39:838846.
  21. Gardner TB, Vege SS, Pearson RK, Chari ST. Fluid resuscitation in acute pancreatitis. Clin Gastroenterol Hepatol 2008; 6:10701076.
  22. Petrov MS, Pylypchuk RD, Emelyanov NV. Systematic review: nutritional support in acute pancreatitis. Aliment Pharmacol Ther 2008; 28:704712.
  23. Marik PE, Zaloga GP. Meta-analysis of parenteral nutrition versus enteral nutrition in patients with acute pancreatitis. BMJ 2004; 328:1407.
  24. Balthazar EJ, Robinson DL, Megibow AJ, Ranson JH. Acute pancreatitis: value of CT in establishing prognosis. Radiology 1990; 174:331336.
  25. Balthazar EJ. Acute pancreatitis: assessment of severity with clinical and CT evaluation. Radiology 2002; 223:603613.
  26. Foitzik T, Bassi DG, Schmidt J, et al. Intravenous contrast medium accentuates the severity of acute necrotizing pancreatitis in the rat. Gastroenterology 1994; 106:207214.
  27. Carmona-Sanchez R, Uscanga L, Bezaury-Rivas P, Robles-Díaz G, Suazo-Barahona J, Vargas-Vorácková F. Potential harmful effect of iodinated intravenous contrast medium on the clinical course of mild acute pancreatitis. Arch Surg 2000; 135:12801284.
  28. Uhl W, Roggo A, Kirschstein T, et al. Influence of contrast-enhanced computed tomography on couse and outcome in patients with acute pancreatitis. Pancreas 2002; 24:191197.
  29. Foitzik T, Klar E. Non-compliance with guidelines for the management of severe acute pancreatitis among German surgeons. Pancreatology 2007; 7:8085.
  30. Villatoro E, Bassi C, Larvin M. Antibiotic therapy for prophylaxis against infection of pancreatic necrosis in acute pancreatitis. Cochrane Database Syst Rev 2006;CD002941.
  31. Heinrich S, Schafer M, Rousson V, Clavien PA. Evidence-based treatment of acute pancreatitis: a look at established paradigms. Ann Surg 2006; 243:154168.
  32. Neoptolemos JP, Carr-Locke DL, London NJ, Bailey IA, James D, Fossard DP. Controlled trial of urgent endoscopic retrograde cholangiopancreatography and endoscopic sphincterotomy versus conservative treatment for acute pancreatitis due to gallstones. Lancet 1988; 2:979983.
  33. Fan ST, Lai EC, Mok FP, Lo CM, Zheng SS, Wong J. Early treatment of acute biliary pancreatitis by endoscopic papillotomy. N Engl J Med 1993; 328:228232.
  34. Folsch UR, Nitsche R, Ludtke R, Hilgers RA, Creutzfeldt W. Early ERCP and papillotomy compared with conservative treatment for acute biliary pancreatitis. The German Study Group on Acute Biliary Pancreatitis. N Engl J Med 1997; 336:237242.
  35. Ayub K, Imada R, Slavin J. Endoscopic retrograde cholangiopancreatography in gallstone associated pancreatitis. Cochrane Database Syst Rev 2004;CD003630
  36. Oria A, Cimmino D, Ocampo C, et al. Early endoscopic intervention versus early conservative management in patients with acute gallstone pancreatitis and biliopancreatic obstruction. A randomized clinical trial. Ann Surg 2007; 245:1017.
  37. Frei GJ, Frei VT, Thirlby RC, McClelland RN. Biliary pancreatitis: clinical presentation and surgical management. Am J Surg 1986; 151:170175.
  38. Uhl W, Warshaw A, Imrie C, et al; International Association of Pancreatology. IAP guidelines on the surgical management of acute pancreatitis. Pancreatology 2002; 2:565573.
Issue
Cleveland Clinic Journal of Medicine - 76(12)
Issue
Cleveland Clinic Journal of Medicine - 76(12)
Page Number
697-704
Page Number
697-704
Publications
Publications
Topics
Article Type
Display Headline
Acute pancreatitis: Problems in adherence to guidelines
Display Headline
Acute pancreatitis: Problems in adherence to guidelines
Sections
Inside the Article

KEY POINTS

  • Serum amylase and lipase levels are often needlessly measured every day.
  • Often, severity assessments are not performed regularly or acted on.
  • Often, not enough fluid is replaced, or fluid status is not adequately monitored.
  • In many severe cases, enteral or parenteral feeding is not started soon enough.
  • Computed tomography is not done in many patients with severe acute pancreatitis, or it is performed too soon.
  • In many cases of suspected infected necrosis, fine-needle aspiration is not done.
  • Broad-spectrum antibiotics are often used inappropriately in patients with mild acute pancreatitis and in patients with sterile necrotizing pancreatitis who are clinically stable and have no signs of sepsis.
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media

Is an ACE inhibitor plus an ARB more effective than either drug alone?

Article Type
Changed
Thu, 04/12/2018 - 11:08
Display Headline
Is an ACE inhibitor plus an ARB more effective than either drug alone?

No. Although randomized, controlled trials have shown convincingly that angiotensin-converting enzyme (ACE) inhibitors reduce the rates of death, myocardial infarction, stroke, and heart failure in patients with known coronary artery disease or left ventricular dysfunction,1 and that angiotensin receptor blockers (ARBs) are “noninferior” to and better tolerated than ACE inhibitors, causing less angioedema and cough but costing more,2 dual renin-angiotensin system (RAS) blockade—an ACE inhibitor plus an ARB—has never been shown to reduce the rates of morbidity or death from any cause.

In fact, the Ongoing Telmisartan Alone and in Combination With Ramipril Global Endpoint Trial (ONTARGET)3,4 found that dual RAS blockade was no more beneficial than monotherapy with an ACE inhibitor or an ARB in preventing serious outcomes in patients with known vascular disease or diabetes with end-organ damage. Furthermore, patients on dual RAS blockade had higher rates of renal insufficiency, hyperkalemia, and hypotension.

THE RATIONALE FOR DUAL RAS BLOCKADE

Dual RAS blockade was first proposed in the early 1990s as a way to avoid the “escape phenomenon” (incomplete suppression of angiotensin II) with ACE inhibitor monotherapy.5 Indeed, studies in rats showed a synergistic effect on blood pressure with an ACE inhibitor combined with an ARB,6 and these results were encouraging enough for the medical community to make a remarkably quick transition to adopting dual RAS blockade in clinical practice.

The concept of dual RAS blockade was so appealing that effects on surrogate end points—lower blood pressure, less protein in the urine, and improved endothelial function—were accepted as free passes, obviating the need for evidence of an effect on hard end points such as lower rates of cardiovascular death or kidney failure. Currently, in the United States, about 1.5% of all patients on RAS blockers are currently receiving both an ACE inhibitor and an ARB.

CONDITIONS IN WHICH DUAL RAS BLOCKADE WAS THOUGHT BENEFICIAL

Hypertension

The European Society of Cardiology’s 2007 clinical practice guidelines7 say that treatment with an ACE inhibitor plus an ARB is preferred for hypertensive patients with metabolic syndrome and its major components (eg, abdominal obesity, insulin resistance, frank diabetes).

Dulton et al, in a meta-analysis,8 calculated that the combination of an ACE inhibitor and an ARB lowered 24-hour blood pressure by 4/3 mm Hg more than monotherapy did. However, most of the studies were of short duration (6 to 8 weeks) and used submaximal doses or once-daily doses of a short-acting ACE inhibitor. Interestingly, studies that used a long-acting ACE inhibitor or a larger dose of a short-acting ACE inhibitor generally showed no additive effect on blood pressure when an ARB was added.

Hence, more evidence from larger randomized and appropriately designed studies is needed before we can conclude that dual RAS blockade is safe and significantly superior to monotherapy in blood pressure control.

Proteinuria

Proteinuria is a surrogate end point for cardiovascular death and is a marker as well as a cause of progressive renal insufficiency. It therefore seemed rational that modifying the degree of proteinuria would translate into robust clinical benefits. Several studies9 showed better renal outcomes, such as fewer patients needing dialysis with combination therapy than with an ACE inhibitor or ARB alone. However, this has never been proven in an adequately powered trial.

ONTARGET was a perfect opportunity to convert what seemed like reliable mechanistic information into solid outcome data.3 The trial enrolled 25,620 patients with established atherosclerotic disease or with diabetes and evidence of end-organ damage. At baseline, 13.1% had microalbuminuria and 4.0% had macroalbuminuria.3 The amount of protein in the urine increased by a significantly lesser amount in the ARB group and in the dualtherapy group than in the group taking only an ACE inhibitor, but in the dual-therapy group this apparent advantage came at the expense of hard end points: more patients reached the primary composite end point of needing dialysis, doubling of their serum creatinine level, or death.

Reducing proteinuria could be an important benefit, but it certainly does not outweigh the risk of increased rates of renal failure and death.

Atherosclerosis and acute coronary syndrome

The road to myocardial infarction begins with inflammation in the “shoulders” of atherosclerotic plaques, which subsequently rupture. Tissue ACE activity and expression of the angiotensin II type 1 receptor are significantly increased in patients with acute coronary syndrome and primarily co-localized to the shoulder regions of the plaque.10 Giving an ACE inhibitor or an ARB to patients who have unstable angina or who have had a myocardial infarction may decrease the rate of reinfarction and lessens the inflammatory process in the atherosclerotic plaque.

Large randomized clinical trials such as HOPE (Heart Outcomes Prevention Evaluation)11 and EUROPA (European Trial on Reduction of Cardiac Events With Perindopril in Stable Coronary Artery Disease)12 showed a lower rate of cardiovascular death in patients with established coronary artery disease and normal left ventricular function if they received an ACE inhibitor. In the HOPE trial, the rate of cardiovascular death was 25% lower in patients treated with ramipril (Altace) vs placebo.11 (The year after HOPE was published, the number of prescriptions for ramipril went up 400%). Interestingly, studies of ARBs for secondary prevention failed to show any lowering of the rate of cardiovascular death or myocardial infarction.13

In ONTARGET,4 although the combination of telmisartan (Micardis) and ramipril had a greater effect on blood pressure, it was not significantly better than ramipril alone in terms of the primary outcome of death from cardiovascular causes, myocardial infarction, stroke, or hospitalization for heart failure (relative risk 0.99).

 

 

Heart failure

The bulk of data on dual RAS blockade in heart failure patients comes from three large randomized trials: CHARM-Added (Candesartan in Heart Failure: Assessment of Reduction in Mortality and Morbidity),14 VALIANT (Valsartan in Acute Myocardial Infarction Trial),15 and VAL-HeFT (Valsartan Heart Failure Trial).16

CHARM-Added14 was the only trial that showed a reduction in cardiovascular deaths with dual RAS therapy (absolute risk reduction 3.6%). It also showed a lower rate of hospitalization for heart failure (absolute risk reduction 4%). However, the rate of allcause mortality was not different between the groups. Of note, more patients receiving dual RAS blockade had to stop taking the study drug because of adverse effects.

Val-HeFT16 showed, in a post hoc analysis, higher rates of morbidity (cardiac arrest, hospitalization for heart failure, or receipt of intravenous inotropic or vasodilator therapy for at least 4 hours) and death when the ARB valsartan (Diovan) was added to the combination of an ACE inhibitor plus a beta-blocker.

A recent meta-analysis17 of safety and tolerability of dual RAS blockade compared with an ACE inhibitor alone found a higher risk of discontinuation because of adverse effects such as hyperkalemia, renal dysfunction, and hypotension in patients on dual RAS blockade. The authors concluded that, given the adverse effects and the lack of consistent survival benefit, the available data do not support the routine addition of an ARB to ACE inhibitor therapy in heart failure patients.

WHAT ABOUT DIRECT RENIN INHIBITORS?

Another class of RAS blockers is available: direct renin inhibitors. Therefore, dual RAS blockade can be achieved by combining an ACE inhibitor with an ARB, an ACE inhibitor with a direct renin inhibitor, or an ARB with a direct renin inhibitor.

We have some outcome data on the combination of an ACE inhibitor plus an ARB,3,4,17 but none for the other two possible dual RAS combinations. Thus far, we know that dual RAS blockade with an ARB and an ACE inhibitor is not beneficial in patients like those in ONTARGET, and that it has questionable benefit in heart failure. However, little is known about combining a direct renin inhibitor with either an ACE inhibitor or an ARB.

Since ARBs and ACE inhibitors both increase plasma renin activity and only partially block the RAS, the argument has been put forward that the addition of a drug such as a direct renin inhibitor, which really decreases plasma renin activity, has the potential to be more beneficial than blockade with either an ACE inhibitor or an ARB. In theory, this is an attractive concept and certainly deserves scrutiny in outcome studies such as ALTITUDE (Aliskiren Trial in Type 2 Diabetes Using Cardio-Renal Endpoints).18

SURROGATE END POINTS: A CAVEAT

As defined by Temple,19 a surrogate end point of a clinical trial is a laboratory measurement or a physical sign used as a substitute for a clinically meaningful end point that measures directly how patients feel or function, or if they survive. Effects on surrogate end points often fail to predict the true clinical effects of an intervention, as the ONTARGET data demonstrated. Among several explanations for this failure is that interventions may affect the clinical outcome by unintended, unanticipated, and unrecognized mechanisms that operate independently of the disease process.20 Nonetheless, surrogate end point cosmetics remains attractive for many clinicians.

The ONTARGET findings indicate that there is no clinically important benefit in adding an ARB for patients with hypertension, proteinuria, heart failure, or coronary artery disease if they are already being treated with an ACE inhibitor. This would indicate that dual RAS blockade should be avoided in clinical practice until we are provided with better evidence.

References
  1. Father MD, Yusuf S, Kober L, et al. Long-term ACE-inhibitor therapy in patients with heart failure or left ventricular dysfunction: a systemic overview of data from individual patients. ACEInhibitor Myocardial Infarction Collaborative Group. Lancet 2000; 355:15751581.
  2. Pitt B, Poole-Wilson PA, Segal R, et al. Effect of losartan compared with captopril on mortality in patients with symptomatic heart failure: randomized trial—the Losartan Heart Failure Survival Study ELITE II. Lancet 2000; 355:15821587.
  3. Mann JF, Schmiede RE, McQueen M, et al; ONTARGET investigators. Renal outcomes with telmisartan, ramipril, or both, in people at high vascular risk (the ONTARGET study): a multicentre, randomized, double-blind, controlled trial. Lancet 2008; 372:547553.
  4. Yusuf S, Teo KK, Pogue J, et al; ONTARGET Investigators Telmisartan, ramipril, or both in patients at high risk for vascular events, N Engl J Med 2008; 358:15471559.
  5. van den Meiracker AH, Man in ‘t Veld AJ, Admiraal PJ, et al. Partial escape of angiotensin converting enzyme (ACE) inhibition during prolonged ACE inhibitor treatment: dose it exist and does it affect the antihypertensive response? J Hypertens 1992; 10:803812.
  6. Menard J, Campbell DJ, Azizi M, Gonzales MF. Synergistic effects of ACE inhibition and Ang II antagonism on blood pressure, cardiac weight, and renin in spontaneously hypertensive rats. Circulation 1997; 96:30723078.
  7. Mancia G, De Backer G, Dominiczak A, et al; Task Force for the Management of Arterial Hypertension of the European Society of Hypertension. 2007 guidelines for the management of arterial hypertension. J Hypertens 2007; 25:11051187.
  8. Dulton TW, He FJ, MacGregor GA. Systematic review of combined angiotensin-converting enzyme inhibition and angiotensin receptor blockade in hypertension. Hypertension 2005; 45:880886.
  9. Kunz R, Fredrich C, Wolbers M, Mann JF. Meta-analysis: effect of monotherapy and combination therapy with inhibitors of the renin angiotensin system on proteinuria in renal disease. Ann Intern Med 2008; 148:3048.
  10. Schieffer B, Schieffer E, Hilfiker-Kleiner D, et al. Expression of angiotensin II and interleukin 6 in human coronary atherosclerotic plaques: potential implications for inflammation and plaque instability. Circulation 2000; 101:13721378.
  11. Yusuf S, Sleight P, Pogue J, Bosch J, Davies R, Dagenais G. Effects of an angiotensin-converting-enzyme inhibitor, ramipril, on cardiovascular events in high-risk patients. The Heart Outcomes Prevention Evaluation Study Investigators. N Engl J Med 2000; 342:145153.
  12. Fox KM; European trial On reduction of cardiac events with Perindopril in stable coronary Artery disease Investigators. Efficacy of perindopril in reduction of cardiovascular events among patients with stable coronary artery disease: randomized, double-blind, placebo-controlled, multicentre trial (the EUROPA study). Lancet 2003; 362:782788.
  13. Dahlöf B, Devereux RB, Kjeldsen SE, et al; LIFE Study Group. Cardiovascular morbidity and mortality in the Losartan Intervention For Endpoint reduction in hypertension study (LIFE): a randomized trial against atenolol. Lancet 2002; 359:9951003.
  14. McMurray JJ, Ostergren J, Swedberg K, et al; CHARM Investigators and Committees. Effects of candesartan in patients with chronic heart failure and reduced leftventricular systolic function taking angiotensin converting enzyme inhibitors: the CHARM-Added trial. Lancet 2003; 362:767771.
  15. Pfeffer MA, McMurray JJ, Velzquez E, et al; Valsartan in Acute Myocardial Infarction Trial Investigators. Valsartan, captopril, or both in myocardial infarction complicated by heart failure, left ventricular dysfunction, or both. N Engl J Med 2003; 349:18931906.
  16. Cohn JN, Tognoni GValsartan Heart Failure Trial Investigators. A randomized trial of the angiotensin receptor blocker valsartan in chronic heart failure. N Engl J Med 2001: 345:16671675.
  17. Lakhdar R, Al-Mallah MH, Lanfear DE. Safety and tolerability of angiotensin-converting enzyme inhibitor versus the combination of angiotensin-converting enzyme inhibitor and angiotensin receptor blocker in patients with left ventricular dysfunction: a systematic review and meta-analysis of randomized controlled trials. J Card Fail 2008; 14:181188.
  18. Parving H-H, Brenner BM, McMurray JJV, et al. Aliskiren Trial in Type 2 Diabetes Using Cardio-Renal Endpoints (ALTITUDE): rationale and study design [published online ahead of print January 14, 2009]. Nephrol Dial Transplant. doi:10.1093/ndt/gfn721.
  19. Temple RJ. A regulatory authority’s opinion about surrogate endpoints. In:Nimmo WS, Tucker GT, eds. Clinical Measurement in Drug Evaluation. J Wiley: New York, 1995.
  20. Messerli FH. The sudden demise of dual renin-angiotensin system blockade or the soft science of the surrogate end point. J Am Coll Cardiol 2009; 53:468470.
Article PDF
Author and Disclosure Information

Franz H. Messerli, MD
Division of Cardiology, St. Luke’s Roosevelt Hospital Center, New York, NY

Melana Yuzefpolskaya, MD
Division of Cardiology, St. Luke’s Roosevelt Hospital Center, New York, NY

Address: Franz H. Messerli, MD, Division of Cardiology, St. Luke’s Roosevelt Hospital Center, 1000 10th Avenue, Suite 3B-30, New York, NY 10019; e-mail [email protected]

Dr. Messerli has been an ad hoc consultant and speaker for GlaxoSmithKline, Novartis, Boehringer Ingelheim, Forest, Daiichi-Sankyo, Sanofi, and Savient Pharmaceuticals, and has received grant support from GlaxoSmithKline, Novartis, Forest, Daiichi-Sankyo, and Boehringer Ingelheim.

Issue
Cleveland Clinic Journal of Medicine - 76(12)
Publications
Topics
Page Number
693-696
Sections
Author and Disclosure Information

Franz H. Messerli, MD
Division of Cardiology, St. Luke’s Roosevelt Hospital Center, New York, NY

Melana Yuzefpolskaya, MD
Division of Cardiology, St. Luke’s Roosevelt Hospital Center, New York, NY

Address: Franz H. Messerli, MD, Division of Cardiology, St. Luke’s Roosevelt Hospital Center, 1000 10th Avenue, Suite 3B-30, New York, NY 10019; e-mail [email protected]

Dr. Messerli has been an ad hoc consultant and speaker for GlaxoSmithKline, Novartis, Boehringer Ingelheim, Forest, Daiichi-Sankyo, Sanofi, and Savient Pharmaceuticals, and has received grant support from GlaxoSmithKline, Novartis, Forest, Daiichi-Sankyo, and Boehringer Ingelheim.

Author and Disclosure Information

Franz H. Messerli, MD
Division of Cardiology, St. Luke’s Roosevelt Hospital Center, New York, NY

Melana Yuzefpolskaya, MD
Division of Cardiology, St. Luke’s Roosevelt Hospital Center, New York, NY

Address: Franz H. Messerli, MD, Division of Cardiology, St. Luke’s Roosevelt Hospital Center, 1000 10th Avenue, Suite 3B-30, New York, NY 10019; e-mail [email protected]

Dr. Messerli has been an ad hoc consultant and speaker for GlaxoSmithKline, Novartis, Boehringer Ingelheim, Forest, Daiichi-Sankyo, Sanofi, and Savient Pharmaceuticals, and has received grant support from GlaxoSmithKline, Novartis, Forest, Daiichi-Sankyo, and Boehringer Ingelheim.

Article PDF
Article PDF

No. Although randomized, controlled trials have shown convincingly that angiotensin-converting enzyme (ACE) inhibitors reduce the rates of death, myocardial infarction, stroke, and heart failure in patients with known coronary artery disease or left ventricular dysfunction,1 and that angiotensin receptor blockers (ARBs) are “noninferior” to and better tolerated than ACE inhibitors, causing less angioedema and cough but costing more,2 dual renin-angiotensin system (RAS) blockade—an ACE inhibitor plus an ARB—has never been shown to reduce the rates of morbidity or death from any cause.

In fact, the Ongoing Telmisartan Alone and in Combination With Ramipril Global Endpoint Trial (ONTARGET)3,4 found that dual RAS blockade was no more beneficial than monotherapy with an ACE inhibitor or an ARB in preventing serious outcomes in patients with known vascular disease or diabetes with end-organ damage. Furthermore, patients on dual RAS blockade had higher rates of renal insufficiency, hyperkalemia, and hypotension.

THE RATIONALE FOR DUAL RAS BLOCKADE

Dual RAS blockade was first proposed in the early 1990s as a way to avoid the “escape phenomenon” (incomplete suppression of angiotensin II) with ACE inhibitor monotherapy.5 Indeed, studies in rats showed a synergistic effect on blood pressure with an ACE inhibitor combined with an ARB,6 and these results were encouraging enough for the medical community to make a remarkably quick transition to adopting dual RAS blockade in clinical practice.

The concept of dual RAS blockade was so appealing that effects on surrogate end points—lower blood pressure, less protein in the urine, and improved endothelial function—were accepted as free passes, obviating the need for evidence of an effect on hard end points such as lower rates of cardiovascular death or kidney failure. Currently, in the United States, about 1.5% of all patients on RAS blockers are currently receiving both an ACE inhibitor and an ARB.

CONDITIONS IN WHICH DUAL RAS BLOCKADE WAS THOUGHT BENEFICIAL

Hypertension

The European Society of Cardiology’s 2007 clinical practice guidelines7 say that treatment with an ACE inhibitor plus an ARB is preferred for hypertensive patients with metabolic syndrome and its major components (eg, abdominal obesity, insulin resistance, frank diabetes).

Dulton et al, in a meta-analysis,8 calculated that the combination of an ACE inhibitor and an ARB lowered 24-hour blood pressure by 4/3 mm Hg more than monotherapy did. However, most of the studies were of short duration (6 to 8 weeks) and used submaximal doses or once-daily doses of a short-acting ACE inhibitor. Interestingly, studies that used a long-acting ACE inhibitor or a larger dose of a short-acting ACE inhibitor generally showed no additive effect on blood pressure when an ARB was added.

Hence, more evidence from larger randomized and appropriately designed studies is needed before we can conclude that dual RAS blockade is safe and significantly superior to monotherapy in blood pressure control.

Proteinuria

Proteinuria is a surrogate end point for cardiovascular death and is a marker as well as a cause of progressive renal insufficiency. It therefore seemed rational that modifying the degree of proteinuria would translate into robust clinical benefits. Several studies9 showed better renal outcomes, such as fewer patients needing dialysis with combination therapy than with an ACE inhibitor or ARB alone. However, this has never been proven in an adequately powered trial.

ONTARGET was a perfect opportunity to convert what seemed like reliable mechanistic information into solid outcome data.3 The trial enrolled 25,620 patients with established atherosclerotic disease or with diabetes and evidence of end-organ damage. At baseline, 13.1% had microalbuminuria and 4.0% had macroalbuminuria.3 The amount of protein in the urine increased by a significantly lesser amount in the ARB group and in the dualtherapy group than in the group taking only an ACE inhibitor, but in the dual-therapy group this apparent advantage came at the expense of hard end points: more patients reached the primary composite end point of needing dialysis, doubling of their serum creatinine level, or death.

Reducing proteinuria could be an important benefit, but it certainly does not outweigh the risk of increased rates of renal failure and death.

Atherosclerosis and acute coronary syndrome

The road to myocardial infarction begins with inflammation in the “shoulders” of atherosclerotic plaques, which subsequently rupture. Tissue ACE activity and expression of the angiotensin II type 1 receptor are significantly increased in patients with acute coronary syndrome and primarily co-localized to the shoulder regions of the plaque.10 Giving an ACE inhibitor or an ARB to patients who have unstable angina or who have had a myocardial infarction may decrease the rate of reinfarction and lessens the inflammatory process in the atherosclerotic plaque.

Large randomized clinical trials such as HOPE (Heart Outcomes Prevention Evaluation)11 and EUROPA (European Trial on Reduction of Cardiac Events With Perindopril in Stable Coronary Artery Disease)12 showed a lower rate of cardiovascular death in patients with established coronary artery disease and normal left ventricular function if they received an ACE inhibitor. In the HOPE trial, the rate of cardiovascular death was 25% lower in patients treated with ramipril (Altace) vs placebo.11 (The year after HOPE was published, the number of prescriptions for ramipril went up 400%). Interestingly, studies of ARBs for secondary prevention failed to show any lowering of the rate of cardiovascular death or myocardial infarction.13

In ONTARGET,4 although the combination of telmisartan (Micardis) and ramipril had a greater effect on blood pressure, it was not significantly better than ramipril alone in terms of the primary outcome of death from cardiovascular causes, myocardial infarction, stroke, or hospitalization for heart failure (relative risk 0.99).

 

 

Heart failure

The bulk of data on dual RAS blockade in heart failure patients comes from three large randomized trials: CHARM-Added (Candesartan in Heart Failure: Assessment of Reduction in Mortality and Morbidity),14 VALIANT (Valsartan in Acute Myocardial Infarction Trial),15 and VAL-HeFT (Valsartan Heart Failure Trial).16

CHARM-Added14 was the only trial that showed a reduction in cardiovascular deaths with dual RAS therapy (absolute risk reduction 3.6%). It also showed a lower rate of hospitalization for heart failure (absolute risk reduction 4%). However, the rate of allcause mortality was not different between the groups. Of note, more patients receiving dual RAS blockade had to stop taking the study drug because of adverse effects.

Val-HeFT16 showed, in a post hoc analysis, higher rates of morbidity (cardiac arrest, hospitalization for heart failure, or receipt of intravenous inotropic or vasodilator therapy for at least 4 hours) and death when the ARB valsartan (Diovan) was added to the combination of an ACE inhibitor plus a beta-blocker.

A recent meta-analysis17 of safety and tolerability of dual RAS blockade compared with an ACE inhibitor alone found a higher risk of discontinuation because of adverse effects such as hyperkalemia, renal dysfunction, and hypotension in patients on dual RAS blockade. The authors concluded that, given the adverse effects and the lack of consistent survival benefit, the available data do not support the routine addition of an ARB to ACE inhibitor therapy in heart failure patients.

WHAT ABOUT DIRECT RENIN INHIBITORS?

Another class of RAS blockers is available: direct renin inhibitors. Therefore, dual RAS blockade can be achieved by combining an ACE inhibitor with an ARB, an ACE inhibitor with a direct renin inhibitor, or an ARB with a direct renin inhibitor.

We have some outcome data on the combination of an ACE inhibitor plus an ARB,3,4,17 but none for the other two possible dual RAS combinations. Thus far, we know that dual RAS blockade with an ARB and an ACE inhibitor is not beneficial in patients like those in ONTARGET, and that it has questionable benefit in heart failure. However, little is known about combining a direct renin inhibitor with either an ACE inhibitor or an ARB.

Since ARBs and ACE inhibitors both increase plasma renin activity and only partially block the RAS, the argument has been put forward that the addition of a drug such as a direct renin inhibitor, which really decreases plasma renin activity, has the potential to be more beneficial than blockade with either an ACE inhibitor or an ARB. In theory, this is an attractive concept and certainly deserves scrutiny in outcome studies such as ALTITUDE (Aliskiren Trial in Type 2 Diabetes Using Cardio-Renal Endpoints).18

SURROGATE END POINTS: A CAVEAT

As defined by Temple,19 a surrogate end point of a clinical trial is a laboratory measurement or a physical sign used as a substitute for a clinically meaningful end point that measures directly how patients feel or function, or if they survive. Effects on surrogate end points often fail to predict the true clinical effects of an intervention, as the ONTARGET data demonstrated. Among several explanations for this failure is that interventions may affect the clinical outcome by unintended, unanticipated, and unrecognized mechanisms that operate independently of the disease process.20 Nonetheless, surrogate end point cosmetics remains attractive for many clinicians.

The ONTARGET findings indicate that there is no clinically important benefit in adding an ARB for patients with hypertension, proteinuria, heart failure, or coronary artery disease if they are already being treated with an ACE inhibitor. This would indicate that dual RAS blockade should be avoided in clinical practice until we are provided with better evidence.

No. Although randomized, controlled trials have shown convincingly that angiotensin-converting enzyme (ACE) inhibitors reduce the rates of death, myocardial infarction, stroke, and heart failure in patients with known coronary artery disease or left ventricular dysfunction,1 and that angiotensin receptor blockers (ARBs) are “noninferior” to and better tolerated than ACE inhibitors, causing less angioedema and cough but costing more,2 dual renin-angiotensin system (RAS) blockade—an ACE inhibitor plus an ARB—has never been shown to reduce the rates of morbidity or death from any cause.

In fact, the Ongoing Telmisartan Alone and in Combination With Ramipril Global Endpoint Trial (ONTARGET)3,4 found that dual RAS blockade was no more beneficial than monotherapy with an ACE inhibitor or an ARB in preventing serious outcomes in patients with known vascular disease or diabetes with end-organ damage. Furthermore, patients on dual RAS blockade had higher rates of renal insufficiency, hyperkalemia, and hypotension.

THE RATIONALE FOR DUAL RAS BLOCKADE

Dual RAS blockade was first proposed in the early 1990s as a way to avoid the “escape phenomenon” (incomplete suppression of angiotensin II) with ACE inhibitor monotherapy.5 Indeed, studies in rats showed a synergistic effect on blood pressure with an ACE inhibitor combined with an ARB,6 and these results were encouraging enough for the medical community to make a remarkably quick transition to adopting dual RAS blockade in clinical practice.

The concept of dual RAS blockade was so appealing that effects on surrogate end points—lower blood pressure, less protein in the urine, and improved endothelial function—were accepted as free passes, obviating the need for evidence of an effect on hard end points such as lower rates of cardiovascular death or kidney failure. Currently, in the United States, about 1.5% of all patients on RAS blockers are currently receiving both an ACE inhibitor and an ARB.

CONDITIONS IN WHICH DUAL RAS BLOCKADE WAS THOUGHT BENEFICIAL

Hypertension

The European Society of Cardiology’s 2007 clinical practice guidelines7 say that treatment with an ACE inhibitor plus an ARB is preferred for hypertensive patients with metabolic syndrome and its major components (eg, abdominal obesity, insulin resistance, frank diabetes).

Dulton et al, in a meta-analysis,8 calculated that the combination of an ACE inhibitor and an ARB lowered 24-hour blood pressure by 4/3 mm Hg more than monotherapy did. However, most of the studies were of short duration (6 to 8 weeks) and used submaximal doses or once-daily doses of a short-acting ACE inhibitor. Interestingly, studies that used a long-acting ACE inhibitor or a larger dose of a short-acting ACE inhibitor generally showed no additive effect on blood pressure when an ARB was added.

Hence, more evidence from larger randomized and appropriately designed studies is needed before we can conclude that dual RAS blockade is safe and significantly superior to monotherapy in blood pressure control.

Proteinuria

Proteinuria is a surrogate end point for cardiovascular death and is a marker as well as a cause of progressive renal insufficiency. It therefore seemed rational that modifying the degree of proteinuria would translate into robust clinical benefits. Several studies9 showed better renal outcomes, such as fewer patients needing dialysis with combination therapy than with an ACE inhibitor or ARB alone. However, this has never been proven in an adequately powered trial.

ONTARGET was a perfect opportunity to convert what seemed like reliable mechanistic information into solid outcome data.3 The trial enrolled 25,620 patients with established atherosclerotic disease or with diabetes and evidence of end-organ damage. At baseline, 13.1% had microalbuminuria and 4.0% had macroalbuminuria.3 The amount of protein in the urine increased by a significantly lesser amount in the ARB group and in the dualtherapy group than in the group taking only an ACE inhibitor, but in the dual-therapy group this apparent advantage came at the expense of hard end points: more patients reached the primary composite end point of needing dialysis, doubling of their serum creatinine level, or death.

Reducing proteinuria could be an important benefit, but it certainly does not outweigh the risk of increased rates of renal failure and death.

Atherosclerosis and acute coronary syndrome

The road to myocardial infarction begins with inflammation in the “shoulders” of atherosclerotic plaques, which subsequently rupture. Tissue ACE activity and expression of the angiotensin II type 1 receptor are significantly increased in patients with acute coronary syndrome and primarily co-localized to the shoulder regions of the plaque.10 Giving an ACE inhibitor or an ARB to patients who have unstable angina or who have had a myocardial infarction may decrease the rate of reinfarction and lessens the inflammatory process in the atherosclerotic plaque.

Large randomized clinical trials such as HOPE (Heart Outcomes Prevention Evaluation)11 and EUROPA (European Trial on Reduction of Cardiac Events With Perindopril in Stable Coronary Artery Disease)12 showed a lower rate of cardiovascular death in patients with established coronary artery disease and normal left ventricular function if they received an ACE inhibitor. In the HOPE trial, the rate of cardiovascular death was 25% lower in patients treated with ramipril (Altace) vs placebo.11 (The year after HOPE was published, the number of prescriptions for ramipril went up 400%). Interestingly, studies of ARBs for secondary prevention failed to show any lowering of the rate of cardiovascular death or myocardial infarction.13

In ONTARGET,4 although the combination of telmisartan (Micardis) and ramipril had a greater effect on blood pressure, it was not significantly better than ramipril alone in terms of the primary outcome of death from cardiovascular causes, myocardial infarction, stroke, or hospitalization for heart failure (relative risk 0.99).

 

 

Heart failure

The bulk of data on dual RAS blockade in heart failure patients comes from three large randomized trials: CHARM-Added (Candesartan in Heart Failure: Assessment of Reduction in Mortality and Morbidity),14 VALIANT (Valsartan in Acute Myocardial Infarction Trial),15 and VAL-HeFT (Valsartan Heart Failure Trial).16

CHARM-Added14 was the only trial that showed a reduction in cardiovascular deaths with dual RAS therapy (absolute risk reduction 3.6%). It also showed a lower rate of hospitalization for heart failure (absolute risk reduction 4%). However, the rate of allcause mortality was not different between the groups. Of note, more patients receiving dual RAS blockade had to stop taking the study drug because of adverse effects.

Val-HeFT16 showed, in a post hoc analysis, higher rates of morbidity (cardiac arrest, hospitalization for heart failure, or receipt of intravenous inotropic or vasodilator therapy for at least 4 hours) and death when the ARB valsartan (Diovan) was added to the combination of an ACE inhibitor plus a beta-blocker.

A recent meta-analysis17 of safety and tolerability of dual RAS blockade compared with an ACE inhibitor alone found a higher risk of discontinuation because of adverse effects such as hyperkalemia, renal dysfunction, and hypotension in patients on dual RAS blockade. The authors concluded that, given the adverse effects and the lack of consistent survival benefit, the available data do not support the routine addition of an ARB to ACE inhibitor therapy in heart failure patients.

WHAT ABOUT DIRECT RENIN INHIBITORS?

Another class of RAS blockers is available: direct renin inhibitors. Therefore, dual RAS blockade can be achieved by combining an ACE inhibitor with an ARB, an ACE inhibitor with a direct renin inhibitor, or an ARB with a direct renin inhibitor.

We have some outcome data on the combination of an ACE inhibitor plus an ARB,3,4,17 but none for the other two possible dual RAS combinations. Thus far, we know that dual RAS blockade with an ARB and an ACE inhibitor is not beneficial in patients like those in ONTARGET, and that it has questionable benefit in heart failure. However, little is known about combining a direct renin inhibitor with either an ACE inhibitor or an ARB.

Since ARBs and ACE inhibitors both increase plasma renin activity and only partially block the RAS, the argument has been put forward that the addition of a drug such as a direct renin inhibitor, which really decreases plasma renin activity, has the potential to be more beneficial than blockade with either an ACE inhibitor or an ARB. In theory, this is an attractive concept and certainly deserves scrutiny in outcome studies such as ALTITUDE (Aliskiren Trial in Type 2 Diabetes Using Cardio-Renal Endpoints).18

SURROGATE END POINTS: A CAVEAT

As defined by Temple,19 a surrogate end point of a clinical trial is a laboratory measurement or a physical sign used as a substitute for a clinically meaningful end point that measures directly how patients feel or function, or if they survive. Effects on surrogate end points often fail to predict the true clinical effects of an intervention, as the ONTARGET data demonstrated. Among several explanations for this failure is that interventions may affect the clinical outcome by unintended, unanticipated, and unrecognized mechanisms that operate independently of the disease process.20 Nonetheless, surrogate end point cosmetics remains attractive for many clinicians.

The ONTARGET findings indicate that there is no clinically important benefit in adding an ARB for patients with hypertension, proteinuria, heart failure, or coronary artery disease if they are already being treated with an ACE inhibitor. This would indicate that dual RAS blockade should be avoided in clinical practice until we are provided with better evidence.

References
  1. Father MD, Yusuf S, Kober L, et al. Long-term ACE-inhibitor therapy in patients with heart failure or left ventricular dysfunction: a systemic overview of data from individual patients. ACEInhibitor Myocardial Infarction Collaborative Group. Lancet 2000; 355:15751581.
  2. Pitt B, Poole-Wilson PA, Segal R, et al. Effect of losartan compared with captopril on mortality in patients with symptomatic heart failure: randomized trial—the Losartan Heart Failure Survival Study ELITE II. Lancet 2000; 355:15821587.
  3. Mann JF, Schmiede RE, McQueen M, et al; ONTARGET investigators. Renal outcomes with telmisartan, ramipril, or both, in people at high vascular risk (the ONTARGET study): a multicentre, randomized, double-blind, controlled trial. Lancet 2008; 372:547553.
  4. Yusuf S, Teo KK, Pogue J, et al; ONTARGET Investigators Telmisartan, ramipril, or both in patients at high risk for vascular events, N Engl J Med 2008; 358:15471559.
  5. van den Meiracker AH, Man in ‘t Veld AJ, Admiraal PJ, et al. Partial escape of angiotensin converting enzyme (ACE) inhibition during prolonged ACE inhibitor treatment: dose it exist and does it affect the antihypertensive response? J Hypertens 1992; 10:803812.
  6. Menard J, Campbell DJ, Azizi M, Gonzales MF. Synergistic effects of ACE inhibition and Ang II antagonism on blood pressure, cardiac weight, and renin in spontaneously hypertensive rats. Circulation 1997; 96:30723078.
  7. Mancia G, De Backer G, Dominiczak A, et al; Task Force for the Management of Arterial Hypertension of the European Society of Hypertension. 2007 guidelines for the management of arterial hypertension. J Hypertens 2007; 25:11051187.
  8. Dulton TW, He FJ, MacGregor GA. Systematic review of combined angiotensin-converting enzyme inhibition and angiotensin receptor blockade in hypertension. Hypertension 2005; 45:880886.
  9. Kunz R, Fredrich C, Wolbers M, Mann JF. Meta-analysis: effect of monotherapy and combination therapy with inhibitors of the renin angiotensin system on proteinuria in renal disease. Ann Intern Med 2008; 148:3048.
  10. Schieffer B, Schieffer E, Hilfiker-Kleiner D, et al. Expression of angiotensin II and interleukin 6 in human coronary atherosclerotic plaques: potential implications for inflammation and plaque instability. Circulation 2000; 101:13721378.
  11. Yusuf S, Sleight P, Pogue J, Bosch J, Davies R, Dagenais G. Effects of an angiotensin-converting-enzyme inhibitor, ramipril, on cardiovascular events in high-risk patients. The Heart Outcomes Prevention Evaluation Study Investigators. N Engl J Med 2000; 342:145153.
  12. Fox KM; European trial On reduction of cardiac events with Perindopril in stable coronary Artery disease Investigators. Efficacy of perindopril in reduction of cardiovascular events among patients with stable coronary artery disease: randomized, double-blind, placebo-controlled, multicentre trial (the EUROPA study). Lancet 2003; 362:782788.
  13. Dahlöf B, Devereux RB, Kjeldsen SE, et al; LIFE Study Group. Cardiovascular morbidity and mortality in the Losartan Intervention For Endpoint reduction in hypertension study (LIFE): a randomized trial against atenolol. Lancet 2002; 359:9951003.
  14. McMurray JJ, Ostergren J, Swedberg K, et al; CHARM Investigators and Committees. Effects of candesartan in patients with chronic heart failure and reduced leftventricular systolic function taking angiotensin converting enzyme inhibitors: the CHARM-Added trial. Lancet 2003; 362:767771.
  15. Pfeffer MA, McMurray JJ, Velzquez E, et al; Valsartan in Acute Myocardial Infarction Trial Investigators. Valsartan, captopril, or both in myocardial infarction complicated by heart failure, left ventricular dysfunction, or both. N Engl J Med 2003; 349:18931906.
  16. Cohn JN, Tognoni GValsartan Heart Failure Trial Investigators. A randomized trial of the angiotensin receptor blocker valsartan in chronic heart failure. N Engl J Med 2001: 345:16671675.
  17. Lakhdar R, Al-Mallah MH, Lanfear DE. Safety and tolerability of angiotensin-converting enzyme inhibitor versus the combination of angiotensin-converting enzyme inhibitor and angiotensin receptor blocker in patients with left ventricular dysfunction: a systematic review and meta-analysis of randomized controlled trials. J Card Fail 2008; 14:181188.
  18. Parving H-H, Brenner BM, McMurray JJV, et al. Aliskiren Trial in Type 2 Diabetes Using Cardio-Renal Endpoints (ALTITUDE): rationale and study design [published online ahead of print January 14, 2009]. Nephrol Dial Transplant. doi:10.1093/ndt/gfn721.
  19. Temple RJ. A regulatory authority’s opinion about surrogate endpoints. In:Nimmo WS, Tucker GT, eds. Clinical Measurement in Drug Evaluation. J Wiley: New York, 1995.
  20. Messerli FH. The sudden demise of dual renin-angiotensin system blockade or the soft science of the surrogate end point. J Am Coll Cardiol 2009; 53:468470.
References
  1. Father MD, Yusuf S, Kober L, et al. Long-term ACE-inhibitor therapy in patients with heart failure or left ventricular dysfunction: a systemic overview of data from individual patients. ACEInhibitor Myocardial Infarction Collaborative Group. Lancet 2000; 355:15751581.
  2. Pitt B, Poole-Wilson PA, Segal R, et al. Effect of losartan compared with captopril on mortality in patients with symptomatic heart failure: randomized trial—the Losartan Heart Failure Survival Study ELITE II. Lancet 2000; 355:15821587.
  3. Mann JF, Schmiede RE, McQueen M, et al; ONTARGET investigators. Renal outcomes with telmisartan, ramipril, or both, in people at high vascular risk (the ONTARGET study): a multicentre, randomized, double-blind, controlled trial. Lancet 2008; 372:547553.
  4. Yusuf S, Teo KK, Pogue J, et al; ONTARGET Investigators Telmisartan, ramipril, or both in patients at high risk for vascular events, N Engl J Med 2008; 358:15471559.
  5. van den Meiracker AH, Man in ‘t Veld AJ, Admiraal PJ, et al. Partial escape of angiotensin converting enzyme (ACE) inhibition during prolonged ACE inhibitor treatment: dose it exist and does it affect the antihypertensive response? J Hypertens 1992; 10:803812.
  6. Menard J, Campbell DJ, Azizi M, Gonzales MF. Synergistic effects of ACE inhibition and Ang II antagonism on blood pressure, cardiac weight, and renin in spontaneously hypertensive rats. Circulation 1997; 96:30723078.
  7. Mancia G, De Backer G, Dominiczak A, et al; Task Force for the Management of Arterial Hypertension of the European Society of Hypertension. 2007 guidelines for the management of arterial hypertension. J Hypertens 2007; 25:11051187.
  8. Dulton TW, He FJ, MacGregor GA. Systematic review of combined angiotensin-converting enzyme inhibition and angiotensin receptor blockade in hypertension. Hypertension 2005; 45:880886.
  9. Kunz R, Fredrich C, Wolbers M, Mann JF. Meta-analysis: effect of monotherapy and combination therapy with inhibitors of the renin angiotensin system on proteinuria in renal disease. Ann Intern Med 2008; 148:3048.
  10. Schieffer B, Schieffer E, Hilfiker-Kleiner D, et al. Expression of angiotensin II and interleukin 6 in human coronary atherosclerotic plaques: potential implications for inflammation and plaque instability. Circulation 2000; 101:13721378.
  11. Yusuf S, Sleight P, Pogue J, Bosch J, Davies R, Dagenais G. Effects of an angiotensin-converting-enzyme inhibitor, ramipril, on cardiovascular events in high-risk patients. The Heart Outcomes Prevention Evaluation Study Investigators. N Engl J Med 2000; 342:145153.
  12. Fox KM; European trial On reduction of cardiac events with Perindopril in stable coronary Artery disease Investigators. Efficacy of perindopril in reduction of cardiovascular events among patients with stable coronary artery disease: randomized, double-blind, placebo-controlled, multicentre trial (the EUROPA study). Lancet 2003; 362:782788.
  13. Dahlöf B, Devereux RB, Kjeldsen SE, et al; LIFE Study Group. Cardiovascular morbidity and mortality in the Losartan Intervention For Endpoint reduction in hypertension study (LIFE): a randomized trial against atenolol. Lancet 2002; 359:9951003.
  14. McMurray JJ, Ostergren J, Swedberg K, et al; CHARM Investigators and Committees. Effects of candesartan in patients with chronic heart failure and reduced leftventricular systolic function taking angiotensin converting enzyme inhibitors: the CHARM-Added trial. Lancet 2003; 362:767771.
  15. Pfeffer MA, McMurray JJ, Velzquez E, et al; Valsartan in Acute Myocardial Infarction Trial Investigators. Valsartan, captopril, or both in myocardial infarction complicated by heart failure, left ventricular dysfunction, or both. N Engl J Med 2003; 349:18931906.
  16. Cohn JN, Tognoni GValsartan Heart Failure Trial Investigators. A randomized trial of the angiotensin receptor blocker valsartan in chronic heart failure. N Engl J Med 2001: 345:16671675.
  17. Lakhdar R, Al-Mallah MH, Lanfear DE. Safety and tolerability of angiotensin-converting enzyme inhibitor versus the combination of angiotensin-converting enzyme inhibitor and angiotensin receptor blocker in patients with left ventricular dysfunction: a systematic review and meta-analysis of randomized controlled trials. J Card Fail 2008; 14:181188.
  18. Parving H-H, Brenner BM, McMurray JJV, et al. Aliskiren Trial in Type 2 Diabetes Using Cardio-Renal Endpoints (ALTITUDE): rationale and study design [published online ahead of print January 14, 2009]. Nephrol Dial Transplant. doi:10.1093/ndt/gfn721.
  19. Temple RJ. A regulatory authority’s opinion about surrogate endpoints. In:Nimmo WS, Tucker GT, eds. Clinical Measurement in Drug Evaluation. J Wiley: New York, 1995.
  20. Messerli FH. The sudden demise of dual renin-angiotensin system blockade or the soft science of the surrogate end point. J Am Coll Cardiol 2009; 53:468470.
Issue
Cleveland Clinic Journal of Medicine - 76(12)
Issue
Cleveland Clinic Journal of Medicine - 76(12)
Page Number
693-696
Page Number
693-696
Publications
Publications
Topics
Article Type
Display Headline
Is an ACE inhibitor plus an ARB more effective than either drug alone?
Display Headline
Is an ACE inhibitor plus an ARB more effective than either drug alone?
Sections
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media

Low bone density is not always bisphosphonate deficiency

Article Type
Changed
Thu, 04/12/2018 - 10:55
Display Headline
Low bone density is not always bisphosphonate deficiency

In general medical practice, low bone mineral density is generally assumed to be osteoporosis. Once vitamin D levels have been evaluated (and corrected, if low) and a few potential causes of secondary osteoporosis excluded, patients with significantly low T scores are often treated with bisphosphonates to blunt the overall osteoporotic process and reduce the likelihood of future fragility fractures.

A few common conditions dramatically underscore the potential difficulties in distinguishing the hyperosteolytic biology of osteoporosis from disorders of bone hypoproduction or defective mineralization. Patients with a severe or complicated gastrointestinal malabsorptive state such as a history of gastric bypass surgery are included in this group. Management of transplantation patients may also be challenging.

But perhaps the most complicated metabolic bone patients to manage are those with severe chronic kidney disease. In this issue of the Journal, Dr. Paul Miller and, in an accompanying commentary, Dr. Maria Coco discuss the problems, some potential bone-protective strategies, and some of the controversies faced by clinicians treating bone disease in patients with chronic kidney disease.

While patients with chronic kidney disease who have low T scores are often comanaged by nephrologists and specialists in metabolic bone disease, the discussion of the pathophysiologic pathways resulting in reduced bone density is germane to many of us. A documented low T score does not equal osteoporosis and thus should not lead us to automatically prescribe a bisphosphonate.

Clues to the presence of a disease associated with secondary osteoporosis or osteomalacia should be sought in any patient with a low T score. Some of these clues are adenopathy on examination, a personal or striking family history of nephrolithiasis, unexplained anemia, thyroid disease, a low anion gap, an unexplained change in blood pressure, a particularly alkaline urine, frequent loose stools, and disturbances of phosphate or calcium.

The era of ignoring osteoporosis is fortunately coming to a close. But we should not cavalierly go where the generation of internists before us could not go—to our prescription pads. Low bone density is not a one-size-fits-all disorder. We need to carefully consider other diagnostic and therapeutic options before assuming that low bone density is due to osteoporosis in every patient. These two articles should stimulate serious thought about possible alternative diagnoses to the now frequently diagnosed “osteoporosis.”

Article PDF
Author and Disclosure Information

Brian F. Mandell, MD, PhD
Editor in Chief

Issue
Cleveland Clinic Journal of Medicine - 76(12)
Publications
Topics
Page Number
683
Sections
Author and Disclosure Information

Brian F. Mandell, MD, PhD
Editor in Chief

Author and Disclosure Information

Brian F. Mandell, MD, PhD
Editor in Chief

Article PDF
Article PDF
Related Articles

In general medical practice, low bone mineral density is generally assumed to be osteoporosis. Once vitamin D levels have been evaluated (and corrected, if low) and a few potential causes of secondary osteoporosis excluded, patients with significantly low T scores are often treated with bisphosphonates to blunt the overall osteoporotic process and reduce the likelihood of future fragility fractures.

A few common conditions dramatically underscore the potential difficulties in distinguishing the hyperosteolytic biology of osteoporosis from disorders of bone hypoproduction or defective mineralization. Patients with a severe or complicated gastrointestinal malabsorptive state such as a history of gastric bypass surgery are included in this group. Management of transplantation patients may also be challenging.

But perhaps the most complicated metabolic bone patients to manage are those with severe chronic kidney disease. In this issue of the Journal, Dr. Paul Miller and, in an accompanying commentary, Dr. Maria Coco discuss the problems, some potential bone-protective strategies, and some of the controversies faced by clinicians treating bone disease in patients with chronic kidney disease.

While patients with chronic kidney disease who have low T scores are often comanaged by nephrologists and specialists in metabolic bone disease, the discussion of the pathophysiologic pathways resulting in reduced bone density is germane to many of us. A documented low T score does not equal osteoporosis and thus should not lead us to automatically prescribe a bisphosphonate.

Clues to the presence of a disease associated with secondary osteoporosis or osteomalacia should be sought in any patient with a low T score. Some of these clues are adenopathy on examination, a personal or striking family history of nephrolithiasis, unexplained anemia, thyroid disease, a low anion gap, an unexplained change in blood pressure, a particularly alkaline urine, frequent loose stools, and disturbances of phosphate or calcium.

The era of ignoring osteoporosis is fortunately coming to a close. But we should not cavalierly go where the generation of internists before us could not go—to our prescription pads. Low bone density is not a one-size-fits-all disorder. We need to carefully consider other diagnostic and therapeutic options before assuming that low bone density is due to osteoporosis in every patient. These two articles should stimulate serious thought about possible alternative diagnoses to the now frequently diagnosed “osteoporosis.”

In general medical practice, low bone mineral density is generally assumed to be osteoporosis. Once vitamin D levels have been evaluated (and corrected, if low) and a few potential causes of secondary osteoporosis excluded, patients with significantly low T scores are often treated with bisphosphonates to blunt the overall osteoporotic process and reduce the likelihood of future fragility fractures.

A few common conditions dramatically underscore the potential difficulties in distinguishing the hyperosteolytic biology of osteoporosis from disorders of bone hypoproduction or defective mineralization. Patients with a severe or complicated gastrointestinal malabsorptive state such as a history of gastric bypass surgery are included in this group. Management of transplantation patients may also be challenging.

But perhaps the most complicated metabolic bone patients to manage are those with severe chronic kidney disease. In this issue of the Journal, Dr. Paul Miller and, in an accompanying commentary, Dr. Maria Coco discuss the problems, some potential bone-protective strategies, and some of the controversies faced by clinicians treating bone disease in patients with chronic kidney disease.

While patients with chronic kidney disease who have low T scores are often comanaged by nephrologists and specialists in metabolic bone disease, the discussion of the pathophysiologic pathways resulting in reduced bone density is germane to many of us. A documented low T score does not equal osteoporosis and thus should not lead us to automatically prescribe a bisphosphonate.

Clues to the presence of a disease associated with secondary osteoporosis or osteomalacia should be sought in any patient with a low T score. Some of these clues are adenopathy on examination, a personal or striking family history of nephrolithiasis, unexplained anemia, thyroid disease, a low anion gap, an unexplained change in blood pressure, a particularly alkaline urine, frequent loose stools, and disturbances of phosphate or calcium.

The era of ignoring osteoporosis is fortunately coming to a close. But we should not cavalierly go where the generation of internists before us could not go—to our prescription pads. Low bone density is not a one-size-fits-all disorder. We need to carefully consider other diagnostic and therapeutic options before assuming that low bone density is due to osteoporosis in every patient. These two articles should stimulate serious thought about possible alternative diagnoses to the now frequently diagnosed “osteoporosis.”

Issue
Cleveland Clinic Journal of Medicine - 76(12)
Issue
Cleveland Clinic Journal of Medicine - 76(12)
Page Number
683
Page Number
683
Publications
Publications
Topics
Article Type
Display Headline
Low bone density is not always bisphosphonate deficiency
Display Headline
Low bone density is not always bisphosphonate deficiency
Sections
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media