User login
CCJM delivers practical clinical articles relevant to internists, cardiologists, endocrinologists, and other specialists, all written by known experts.
Copyright © 2019 Cleveland Clinic. All rights reserved. The information provided is for educational purposes only. Use of this website is subject to the disclaimer and privacy policy.
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
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')]
Fragility fractures in chronic kidney disease: An opinion-based approach
But even in chronic kidney disease, many fractures are due to postmenopausal or agerelated osteoporosis, and estrogen-deficiency osteoporosis is the most common cause of fragility fractures overall.1–3 Osteoporosis can be diagnosed only after other causes of skeletal fragility have been ruled out. And how to diagnose and treat osteoporosis in the most severe stage of kidney disease is a matter of opinion, as we have almost no data to guide us.
Nevertheless, in the pages that follow, I will outline my admittedly opinion-based approach to diagnosing and managing the causes of fragility fractures in patients with chronic kidney disease.
T SCORES DO NOT DISTINGUISH THE CAUSES OF FRAGILITY
The most common cause of fragility fractures is osteoporosis due to estrogen deficiency.1–3 But since many other medical conditions can lead to osteoporosis, simple diagnostic criteria have been difficult to find.
Before 1994, the diagnosis of osteoporosis was made on the basis of low-trauma fractures.4 Now, we use the World Health Organization criteria,5 based on bone mineral density T scores:
- Normal—a T score of −1.0 standard deviations or higher
- Osteopenia—a T score of less than −1.0 but higher than −2.5
- Osteoporosis—a T score of −2.5 or less
- Severe osteoporosis—a T score of −2.5 or less with a fragility fracture.
However, fractures can also be due to metabolic bone diseases that are not osteoporosis, including renal bone diseases.6–7 While a low T score or a fracture provides a working diagnosis of osteoporosis, it does not help distinguish the different types of osteoporosis and nonosteoporotic metabolic bone diseases. For example, osteomalacia and osteogenesis imperfecta can also cause fragility fractures and can be associated with low bone density. Using these criteria to define osteoporosis is even more problematic in patients with chronic kidney disease.
FIVE STAGES OF CHRONIC KIDNEY DISEASE
The National Kidney Foundation8 classifies the severity of chronic kidney disease on the basis of the glomerular filtration rate (GFR), as measured by 24-hour urine for creatinine clearance, or as estimated by the Cockcroft-Gault equation or, preferably, the Modification of Diet in Renal Disease (MDRD) equation (calculators are available at www.kidney.org/professionals/kdoqi/gfr_calculator.cfm):
- Stage 1—GFR 90 mL/minute/1.73 m2 or higher
- Stage 2—GFR 60 to 89
- Stage 3—GFR 30 to 59
- Stage 4—GFR 15 to 29
- Stage 5—GFR lower than 15, or if the patient is on dialysis. (Another stage, called 5D, was added to the list to denote patients on dialysis, since the metabolic derangements in bone and systemic biology may differ between patients on dialysis vs those not on dialysis.)
This staging system is relevant to the discussion of bone fragility that follows.
CHRONIC KIDNEY DISEASE IS COMMON IN THE ELDERLY
The third National Health and Nutrition Examination Survey9 found that, at least as estimated by the Cockcroft-Gault equation, the GFR declines with age, so that by the age of 70 at least 20% of the US population has stage 4 or 5 chronic kidney disease.
Although the Cockcroft-Gault and MDRD equations may yield lower GFR values in the general population than one would get by measuring creatinine, inulin, or iothalamate clearance,10,11 the point is that both osteoporosis and chronic kidney disease are common.
THE GAMUT OF RENAL OSTEODYSTROPHY
In kidney failure (stage 5 chronic kidney disease), all forms of renal osteodystrophy may be associated with fragility fractures. Renal osteodystrophy can be defined by quantitative bone histomorphometry.12,13 The systemic conditions that may be associated with the bone disease and systemic vascular disease (chronic kidney disease–mineral and bone disorder) are characterized by one or more of the following14:
- Abnormalities of calcium, phosphorus, parathyroid hormone, or vitamin D metabolism
- Abnormalities in bone turnover, mineralization, volume, linear growth, or strength
- Vascular or other soft-tissue calcification.
The National Kidney Foundation14 classifies renal osteodystrophy on the basis of:
- Turnover—high, normal, or low
- Mineralization—normal or abnormal
- Volume—high, normal, or low.
Although this system helps us understand these diseases better, it does not provide a working diagnosis of osteoporosis.14
WHAT IS OSTEOPOROSIS?
In an attempt to define osteoporosis by a pathophysiologic mechanism, the National Institutes of Health15 have held two consensus conferences and have stated that “osteoporosis is a skeletal disorder characterized by impairment in bone strength predisposing a person to an increased risk of fracture. Bone strength primarily reflects the integration of bone density and bone quality.”15 However, the consensus statement also does not provide a working diagnosis of osteoporosis—one that clinicians can apply to management decisions, and one that is also accepted by the US International Classification of Disease codes for reimbursement purposes.
The 1994 World Health Organization criteria offer the most pragmatic operational definition of osteoporosis, and they can be applied in both men and women, as well as in younger patients with medical conditions associated with increased risk of low-trauma fracture.5,16 Although the main purpose of these criteria was to advise international health authorities of the potential future economic impact of osteoporosis, the T score also became the pragmatic diagnostic threshold for defining normal, osteopenia, and osteoporosis in clinical practice.
The T score also calls attention to an important observation: of people who have fractures and subsequently undergo bone densitometry, more are found to have osteopenia than osteoporosis. The reasons are that there are more people with osteopenia than osteoporosis,17,18 and many other factors independent of low bone mineral density contribute to bone strength.19,20
How is osteoporosis diagnosed in stage 1–3 chronic kidney disease?
In patients with chronic kidney disease who develop fragility fractures, the reasonable question is: Is the cause of the fracture osteoporosis or some other metabolic bone disease associated with chronic kidney disease?
The National Kidney Foundation guidelines14 say that the diagnosis of osteoporosis can be established in patients with stage 1, 2, or 3 chronic kidney disease on the basis of either of the World Health Organization criteria, ie, a T score of −2.5 or lower or fragility fractures, as in the postmenopausal population, as long as there are no biochemical abnormalities that suggest chronic kidney disease–mineral and bone disorder.
How is osteoporosis diagnosed in stage 4 or 5 chronic kidney disease?
The answer is neither straightforward nor clearly defined in severe (stage 4 or 5) chronic kidney disease.
In stage 5 and especially in patients on dialysis, the derangements in bone and mineral metabolism become serious enough to impair bone strength and increase the risk of lowtrauma fractures. The risk of hip fracture in stage 5 may be four times higher than in agematched controls.21–24
Adynamic, severe hyperparathyroid bone disease as well as osteomalacia can be associated with a higher risk of fragility fractures than in aged-matched controls in population studies of postmenopausal women or elderly men. These are bone fragility conditions that are not osteoporosis but that can mimic osteoporosis by the World Health Organization criteria.
Thus, when a patient in stage 5 has severe fragility fractures that by themselves may be life-threatening, it is reasonable to ask if the drugs that reduce the risk of fractures in many other osteoporotic conditions (postmenopausal, steroid-induced, elderly male osteoporosis, after solid organ transplantation) can also be used in patients with advanced chronic kidney disease.
The diagnosis of osteoporosis in these patients has no universally accepted criteria. The diagnosis is best suggested by excluding other forms of renal osteodystrophy by quantitative histomorphometry or by attempting to classify the form of renal osteodystrophy by noninvasive means of assessing bone turnover, mineralization, and volume. However, we lack clinical tools to make these distinctions in individual patients.
While many promising radiologic techniques that examine bone microarchitecture offer hope of being able to define turnover, mineralization, and volume noninvasively in severe chronic kidney disease, they are investigational and unproven at this time in discriminating between renal osteodystrophy and osteoporosis.6,25–27 As we increase our understanding of the relationships between turnover, mineralization, volume, and bone strength, these noninvasive imaging technologies may become the means to correlate turnover, mineralization, and volume to bone strength and open up an entirely new way to classify skeletal strength.
In the meantime, the clinician is left with quantitative bone histomorphometry (which requires biopsy) and biochemical markers of bone turnover to characterize the bone disease that may be responsible for low-trauma fractures in stage 5 chronic kidney disease. The clinician should first use biochemical markers before bone biopsy to distinguish the form of renal osteodystrophy, as this distinction may be able to prevent unnecessary biopsy.
Biochemical markers of bone metabolism
In chronic kidney disease, the bone biochemical tests that nephrologists usually assess during the course of declining renal function are the serum levels of:
- Phosphorus
- Parathyroid hormone
- Calcium
- Other electrolytes
- Total alkaline phosphatase or bone-specific alkaline phosphatase
- 1,25 dihydroxyvitamin D.
In postmenopausal osteoporosis, the biochemical markers of bone turnover that are measured to reflect baseline levels of bone turnover or change in bone turnover in response to drug therapy are:
- The serum or urine collagen cross-links N-telopeptide (NTx) and C-telopeptide (CTx), markers of bone resorption
- Bone-specific alkaline phosphatase (an osteoblast activity marker)
- Serum osteocalcin, a bone formation marker
- Propeptide type 1 collagen (P1NP), a marker of osteoblast activity, highly correlated with bone formation
- 25-hydroxyvitamin D levels.
Biochemical markers of bone turnover cannot be used to diagnose osteoporosis. They can, however, provide clinical guidance as to whether a patient has high or low bone turnover and whether therapy is affecting bone turnover.28–36 Although these markers have value in making these distinctions in groups of patients, they are less sensitive and specific for classifying an individual patient’s bone turnover status.
Bone-specific alkaline phosphatase, parathyroid hormone, and adynamic bone disease
If a patient’s bone-specific alkaline phosphatase level is elevated, adynamic bone disease is highly unlikely. Assuming that other causes of this elevated level (eg, Paget disease of bone, metastatic cancer) have already been excluded, the elevated level could represent either osteomalacia or hyperparathyroid bone disease.
However, a “normal” bone-specific alkaline phosphatase level does not exclude adynamic bone disease, whereas a low level is more often associated with low bone turnover.
An elevated parathyroid hormone level does not exclude adynamic renal bone disease, but a low level (< 150 pg/mL) suggests a lowbone-turnover state. A level six times or more greater than the upper limit of normal is far more likely to be associated with high bone turnover.
Thus, in clinical practice, patients with stage 4 or 5 chronic kidney disease who have elevated bone-specific alkaline phosphatase or very high parathyroid hormone values do not have adynamic bone disease. Furthermore, once other causes of these aberrant biochemical abnormalities have been defined, then “high-bone-turnover osteoporosis” may be a consideration. Certainly, in my opinion, if bone turnover markers suggest low bone turnover, bone biopsy is necessary before starting an antiresorptive agent.35
Quantitative bone histomorphometry
Double tetracycline-labeled quantitative histomorphometry is still the only accepted way to measure turnover, mineralization, and volume in clinical practice.43–45 A committee of the American Society for Bone and Mineral Research has developed histomorphometric criteria for distinguishing among the different types of metabolic bone diseases (osteomalacia, adynamic bone disease, hyperparathyroid bone disease).12 These criteria can be used to distinguish among the various metabolic bone diseases that accompany stage 5 chronic kidney disease, including adynamic bone disease.43,46–48
For patients in stage 5 who have had a fragility fracture, adynamic bone disease should be excluded before the off-label use of an osteoporosis drug that reduces bone turnover, such as a bisphosphonate, calcitonin, estrogen, a selective estrogen receptor modulator, or denosumab (anti-RANK ligand antibody). While there is no evidence, for example, that starting a bisphosphonate in a patient who already has adynamic bone disease is detrimental to either bone strength or systemic vascular calcification (which may be linked to low bone turnover),49 it seems unreasonable to do so until solid prospective data clarify the harm or benefit.50 Preliminary experimental and clinical data suggest that bisphosphonates may even reduce progression of extraosseous calcification and inhibit the development of atherosclerosis.50
Hence, quantitative bone histomorphometry can discriminate among the various forms of renal osteodystrophy. If a distinct form of renal osteodystrophy is not present in a patient with stage 4 or 5 chronic kidney disease who has had a fracture and who, on biopsy, has a low trabecular bone volume, the patient probably has osteoporosis by exclusion.
TREATING OSTEOPOROSIS IN STAGE 1–3 CHRONIC KIDNEY DISEASE
As previously mentioned, patients who have fragility fractures in stage 1, 2, or 3 chronic kidney disease are more likely to have osteoporosis than renal osteodystrophy as the cause of their impaired bone strength. Although several articles have described a higher risk of fragility fractures in patients with age-related reduction in renal function than in agematched patients with normal renal function, the specific metabolic bone disease other than osteoporosis accounting for this bone fragility has not been defined.6
Hence, patients with osteoporosis who are in stage 1, 2, or 3 chronic kidney disease and do not have a known biochemical abnormality that might suggest some form of renal osteodystrophy can and should be considered for treatment with approved drugs that reduce the risk of fractures in postmenopausal, male, or glucocorticoid-induced osteoporosis.51–53 In clinical trials, these agents were shown to be effective in patients with serum creatinine concentrations as high as 2.0 mg/dL or a GFR as low as 30 mL/min, as estimated by the Cockcroft-Gault equation.
While all of the approved agents show evidence of reducing the risk of vertebral fractures, patients at higher risk of fractures or those who have already suffered a nonvertebral fracture are more often considered candidates for treatment with a bisphosphonate or teriparatide (Forteo), both of which have shown evidence of reducing the risk of all fractures.
Bisphosphonates in stage 1–3 chronic kidney disease
There is prospective evidence that patients with an age-related reduction in GFR down to 30 mL/min benefit from oral and intravenous bisphosphonates, since all of the clinical trials that led to the approval of bisphosphonates included patients with GFRs as low as this.54–57 Bisphosphonates seem to have an excellent safety profile as measured by renal adverse events in patients with a GFR of 30 mL/min or greater.52–59
From the intravenous bisphosphonate studies, it appears that ibandronate (Boniva) at the approved dose of 3 mg intravenously every 3 months and zoledronic acid (Reclast) 5 mg once a year given over 15 minutes are safe in patients with a GFR greater than 30 or 35 mL/min.
However, the safety of these drugs might not be the same in patients with preexisting renal parenchymal disease (eg, in diabetes) or in patients using other agents that could affect renal function (eg, nonsteroidal antiinflammatory drugs). Therefore, caution is still needed when deciding to use intravenous bisphosphonates in specific higher-risk renal subpopulations.
In the clinical trials of zoledronic acid, a substantial proportion of patients had diabetes, and no difference was seen in adverse renal effects between diabetic and nondiabetic patients. Also, GFRs declined equally between the treated and placebo groups over time and were no different at the end of 3 years.55 However, in patients in whom serum creatinine was measured 9 to 11 days after the infusion of zoledronic acid, there was a small but statistically significant transient increase in serum creatinine concentration (0.5–2.0 mg/dL above baseline) after the second annual infusion. 58 The serum creatinine concentrations returned to their baseline values in all of these patients before the next annual infusion.
It is important that infusions of zoledronic acid be given no faster than over 15 minutes. More rapid infusion has been associated with acute renal failure, suggesting that the tubular damage that mimics acute tubular necrosis is related to the maximal concentration and not to the area under the curve. I infuse zoledronic acid over 30 minutes in patients with normal renal function or in those with stage 1, 2, or 3 chronic kidney disease.
Teriparatide
Teriparatide’s approval trial did not require baseline measurements of GFR, but patients were enrolled only if their baseline serum creatinine concentrations were less than 2.0 mg/dL.60 In a post hoc analysis, a small subset of patients had GFRs as low as 30 mL/min as estimated by the Cockcroft-Gault equation. In these patients, teriparatide 20 or 40 μg/day had an anabolic effect as measured by increases in osteoblast activity markers and bone mineral density, similar to that seen in patients with higher estimated GFRs and without any adverse renal effects.61
There are no data on using teriparatide in stage 4 or 5 chronic kidney disease, and I emphasize that in all of the clinical trials of teriparatide, all patients, even those with estimated GFRs as low as 30 mL/min, had normal baseline serum intact parathyroid hormone levels. It is possible that the bone biologic response could differ between patients with chronic kidney disease who have an elevated as compared with a normal serum parathyroid hormone level. This issue should be investigated.
TREATING OSTEOPOROSIS IN STAGE 4 OR 5 CHRONIC KIDNEY DISEASE
Treatment decisions are more difficult in patients with stage 4 and especially stage 5 chronic kidney disease who have had fragility fractures. This is even the case when the clinician has determined to the best of his or her ability that the patient has osteoporosis rather than renal osteodystrophy.
There are no prospective data showing any of the approved drugs to be effective in treating osteoporosis in patients whose GFRs are lower than 30 mL/min. However, a post hoc analysis of pooled data from nine clinical trials62 found that risedronate (Actonel) 5 mg/day reduced the incidence of new vertebral fractures. Another post hoc analysis, from the Fracture Intervention Trial,63 found that alendronate (Fosamax) 5 mg/day for the first 2 years and 10 mg/day for the 3rd year reduced the incidence of all clinical fractures. In neither of these post hoc analyses did the drug affect the serum creatinine concentration. The patients—postmenopausal women—had GFRs as low as 15 mL/min as estimated by the Cockcroft-Gault equation. Similar post hoc data have been published on raloxifene (Evista).64
There are no data on the efficacy (reduction in fracture risk) or safety of any bisphosphonate in patients with GFRs lower than 15 mL/min (stage 5 chronic kidney disease). Nevertheless, the question often arises when fragility fractures occur in this population. Here, only opinion and controversy exist, and we fervently await good science and randomized prospective data.
How to manage renal bone disease after transplantation is a distinctly separate issue in which bisphosphonate use may be even more controversial than in end-stage renal disease.65,66
In my opinion, patients without fractures with stage 5 chronic kidney disease should not be given bisphosphonates or teriparatide offlabel. Treating only on the basis of low bone mineral density and other risk factors seems to be associated with greater risk than benefit.
In stage 5 patients suffering fragility fractures, a bisphosphonate may be considered, but only after renal osteodystrophy has been thoroughly ruled out, which most often requires a bone biopsy.43,67,68 In skilled hands, transiliac bone biopsy is a safe procedure with little morbidity.
If osteoporosis appears to be the cause of the fracture, and if one chooses to use a bisphosphonate and the patient gives his or her informed consent, then I would give half the usual dose and restrict the therapy to no more than 3 years. The reason for halving the dose is based on the known pharmacokinetics of bisphosphonates in people with normal renal function: 50% of a given dose goes to bone and 50% is excreted by the kidney. Furthermore, the dialyzability of bisphosphonates has not been well studied. Limiting the treatment to 3 years is based on the unknown but probably greater bone retention of bisphosphonates when excretion is impaired.
I must emphasize that these approaches are not based on any evidence of efficacy, but rather are considered in extreme cases of often-recurrent fragility fractures in which the fractures per se pose a great risk of morbidity and death. These approaches should be clearly discussed with the patient, undertaken by specialists knowledgeable in complex metabolic bone disease management, and initiated only after the skeletal fragility disorder is well characterized.
SUMMING UP
No consensus exists on the criteria for diagnosing osteoporosis in stage 4 or 5 chronic kidney disease.
In higher-risk patients in stage 1, 2, or 3 chronic kidney disease who have osteoporosis, it appears that any drug approved for osteoporosis can be used, eg, a bisphosphonate, teriparatide, or both.
Considerations for management are far more complex in stage 4 or 5 because of the increased prevalence of other metabolic bone diseases and renal osteodystrophy, and because the World Health Organization criteria cannot be used to diagnose osteoporosis. In stage 5, the differential diagnosis requires careful analysis of a broad range of biochemical markers of bone turnover and, at times, quantitative bone histomorphometry, especially if one is considering using a bisphosphonate. It is unknown if bisphosphonates, by reducing bone turnover in a preexisting low-bone-turnover state, would help or harm bone or would lead to less or more cardiovascular disease. These questions must be addressed by better science and prospective data.
In the future, newer noninvasive radiologic tools to measure microstructure and mineralization of bone promise to help us better understand osteoporosis and renal osteodystrophy in a noninvasive manner.
In clinical practice, at the current time and with current limited knowledge, treatment of osteoporosis in stage 4 or 5 chronic kidney disease is opinion-based. Nevertheless, in very specific clinical cases of severe fragility fractures that, by themselves, may cause disability and death, bisphosphonates should be considered by experts in bone metabolism and, as with any off-label application, after careful informed discussions with the patient.
- Melton LJ. Epidemiology worldwide. Endocrinol Metab Clin North Am 2003; 32:1–13.
- Burge R, Dawson-Hughes B, Solomon DH, Wong JB, King A, Tosteson A. Incidence and economic burden of osteoporosis-related fractures in the United States, 2005–2025. J Bone Miner Res 2007; 22:465–475.
- Barrett-Connor E, Siris ES, Wehren LE, et al. Osteoporosis and fracture risk in women of different ethnic groups. J Bone Miner Res 2005; 20:185–194.
- Assessment of fracture risk and its application to screening for postmenopausal osteoporosis. Report of a WHO Study Group. World Health Organ Tech Rep Ser 1994; 843:1–129.
- Miller PD, Bonnick SL. Clinical application of bone densitometry. In:Favus MJ, editor. Primer on the Metabolic Bone Diseases and Disorders of Mineral Metabolism. 4th ed. American Society for Bone and Mineral Research. Philadelphia, PA: Lippincott Williams & Wilkins; 1999.
- Nickolas TL, Leonard MB, Shane E. Chronic kidney disease and bone fracture: a growing concern. Kidney Int 2008; 74:721–731.
- Gal-Moscovici A, Sprague SM. Osteoporosis and chronic kidney disease. Semin Dial 2007; 20:423–430.
- National Kidney Foundation. Clinical practice guidelines for bone metabolism and disease in chronic kidney disease. Am J Kidney Dis 2003; 42(suppl 3):S1–S201.
- Coresh J, Astor BC, Greene T, Eknoyan G, Levey AS. Prevalence of chronic kidney disease and decreased kidney function in the adult US population: Third National Health and Nutrition Examination Survey. Am J Kidney Dis 2003; 41:1–12.
- Bennett WM. Reporting eGFR. Clin J Am Soc Nephrol 2008; 3:1561–1562.
- Glassock RJ, Winearls C. Screening for CKD with eGFR: doubts and dangers. Clin J Am Soc Nephrol 2008; 3:1563–1568.
- Parfitt AM, Drezner M, Glorieux F, et al. Bone histomorphometry: standardization of nomenclature, symbols, and units. Report of the ASBMR Histomorphometry Nomenclature Committee. J Bone Miner Res 1987; 2:595–610.
- Andress DL, Sherrard DJ. The osteodystrophy of chronic renal failure. In:Schrier RW, editor: Diseases of the Kidney and Urinary Tract. Philadelphia, PA: Lippincott Williams & Wilkins; 2007:2431–2453.
- Moe S, Drueke T, Cunningham J, et al. Definition, evaluation, and classification of renal osteodystrophy: a position statement from Kidney Disease: Improving Global Outcomes (KDIGO). Kidney Int 2006; 69:1945–1953.
- NIH Consensus Development Panel Osteoporosis Prevention Diagnosis and Therapy. Osteoporosis prevention, diagnosis, and therapy. JAMA 2001; 285:785–795.
- Baim S, Binkley N, Bilezikian JP, et al. Official positions of the International Society for Clinical Densitometry and executive summary of the 2007 ISCD Position Development Conference. J Clin Densitom 2008; 11:75–91.
- Siris E, Miller P, Barrett-Connor E, et al. Identification and fracture outcomes of undiagnosed low bone mineral density in postmenopausal women: results from the National Osteoporosis Risk Assessment (NORA). JAMA 2001; 286:2815–2822.
- Schuit SCE, Oei HH, Witteman JC, et al. Fracture incidence and association with bone mineral density in elderly men and women: the Rotterdam Study. Bone 2004; 34:195–202.
- Bouxsein ML. Non-invasive measurements of bone strength: promise and peril. J Musculoskelet Neuronal Interact 2004; 4:404–405.
- Seeman E. Bone quality: the material and structural basis of bone strength. J Bone Miner Metab 2008; 26:1–8.
- Alem AM, Sherrard DJ, Gillen DL, et al. Increased risk of hip fracture among patients with end-stage renal disease. Kidney Int 2000; 58:396–399.
- Ball AM, Gillen DL, Sherrard D, et al. Risk of hip fracture among dialysis and renal transplant recipients. JAMA 2002; 288:3014–3018.
- Jadoul M, Albert JM, Akiba T, et al. Incidence and risk factors for hip or other bone fractures among hemodialysis patients in the Dialysis Outcomes and Practice Patterns Study. Kidney Int 2006; 70:1358–1366.
- Stehman-Breen CO, Sherrard DJ, Alem AM, et al. Risk factors for hip fracture among patients with end-stage renal disease. Kidney Int 2000; 58:2200–2205.
- Wehrli FW, Leonard MB, Saha PK, Gomberg BR. Quantitative highresolution magnetic resonance imaging reveals structural implications of renal osteodystrophy on trabecular and cortical bone. J Magn Reson Imaging 2004; 20:83–89.
- Genant HK, Lang TF, Engelke K, et al. Advances in the noninvasive assessment of bone density, quality, and structure. Calcif Tissue Int 1996; 59(suppl 1):S10–S15.
- Roschger P, Paschalis EP, Fratzl P, Klaushofer K. Bone mineralization density distribution in health and disease. Bone 2008; 42:456–466.
- Miller PD. Bone density and markers of bone turnover in predicting fracture risk and how changes in these measures predict fracture risk reduction. Curr Osteoporos Rep 2005; 3:103–110.
- Miller PD, Hochberg MC, Wehren LE, Ross PD, Wasnich RD. How useful are measures of BMD and bone turnover? Curr Med Res Opin 2005; 21:545–554.
- Chavassieux PM, Delmas PD. Bone remodeling: biochemical markers or bone biopsy? J Bone Miner Res 2006; 21:178–179.
- Garnero P. Biomarkers for osteoporosis management: utility in diagnosis, fracture risk prediction and therapy monitoring. Mol Diagn Ther 2008; 12:157–170.
- Hochberg M, Greenspan S, Wasnich R, Miller P, Thompson D, Ross P. Changes in bone density and turnover explain the reductions in incidence of nonvertebral fractures that occur during treatment with antiresorptive agents. J Clin Endocrinol Metab 2002; 87:1586–1592.
- Bouxsein ML, Delmas PD. Considerations for development of surrogate endpoints for antifracture efficacy of new treatments in osteoporosis: a perspective. J Bone Miner Res 2008; 23:1155–1167.
- Chen P, Satterwhite JH, Licatta AA, et al. Early changes in biochemical markers of bone formation predict BMD response to teriparatide in postmenopausal women with osteoporosis. J Bone Miner Res 2005; 20:962–970.
- Miller PD, Delmas PD, Lindsay R, et al. Early responsiveness of women with osteoporosis to teriparatide after therapy with alendronate or risedronate. J Clin Endocrinol Metab 2008; 93:3785–3793.
- Baim S, Miller PD. Assessing the clinical utility of serum CTX in postmenopausal osteoporosis and its use in predicting risk of osteonecrosis of the jaw. J Bone Miner Res 2009; 24:561–574.
- Miller PD, Lerma EV. Renal bone diseases. In:Kleerekoper M, Siris E, McClung M, editors. The Bone and Mineral Manual—A Practical Guide. 2nd ed. Burlington, MA: Elsevier Academic Press; 2005:127–138.
- Miller PD, Shane E. Management of transplantation renal bone disease: Interplay of bone mineral density and decisions regarding bisphosphonate use. In:Weir MR, editor. Medical Management of Kidney Transplantation. Philadelphia, PA: Lippincott Williams & Wilkins; 2004:359–375.
- Levin A, Bakris GL, Molitch M, et al. Prevalence of abnormal serum vitamin D, PTH, calcium, and phosphorus in patients with chronic kidney disease: results of the study to evaluate early kidney disease. Kidney int 2007; 71:31–38.
- Vassalotti JA, Uribarri J, Chen SC, et al. Trends in mineral metabolism: Kidney Early Evaluation Program (KEEP) and the National Health and Nutrition Examination Survey (NHANES) 1999–2004. Am J Kidney Dis 2008; 51(suppl 2):S56–S68.
- Meier C, Seibel MJ, Kraenzlin ME. Use of bone turnover markers in the real world: are we there yet? J Bone Miner Res 2009; 24:386–388.
- Lehmann G, Ott U, Kaemmerer D, Schuetze J, Wolf G. Bone histomorphometry and biochemical markers of bone turnover in patients with chronic kidney disease stages 3–5. Clin Nephrol 2008; 70:296–305.
- Miller PD. The role of bone biopsy in patients with chronic renal failure. Clin J Am Soc Nephrol 2008; 3(suppl 3):S140–S150.
- Frost HM. Tetracycline-based histological analysis of bone remodeling. Calcif Tissue Res 1969; 3:211–237.
- Hitt O, Jaworski ZF, Shimizu AG, Frost HM. Tissue-level bone formation rates in chronic renal failure, measured by means of tetracycline bone labeling. Can J Physiol Pharmacol 1970; 48:824–828.
- Coen G. Adynamic bone disease: an update and overview. J Nephrol 2005; 18:117–122.
- Parfitt AM. Renal bone disease: a new conceptual framework for the interpretation of bone histomorphometry. Curr Opin Nephrol Hypertens 2003; 12:387–403.
- Brandenburg VM, Floege J. Adynamic bone diseaseùbone and beyond. NDT Plus 2008; 3:135–147. doi:10.1093/ndtplus/sfn040.
- Hruska KA, Saab G, Mathew S, Lund R. Renal osteodystrophy, phosphate homeostasis, and vascular calcification. Semin Dial 2007; 20:309–315.
- Toussaint ND, Elder GJ, Kerr PG. Bisphosphonates in chronic kidney disease; balancing potential benefits and adverse effects on bone and soft tissue. Clin J Am Soc Nephrol 2009; 4:221–233.
- Miller PD. Is there a role for bisphosphonates in chronic kidney disease? Semin Dial 2007; 20:186–190.
- Miller PD. Bisphosphonates: pharmacology and use in the treatment of osteoporosis. In:Marcus R, Feldman D, Nelson DA, Rosen CJ, editors. Osteoporosis. 3rd ed. Boston, MA: Elsevier Academic Press; 2008:1725–1736.
- Russell RG, Watts NB, Ebetino FH, Rogers MJ. Mechanisms of action of bisphosphonates: similarities and differences and their potential influence on clinical efficacy. Osteoporosis Int 2008; 19:733–759.
- Eisman JA, Civitelli R, Adami S, et al. Efficacy and tolerability of intravenous ibandronate injections in postmenopausal osteoporosis: 2-year results from the DIVA study. J Rheumatol 2008; 35:488–497.
- Black DM, Delmas PD, Eastell RR, et al. Once-yearly zoledronic acid for treatment of postmenopausal osteoporosis. N Engl J Med 2007; 356:1809–1822.
- Lewiecki EM, Miller PD. Renal safety of intravenous bisphosphonates in the treatment of osteoporosis. Expert Opin Drug Saf 2007; 6:663–672.
- Miller PD. Anti-resorptives in the management of osteoporosis. Best Pract Res Clin Endocrinol Metab 2008; 22:849–868.
- Perazella MA, Markowitz GS. Bisphosphonate nephrotoxicity. Kidney Int 2008; 74:1385–1393.
- Boonen S, Sellmeyer DE, Lippuner K, et al. Renal safety of annual zoledronic acid infusions in osteoporotic postmenopausal women. Kidney Int 2008; 74:641–648.
- Neer RM, Arnaud CD, Zanchetta JR, et al. Effect of parathyroid hormone (1–34) on fractures and bone mineral density in postmenopausal women with osteoporosis. N Engl J Med 2001; 344:1434–1441.
- Miller PD, Schwartz EN, Chen P, Misurski DA, Krege JH. Teriparatide in postmenopausal women with osteoporosis and mild or moderate renal impairment. Osteoporosis Int 2007; 18:59–68.
- Miller PD, Roux C, Boonen S, Barton I, Dunlap L, Burgio D. Safety and efficacy of risedronate in patients with age-related reduced renal function as estimated by the Cockcroft and Gault method: a pooled analysis of nine clinical trials. J Bone Miner Res 2005; 20:2105–2115.
- Jamal SA, Bauer DC, Ensrud KE, et al. Alendronate treatment in women with normal to severely impaired renal function: an analysis of the fracture intervention trial. J Bone Miner Res 2007; 22:503–508.
- Ishani A, Blackwell T, Jamal SA, Cummings SR, Ensrud KE; MORE Investigators. The effect of raloxifene treatment in postmenopausal women with CKD. J Am Soc Nephrol 2008; 19:1430–1438.
- Coco M, Glicklich D, Faugere MC, et al. Prevention of bone loss in renal transplant recipients: a prospective, randomized trial of intravenous pamidronate. J Am Soc Nephrol 2003; 14:2669–2676.
- Palmer SC, McGregor DO, Strippoli GF. Interventions for preventing bone disease in kidney transplant recipients. Cochrane Database Syst Rev 2007;CD005015.
- Ferreira MA. Diagnosis of renal osteodystrophy: when and how to use biochemical markers and non-invasive methods; when bone biopsy is needed. Nephrol Dial Transplant 2000; 15(suppl 5):8–14.
- Trueba D, Sawaya BP, Mawad H, Malluche HH. Bone biopsy: indications, techniques, and complications. Semin Dial 2003; 16:341–345.
But even in chronic kidney disease, many fractures are due to postmenopausal or agerelated osteoporosis, and estrogen-deficiency osteoporosis is the most common cause of fragility fractures overall.1–3 Osteoporosis can be diagnosed only after other causes of skeletal fragility have been ruled out. And how to diagnose and treat osteoporosis in the most severe stage of kidney disease is a matter of opinion, as we have almost no data to guide us.
Nevertheless, in the pages that follow, I will outline my admittedly opinion-based approach to diagnosing and managing the causes of fragility fractures in patients with chronic kidney disease.
T SCORES DO NOT DISTINGUISH THE CAUSES OF FRAGILITY
The most common cause of fragility fractures is osteoporosis due to estrogen deficiency.1–3 But since many other medical conditions can lead to osteoporosis, simple diagnostic criteria have been difficult to find.
Before 1994, the diagnosis of osteoporosis was made on the basis of low-trauma fractures.4 Now, we use the World Health Organization criteria,5 based on bone mineral density T scores:
- Normal—a T score of −1.0 standard deviations or higher
- Osteopenia—a T score of less than −1.0 but higher than −2.5
- Osteoporosis—a T score of −2.5 or less
- Severe osteoporosis—a T score of −2.5 or less with a fragility fracture.
However, fractures can also be due to metabolic bone diseases that are not osteoporosis, including renal bone diseases.6–7 While a low T score or a fracture provides a working diagnosis of osteoporosis, it does not help distinguish the different types of osteoporosis and nonosteoporotic metabolic bone diseases. For example, osteomalacia and osteogenesis imperfecta can also cause fragility fractures and can be associated with low bone density. Using these criteria to define osteoporosis is even more problematic in patients with chronic kidney disease.
FIVE STAGES OF CHRONIC KIDNEY DISEASE
The National Kidney Foundation8 classifies the severity of chronic kidney disease on the basis of the glomerular filtration rate (GFR), as measured by 24-hour urine for creatinine clearance, or as estimated by the Cockcroft-Gault equation or, preferably, the Modification of Diet in Renal Disease (MDRD) equation (calculators are available at www.kidney.org/professionals/kdoqi/gfr_calculator.cfm):
- Stage 1—GFR 90 mL/minute/1.73 m2 or higher
- Stage 2—GFR 60 to 89
- Stage 3—GFR 30 to 59
- Stage 4—GFR 15 to 29
- Stage 5—GFR lower than 15, or if the patient is on dialysis. (Another stage, called 5D, was added to the list to denote patients on dialysis, since the metabolic derangements in bone and systemic biology may differ between patients on dialysis vs those not on dialysis.)
This staging system is relevant to the discussion of bone fragility that follows.
CHRONIC KIDNEY DISEASE IS COMMON IN THE ELDERLY
The third National Health and Nutrition Examination Survey9 found that, at least as estimated by the Cockcroft-Gault equation, the GFR declines with age, so that by the age of 70 at least 20% of the US population has stage 4 or 5 chronic kidney disease.
Although the Cockcroft-Gault and MDRD equations may yield lower GFR values in the general population than one would get by measuring creatinine, inulin, or iothalamate clearance,10,11 the point is that both osteoporosis and chronic kidney disease are common.
THE GAMUT OF RENAL OSTEODYSTROPHY
In kidney failure (stage 5 chronic kidney disease), all forms of renal osteodystrophy may be associated with fragility fractures. Renal osteodystrophy can be defined by quantitative bone histomorphometry.12,13 The systemic conditions that may be associated with the bone disease and systemic vascular disease (chronic kidney disease–mineral and bone disorder) are characterized by one or more of the following14:
- Abnormalities of calcium, phosphorus, parathyroid hormone, or vitamin D metabolism
- Abnormalities in bone turnover, mineralization, volume, linear growth, or strength
- Vascular or other soft-tissue calcification.
The National Kidney Foundation14 classifies renal osteodystrophy on the basis of:
- Turnover—high, normal, or low
- Mineralization—normal or abnormal
- Volume—high, normal, or low.
Although this system helps us understand these diseases better, it does not provide a working diagnosis of osteoporosis.14
WHAT IS OSTEOPOROSIS?
In an attempt to define osteoporosis by a pathophysiologic mechanism, the National Institutes of Health15 have held two consensus conferences and have stated that “osteoporosis is a skeletal disorder characterized by impairment in bone strength predisposing a person to an increased risk of fracture. Bone strength primarily reflects the integration of bone density and bone quality.”15 However, the consensus statement also does not provide a working diagnosis of osteoporosis—one that clinicians can apply to management decisions, and one that is also accepted by the US International Classification of Disease codes for reimbursement purposes.
The 1994 World Health Organization criteria offer the most pragmatic operational definition of osteoporosis, and they can be applied in both men and women, as well as in younger patients with medical conditions associated with increased risk of low-trauma fracture.5,16 Although the main purpose of these criteria was to advise international health authorities of the potential future economic impact of osteoporosis, the T score also became the pragmatic diagnostic threshold for defining normal, osteopenia, and osteoporosis in clinical practice.
The T score also calls attention to an important observation: of people who have fractures and subsequently undergo bone densitometry, more are found to have osteopenia than osteoporosis. The reasons are that there are more people with osteopenia than osteoporosis,17,18 and many other factors independent of low bone mineral density contribute to bone strength.19,20
How is osteoporosis diagnosed in stage 1–3 chronic kidney disease?
In patients with chronic kidney disease who develop fragility fractures, the reasonable question is: Is the cause of the fracture osteoporosis or some other metabolic bone disease associated with chronic kidney disease?
The National Kidney Foundation guidelines14 say that the diagnosis of osteoporosis can be established in patients with stage 1, 2, or 3 chronic kidney disease on the basis of either of the World Health Organization criteria, ie, a T score of −2.5 or lower or fragility fractures, as in the postmenopausal population, as long as there are no biochemical abnormalities that suggest chronic kidney disease–mineral and bone disorder.
How is osteoporosis diagnosed in stage 4 or 5 chronic kidney disease?
The answer is neither straightforward nor clearly defined in severe (stage 4 or 5) chronic kidney disease.
In stage 5 and especially in patients on dialysis, the derangements in bone and mineral metabolism become serious enough to impair bone strength and increase the risk of lowtrauma fractures. The risk of hip fracture in stage 5 may be four times higher than in agematched controls.21–24
Adynamic, severe hyperparathyroid bone disease as well as osteomalacia can be associated with a higher risk of fragility fractures than in aged-matched controls in population studies of postmenopausal women or elderly men. These are bone fragility conditions that are not osteoporosis but that can mimic osteoporosis by the World Health Organization criteria.
Thus, when a patient in stage 5 has severe fragility fractures that by themselves may be life-threatening, it is reasonable to ask if the drugs that reduce the risk of fractures in many other osteoporotic conditions (postmenopausal, steroid-induced, elderly male osteoporosis, after solid organ transplantation) can also be used in patients with advanced chronic kidney disease.
The diagnosis of osteoporosis in these patients has no universally accepted criteria. The diagnosis is best suggested by excluding other forms of renal osteodystrophy by quantitative histomorphometry or by attempting to classify the form of renal osteodystrophy by noninvasive means of assessing bone turnover, mineralization, and volume. However, we lack clinical tools to make these distinctions in individual patients.
While many promising radiologic techniques that examine bone microarchitecture offer hope of being able to define turnover, mineralization, and volume noninvasively in severe chronic kidney disease, they are investigational and unproven at this time in discriminating between renal osteodystrophy and osteoporosis.6,25–27 As we increase our understanding of the relationships between turnover, mineralization, volume, and bone strength, these noninvasive imaging technologies may become the means to correlate turnover, mineralization, and volume to bone strength and open up an entirely new way to classify skeletal strength.
In the meantime, the clinician is left with quantitative bone histomorphometry (which requires biopsy) and biochemical markers of bone turnover to characterize the bone disease that may be responsible for low-trauma fractures in stage 5 chronic kidney disease. The clinician should first use biochemical markers before bone biopsy to distinguish the form of renal osteodystrophy, as this distinction may be able to prevent unnecessary biopsy.
Biochemical markers of bone metabolism
In chronic kidney disease, the bone biochemical tests that nephrologists usually assess during the course of declining renal function are the serum levels of:
- Phosphorus
- Parathyroid hormone
- Calcium
- Other electrolytes
- Total alkaline phosphatase or bone-specific alkaline phosphatase
- 1,25 dihydroxyvitamin D.
In postmenopausal osteoporosis, the biochemical markers of bone turnover that are measured to reflect baseline levels of bone turnover or change in bone turnover in response to drug therapy are:
- The serum or urine collagen cross-links N-telopeptide (NTx) and C-telopeptide (CTx), markers of bone resorption
- Bone-specific alkaline phosphatase (an osteoblast activity marker)
- Serum osteocalcin, a bone formation marker
- Propeptide type 1 collagen (P1NP), a marker of osteoblast activity, highly correlated with bone formation
- 25-hydroxyvitamin D levels.
Biochemical markers of bone turnover cannot be used to diagnose osteoporosis. They can, however, provide clinical guidance as to whether a patient has high or low bone turnover and whether therapy is affecting bone turnover.28–36 Although these markers have value in making these distinctions in groups of patients, they are less sensitive and specific for classifying an individual patient’s bone turnover status.
Bone-specific alkaline phosphatase, parathyroid hormone, and adynamic bone disease
If a patient’s bone-specific alkaline phosphatase level is elevated, adynamic bone disease is highly unlikely. Assuming that other causes of this elevated level (eg, Paget disease of bone, metastatic cancer) have already been excluded, the elevated level could represent either osteomalacia or hyperparathyroid bone disease.
However, a “normal” bone-specific alkaline phosphatase level does not exclude adynamic bone disease, whereas a low level is more often associated with low bone turnover.
An elevated parathyroid hormone level does not exclude adynamic renal bone disease, but a low level (< 150 pg/mL) suggests a lowbone-turnover state. A level six times or more greater than the upper limit of normal is far more likely to be associated with high bone turnover.
Thus, in clinical practice, patients with stage 4 or 5 chronic kidney disease who have elevated bone-specific alkaline phosphatase or very high parathyroid hormone values do not have adynamic bone disease. Furthermore, once other causes of these aberrant biochemical abnormalities have been defined, then “high-bone-turnover osteoporosis” may be a consideration. Certainly, in my opinion, if bone turnover markers suggest low bone turnover, bone biopsy is necessary before starting an antiresorptive agent.35
Quantitative bone histomorphometry
Double tetracycline-labeled quantitative histomorphometry is still the only accepted way to measure turnover, mineralization, and volume in clinical practice.43–45 A committee of the American Society for Bone and Mineral Research has developed histomorphometric criteria for distinguishing among the different types of metabolic bone diseases (osteomalacia, adynamic bone disease, hyperparathyroid bone disease).12 These criteria can be used to distinguish among the various metabolic bone diseases that accompany stage 5 chronic kidney disease, including adynamic bone disease.43,46–48
For patients in stage 5 who have had a fragility fracture, adynamic bone disease should be excluded before the off-label use of an osteoporosis drug that reduces bone turnover, such as a bisphosphonate, calcitonin, estrogen, a selective estrogen receptor modulator, or denosumab (anti-RANK ligand antibody). While there is no evidence, for example, that starting a bisphosphonate in a patient who already has adynamic bone disease is detrimental to either bone strength or systemic vascular calcification (which may be linked to low bone turnover),49 it seems unreasonable to do so until solid prospective data clarify the harm or benefit.50 Preliminary experimental and clinical data suggest that bisphosphonates may even reduce progression of extraosseous calcification and inhibit the development of atherosclerosis.50
Hence, quantitative bone histomorphometry can discriminate among the various forms of renal osteodystrophy. If a distinct form of renal osteodystrophy is not present in a patient with stage 4 or 5 chronic kidney disease who has had a fracture and who, on biopsy, has a low trabecular bone volume, the patient probably has osteoporosis by exclusion.
TREATING OSTEOPOROSIS IN STAGE 1–3 CHRONIC KIDNEY DISEASE
As previously mentioned, patients who have fragility fractures in stage 1, 2, or 3 chronic kidney disease are more likely to have osteoporosis than renal osteodystrophy as the cause of their impaired bone strength. Although several articles have described a higher risk of fragility fractures in patients with age-related reduction in renal function than in agematched patients with normal renal function, the specific metabolic bone disease other than osteoporosis accounting for this bone fragility has not been defined.6
Hence, patients with osteoporosis who are in stage 1, 2, or 3 chronic kidney disease and do not have a known biochemical abnormality that might suggest some form of renal osteodystrophy can and should be considered for treatment with approved drugs that reduce the risk of fractures in postmenopausal, male, or glucocorticoid-induced osteoporosis.51–53 In clinical trials, these agents were shown to be effective in patients with serum creatinine concentrations as high as 2.0 mg/dL or a GFR as low as 30 mL/min, as estimated by the Cockcroft-Gault equation.
While all of the approved agents show evidence of reducing the risk of vertebral fractures, patients at higher risk of fractures or those who have already suffered a nonvertebral fracture are more often considered candidates for treatment with a bisphosphonate or teriparatide (Forteo), both of which have shown evidence of reducing the risk of all fractures.
Bisphosphonates in stage 1–3 chronic kidney disease
There is prospective evidence that patients with an age-related reduction in GFR down to 30 mL/min benefit from oral and intravenous bisphosphonates, since all of the clinical trials that led to the approval of bisphosphonates included patients with GFRs as low as this.54–57 Bisphosphonates seem to have an excellent safety profile as measured by renal adverse events in patients with a GFR of 30 mL/min or greater.52–59
From the intravenous bisphosphonate studies, it appears that ibandronate (Boniva) at the approved dose of 3 mg intravenously every 3 months and zoledronic acid (Reclast) 5 mg once a year given over 15 minutes are safe in patients with a GFR greater than 30 or 35 mL/min.
However, the safety of these drugs might not be the same in patients with preexisting renal parenchymal disease (eg, in diabetes) or in patients using other agents that could affect renal function (eg, nonsteroidal antiinflammatory drugs). Therefore, caution is still needed when deciding to use intravenous bisphosphonates in specific higher-risk renal subpopulations.
In the clinical trials of zoledronic acid, a substantial proportion of patients had diabetes, and no difference was seen in adverse renal effects between diabetic and nondiabetic patients. Also, GFRs declined equally between the treated and placebo groups over time and were no different at the end of 3 years.55 However, in patients in whom serum creatinine was measured 9 to 11 days after the infusion of zoledronic acid, there was a small but statistically significant transient increase in serum creatinine concentration (0.5–2.0 mg/dL above baseline) after the second annual infusion. 58 The serum creatinine concentrations returned to their baseline values in all of these patients before the next annual infusion.
It is important that infusions of zoledronic acid be given no faster than over 15 minutes. More rapid infusion has been associated with acute renal failure, suggesting that the tubular damage that mimics acute tubular necrosis is related to the maximal concentration and not to the area under the curve. I infuse zoledronic acid over 30 minutes in patients with normal renal function or in those with stage 1, 2, or 3 chronic kidney disease.
Teriparatide
Teriparatide’s approval trial did not require baseline measurements of GFR, but patients were enrolled only if their baseline serum creatinine concentrations were less than 2.0 mg/dL.60 In a post hoc analysis, a small subset of patients had GFRs as low as 30 mL/min as estimated by the Cockcroft-Gault equation. In these patients, teriparatide 20 or 40 μg/day had an anabolic effect as measured by increases in osteoblast activity markers and bone mineral density, similar to that seen in patients with higher estimated GFRs and without any adverse renal effects.61
There are no data on using teriparatide in stage 4 or 5 chronic kidney disease, and I emphasize that in all of the clinical trials of teriparatide, all patients, even those with estimated GFRs as low as 30 mL/min, had normal baseline serum intact parathyroid hormone levels. It is possible that the bone biologic response could differ between patients with chronic kidney disease who have an elevated as compared with a normal serum parathyroid hormone level. This issue should be investigated.
TREATING OSTEOPOROSIS IN STAGE 4 OR 5 CHRONIC KIDNEY DISEASE
Treatment decisions are more difficult in patients with stage 4 and especially stage 5 chronic kidney disease who have had fragility fractures. This is even the case when the clinician has determined to the best of his or her ability that the patient has osteoporosis rather than renal osteodystrophy.
There are no prospective data showing any of the approved drugs to be effective in treating osteoporosis in patients whose GFRs are lower than 30 mL/min. However, a post hoc analysis of pooled data from nine clinical trials62 found that risedronate (Actonel) 5 mg/day reduced the incidence of new vertebral fractures. Another post hoc analysis, from the Fracture Intervention Trial,63 found that alendronate (Fosamax) 5 mg/day for the first 2 years and 10 mg/day for the 3rd year reduced the incidence of all clinical fractures. In neither of these post hoc analyses did the drug affect the serum creatinine concentration. The patients—postmenopausal women—had GFRs as low as 15 mL/min as estimated by the Cockcroft-Gault equation. Similar post hoc data have been published on raloxifene (Evista).64
There are no data on the efficacy (reduction in fracture risk) or safety of any bisphosphonate in patients with GFRs lower than 15 mL/min (stage 5 chronic kidney disease). Nevertheless, the question often arises when fragility fractures occur in this population. Here, only opinion and controversy exist, and we fervently await good science and randomized prospective data.
How to manage renal bone disease after transplantation is a distinctly separate issue in which bisphosphonate use may be even more controversial than in end-stage renal disease.65,66
In my opinion, patients without fractures with stage 5 chronic kidney disease should not be given bisphosphonates or teriparatide offlabel. Treating only on the basis of low bone mineral density and other risk factors seems to be associated with greater risk than benefit.
In stage 5 patients suffering fragility fractures, a bisphosphonate may be considered, but only after renal osteodystrophy has been thoroughly ruled out, which most often requires a bone biopsy.43,67,68 In skilled hands, transiliac bone biopsy is a safe procedure with little morbidity.
If osteoporosis appears to be the cause of the fracture, and if one chooses to use a bisphosphonate and the patient gives his or her informed consent, then I would give half the usual dose and restrict the therapy to no more than 3 years. The reason for halving the dose is based on the known pharmacokinetics of bisphosphonates in people with normal renal function: 50% of a given dose goes to bone and 50% is excreted by the kidney. Furthermore, the dialyzability of bisphosphonates has not been well studied. Limiting the treatment to 3 years is based on the unknown but probably greater bone retention of bisphosphonates when excretion is impaired.
I must emphasize that these approaches are not based on any evidence of efficacy, but rather are considered in extreme cases of often-recurrent fragility fractures in which the fractures per se pose a great risk of morbidity and death. These approaches should be clearly discussed with the patient, undertaken by specialists knowledgeable in complex metabolic bone disease management, and initiated only after the skeletal fragility disorder is well characterized.
SUMMING UP
No consensus exists on the criteria for diagnosing osteoporosis in stage 4 or 5 chronic kidney disease.
In higher-risk patients in stage 1, 2, or 3 chronic kidney disease who have osteoporosis, it appears that any drug approved for osteoporosis can be used, eg, a bisphosphonate, teriparatide, or both.
Considerations for management are far more complex in stage 4 or 5 because of the increased prevalence of other metabolic bone diseases and renal osteodystrophy, and because the World Health Organization criteria cannot be used to diagnose osteoporosis. In stage 5, the differential diagnosis requires careful analysis of a broad range of biochemical markers of bone turnover and, at times, quantitative bone histomorphometry, especially if one is considering using a bisphosphonate. It is unknown if bisphosphonates, by reducing bone turnover in a preexisting low-bone-turnover state, would help or harm bone or would lead to less or more cardiovascular disease. These questions must be addressed by better science and prospective data.
In the future, newer noninvasive radiologic tools to measure microstructure and mineralization of bone promise to help us better understand osteoporosis and renal osteodystrophy in a noninvasive manner.
In clinical practice, at the current time and with current limited knowledge, treatment of osteoporosis in stage 4 or 5 chronic kidney disease is opinion-based. Nevertheless, in very specific clinical cases of severe fragility fractures that, by themselves, may cause disability and death, bisphosphonates should be considered by experts in bone metabolism and, as with any off-label application, after careful informed discussions with the patient.
But even in chronic kidney disease, many fractures are due to postmenopausal or agerelated osteoporosis, and estrogen-deficiency osteoporosis is the most common cause of fragility fractures overall.1–3 Osteoporosis can be diagnosed only after other causes of skeletal fragility have been ruled out. And how to diagnose and treat osteoporosis in the most severe stage of kidney disease is a matter of opinion, as we have almost no data to guide us.
Nevertheless, in the pages that follow, I will outline my admittedly opinion-based approach to diagnosing and managing the causes of fragility fractures in patients with chronic kidney disease.
T SCORES DO NOT DISTINGUISH THE CAUSES OF FRAGILITY
The most common cause of fragility fractures is osteoporosis due to estrogen deficiency.1–3 But since many other medical conditions can lead to osteoporosis, simple diagnostic criteria have been difficult to find.
Before 1994, the diagnosis of osteoporosis was made on the basis of low-trauma fractures.4 Now, we use the World Health Organization criteria,5 based on bone mineral density T scores:
- Normal—a T score of −1.0 standard deviations or higher
- Osteopenia—a T score of less than −1.0 but higher than −2.5
- Osteoporosis—a T score of −2.5 or less
- Severe osteoporosis—a T score of −2.5 or less with a fragility fracture.
However, fractures can also be due to metabolic bone diseases that are not osteoporosis, including renal bone diseases.6–7 While a low T score or a fracture provides a working diagnosis of osteoporosis, it does not help distinguish the different types of osteoporosis and nonosteoporotic metabolic bone diseases. For example, osteomalacia and osteogenesis imperfecta can also cause fragility fractures and can be associated with low bone density. Using these criteria to define osteoporosis is even more problematic in patients with chronic kidney disease.
FIVE STAGES OF CHRONIC KIDNEY DISEASE
The National Kidney Foundation8 classifies the severity of chronic kidney disease on the basis of the glomerular filtration rate (GFR), as measured by 24-hour urine for creatinine clearance, or as estimated by the Cockcroft-Gault equation or, preferably, the Modification of Diet in Renal Disease (MDRD) equation (calculators are available at www.kidney.org/professionals/kdoqi/gfr_calculator.cfm):
- Stage 1—GFR 90 mL/minute/1.73 m2 or higher
- Stage 2—GFR 60 to 89
- Stage 3—GFR 30 to 59
- Stage 4—GFR 15 to 29
- Stage 5—GFR lower than 15, or if the patient is on dialysis. (Another stage, called 5D, was added to the list to denote patients on dialysis, since the metabolic derangements in bone and systemic biology may differ between patients on dialysis vs those not on dialysis.)
This staging system is relevant to the discussion of bone fragility that follows.
CHRONIC KIDNEY DISEASE IS COMMON IN THE ELDERLY
The third National Health and Nutrition Examination Survey9 found that, at least as estimated by the Cockcroft-Gault equation, the GFR declines with age, so that by the age of 70 at least 20% of the US population has stage 4 or 5 chronic kidney disease.
Although the Cockcroft-Gault and MDRD equations may yield lower GFR values in the general population than one would get by measuring creatinine, inulin, or iothalamate clearance,10,11 the point is that both osteoporosis and chronic kidney disease are common.
THE GAMUT OF RENAL OSTEODYSTROPHY
In kidney failure (stage 5 chronic kidney disease), all forms of renal osteodystrophy may be associated with fragility fractures. Renal osteodystrophy can be defined by quantitative bone histomorphometry.12,13 The systemic conditions that may be associated with the bone disease and systemic vascular disease (chronic kidney disease–mineral and bone disorder) are characterized by one or more of the following14:
- Abnormalities of calcium, phosphorus, parathyroid hormone, or vitamin D metabolism
- Abnormalities in bone turnover, mineralization, volume, linear growth, or strength
- Vascular or other soft-tissue calcification.
The National Kidney Foundation14 classifies renal osteodystrophy on the basis of:
- Turnover—high, normal, or low
- Mineralization—normal or abnormal
- Volume—high, normal, or low.
Although this system helps us understand these diseases better, it does not provide a working diagnosis of osteoporosis.14
WHAT IS OSTEOPOROSIS?
In an attempt to define osteoporosis by a pathophysiologic mechanism, the National Institutes of Health15 have held two consensus conferences and have stated that “osteoporosis is a skeletal disorder characterized by impairment in bone strength predisposing a person to an increased risk of fracture. Bone strength primarily reflects the integration of bone density and bone quality.”15 However, the consensus statement also does not provide a working diagnosis of osteoporosis—one that clinicians can apply to management decisions, and one that is also accepted by the US International Classification of Disease codes for reimbursement purposes.
The 1994 World Health Organization criteria offer the most pragmatic operational definition of osteoporosis, and they can be applied in both men and women, as well as in younger patients with medical conditions associated with increased risk of low-trauma fracture.5,16 Although the main purpose of these criteria was to advise international health authorities of the potential future economic impact of osteoporosis, the T score also became the pragmatic diagnostic threshold for defining normal, osteopenia, and osteoporosis in clinical practice.
The T score also calls attention to an important observation: of people who have fractures and subsequently undergo bone densitometry, more are found to have osteopenia than osteoporosis. The reasons are that there are more people with osteopenia than osteoporosis,17,18 and many other factors independent of low bone mineral density contribute to bone strength.19,20
How is osteoporosis diagnosed in stage 1–3 chronic kidney disease?
In patients with chronic kidney disease who develop fragility fractures, the reasonable question is: Is the cause of the fracture osteoporosis or some other metabolic bone disease associated with chronic kidney disease?
The National Kidney Foundation guidelines14 say that the diagnosis of osteoporosis can be established in patients with stage 1, 2, or 3 chronic kidney disease on the basis of either of the World Health Organization criteria, ie, a T score of −2.5 or lower or fragility fractures, as in the postmenopausal population, as long as there are no biochemical abnormalities that suggest chronic kidney disease–mineral and bone disorder.
How is osteoporosis diagnosed in stage 4 or 5 chronic kidney disease?
The answer is neither straightforward nor clearly defined in severe (stage 4 or 5) chronic kidney disease.
In stage 5 and especially in patients on dialysis, the derangements in bone and mineral metabolism become serious enough to impair bone strength and increase the risk of lowtrauma fractures. The risk of hip fracture in stage 5 may be four times higher than in agematched controls.21–24
Adynamic, severe hyperparathyroid bone disease as well as osteomalacia can be associated with a higher risk of fragility fractures than in aged-matched controls in population studies of postmenopausal women or elderly men. These are bone fragility conditions that are not osteoporosis but that can mimic osteoporosis by the World Health Organization criteria.
Thus, when a patient in stage 5 has severe fragility fractures that by themselves may be life-threatening, it is reasonable to ask if the drugs that reduce the risk of fractures in many other osteoporotic conditions (postmenopausal, steroid-induced, elderly male osteoporosis, after solid organ transplantation) can also be used in patients with advanced chronic kidney disease.
The diagnosis of osteoporosis in these patients has no universally accepted criteria. The diagnosis is best suggested by excluding other forms of renal osteodystrophy by quantitative histomorphometry or by attempting to classify the form of renal osteodystrophy by noninvasive means of assessing bone turnover, mineralization, and volume. However, we lack clinical tools to make these distinctions in individual patients.
While many promising radiologic techniques that examine bone microarchitecture offer hope of being able to define turnover, mineralization, and volume noninvasively in severe chronic kidney disease, they are investigational and unproven at this time in discriminating between renal osteodystrophy and osteoporosis.6,25–27 As we increase our understanding of the relationships between turnover, mineralization, volume, and bone strength, these noninvasive imaging technologies may become the means to correlate turnover, mineralization, and volume to bone strength and open up an entirely new way to classify skeletal strength.
In the meantime, the clinician is left with quantitative bone histomorphometry (which requires biopsy) and biochemical markers of bone turnover to characterize the bone disease that may be responsible for low-trauma fractures in stage 5 chronic kidney disease. The clinician should first use biochemical markers before bone biopsy to distinguish the form of renal osteodystrophy, as this distinction may be able to prevent unnecessary biopsy.
Biochemical markers of bone metabolism
In chronic kidney disease, the bone biochemical tests that nephrologists usually assess during the course of declining renal function are the serum levels of:
- Phosphorus
- Parathyroid hormone
- Calcium
- Other electrolytes
- Total alkaline phosphatase or bone-specific alkaline phosphatase
- 1,25 dihydroxyvitamin D.
In postmenopausal osteoporosis, the biochemical markers of bone turnover that are measured to reflect baseline levels of bone turnover or change in bone turnover in response to drug therapy are:
- The serum or urine collagen cross-links N-telopeptide (NTx) and C-telopeptide (CTx), markers of bone resorption
- Bone-specific alkaline phosphatase (an osteoblast activity marker)
- Serum osteocalcin, a bone formation marker
- Propeptide type 1 collagen (P1NP), a marker of osteoblast activity, highly correlated with bone formation
- 25-hydroxyvitamin D levels.
Biochemical markers of bone turnover cannot be used to diagnose osteoporosis. They can, however, provide clinical guidance as to whether a patient has high or low bone turnover and whether therapy is affecting bone turnover.28–36 Although these markers have value in making these distinctions in groups of patients, they are less sensitive and specific for classifying an individual patient’s bone turnover status.
Bone-specific alkaline phosphatase, parathyroid hormone, and adynamic bone disease
If a patient’s bone-specific alkaline phosphatase level is elevated, adynamic bone disease is highly unlikely. Assuming that other causes of this elevated level (eg, Paget disease of bone, metastatic cancer) have already been excluded, the elevated level could represent either osteomalacia or hyperparathyroid bone disease.
However, a “normal” bone-specific alkaline phosphatase level does not exclude adynamic bone disease, whereas a low level is more often associated with low bone turnover.
An elevated parathyroid hormone level does not exclude adynamic renal bone disease, but a low level (< 150 pg/mL) suggests a lowbone-turnover state. A level six times or more greater than the upper limit of normal is far more likely to be associated with high bone turnover.
Thus, in clinical practice, patients with stage 4 or 5 chronic kidney disease who have elevated bone-specific alkaline phosphatase or very high parathyroid hormone values do not have adynamic bone disease. Furthermore, once other causes of these aberrant biochemical abnormalities have been defined, then “high-bone-turnover osteoporosis” may be a consideration. Certainly, in my opinion, if bone turnover markers suggest low bone turnover, bone biopsy is necessary before starting an antiresorptive agent.35
Quantitative bone histomorphometry
Double tetracycline-labeled quantitative histomorphometry is still the only accepted way to measure turnover, mineralization, and volume in clinical practice.43–45 A committee of the American Society for Bone and Mineral Research has developed histomorphometric criteria for distinguishing among the different types of metabolic bone diseases (osteomalacia, adynamic bone disease, hyperparathyroid bone disease).12 These criteria can be used to distinguish among the various metabolic bone diseases that accompany stage 5 chronic kidney disease, including adynamic bone disease.43,46–48
For patients in stage 5 who have had a fragility fracture, adynamic bone disease should be excluded before the off-label use of an osteoporosis drug that reduces bone turnover, such as a bisphosphonate, calcitonin, estrogen, a selective estrogen receptor modulator, or denosumab (anti-RANK ligand antibody). While there is no evidence, for example, that starting a bisphosphonate in a patient who already has adynamic bone disease is detrimental to either bone strength or systemic vascular calcification (which may be linked to low bone turnover),49 it seems unreasonable to do so until solid prospective data clarify the harm or benefit.50 Preliminary experimental and clinical data suggest that bisphosphonates may even reduce progression of extraosseous calcification and inhibit the development of atherosclerosis.50
Hence, quantitative bone histomorphometry can discriminate among the various forms of renal osteodystrophy. If a distinct form of renal osteodystrophy is not present in a patient with stage 4 or 5 chronic kidney disease who has had a fracture and who, on biopsy, has a low trabecular bone volume, the patient probably has osteoporosis by exclusion.
TREATING OSTEOPOROSIS IN STAGE 1–3 CHRONIC KIDNEY DISEASE
As previously mentioned, patients who have fragility fractures in stage 1, 2, or 3 chronic kidney disease are more likely to have osteoporosis than renal osteodystrophy as the cause of their impaired bone strength. Although several articles have described a higher risk of fragility fractures in patients with age-related reduction in renal function than in agematched patients with normal renal function, the specific metabolic bone disease other than osteoporosis accounting for this bone fragility has not been defined.6
Hence, patients with osteoporosis who are in stage 1, 2, or 3 chronic kidney disease and do not have a known biochemical abnormality that might suggest some form of renal osteodystrophy can and should be considered for treatment with approved drugs that reduce the risk of fractures in postmenopausal, male, or glucocorticoid-induced osteoporosis.51–53 In clinical trials, these agents were shown to be effective in patients with serum creatinine concentrations as high as 2.0 mg/dL or a GFR as low as 30 mL/min, as estimated by the Cockcroft-Gault equation.
While all of the approved agents show evidence of reducing the risk of vertebral fractures, patients at higher risk of fractures or those who have already suffered a nonvertebral fracture are more often considered candidates for treatment with a bisphosphonate or teriparatide (Forteo), both of which have shown evidence of reducing the risk of all fractures.
Bisphosphonates in stage 1–3 chronic kidney disease
There is prospective evidence that patients with an age-related reduction in GFR down to 30 mL/min benefit from oral and intravenous bisphosphonates, since all of the clinical trials that led to the approval of bisphosphonates included patients with GFRs as low as this.54–57 Bisphosphonates seem to have an excellent safety profile as measured by renal adverse events in patients with a GFR of 30 mL/min or greater.52–59
From the intravenous bisphosphonate studies, it appears that ibandronate (Boniva) at the approved dose of 3 mg intravenously every 3 months and zoledronic acid (Reclast) 5 mg once a year given over 15 minutes are safe in patients with a GFR greater than 30 or 35 mL/min.
However, the safety of these drugs might not be the same in patients with preexisting renal parenchymal disease (eg, in diabetes) or in patients using other agents that could affect renal function (eg, nonsteroidal antiinflammatory drugs). Therefore, caution is still needed when deciding to use intravenous bisphosphonates in specific higher-risk renal subpopulations.
In the clinical trials of zoledronic acid, a substantial proportion of patients had diabetes, and no difference was seen in adverse renal effects between diabetic and nondiabetic patients. Also, GFRs declined equally between the treated and placebo groups over time and were no different at the end of 3 years.55 However, in patients in whom serum creatinine was measured 9 to 11 days after the infusion of zoledronic acid, there was a small but statistically significant transient increase in serum creatinine concentration (0.5–2.0 mg/dL above baseline) after the second annual infusion. 58 The serum creatinine concentrations returned to their baseline values in all of these patients before the next annual infusion.
It is important that infusions of zoledronic acid be given no faster than over 15 minutes. More rapid infusion has been associated with acute renal failure, suggesting that the tubular damage that mimics acute tubular necrosis is related to the maximal concentration and not to the area under the curve. I infuse zoledronic acid over 30 minutes in patients with normal renal function or in those with stage 1, 2, or 3 chronic kidney disease.
Teriparatide
Teriparatide’s approval trial did not require baseline measurements of GFR, but patients were enrolled only if their baseline serum creatinine concentrations were less than 2.0 mg/dL.60 In a post hoc analysis, a small subset of patients had GFRs as low as 30 mL/min as estimated by the Cockcroft-Gault equation. In these patients, teriparatide 20 or 40 μg/day had an anabolic effect as measured by increases in osteoblast activity markers and bone mineral density, similar to that seen in patients with higher estimated GFRs and without any adverse renal effects.61
There are no data on using teriparatide in stage 4 or 5 chronic kidney disease, and I emphasize that in all of the clinical trials of teriparatide, all patients, even those with estimated GFRs as low as 30 mL/min, had normal baseline serum intact parathyroid hormone levels. It is possible that the bone biologic response could differ between patients with chronic kidney disease who have an elevated as compared with a normal serum parathyroid hormone level. This issue should be investigated.
TREATING OSTEOPOROSIS IN STAGE 4 OR 5 CHRONIC KIDNEY DISEASE
Treatment decisions are more difficult in patients with stage 4 and especially stage 5 chronic kidney disease who have had fragility fractures. This is even the case when the clinician has determined to the best of his or her ability that the patient has osteoporosis rather than renal osteodystrophy.
There are no prospective data showing any of the approved drugs to be effective in treating osteoporosis in patients whose GFRs are lower than 30 mL/min. However, a post hoc analysis of pooled data from nine clinical trials62 found that risedronate (Actonel) 5 mg/day reduced the incidence of new vertebral fractures. Another post hoc analysis, from the Fracture Intervention Trial,63 found that alendronate (Fosamax) 5 mg/day for the first 2 years and 10 mg/day for the 3rd year reduced the incidence of all clinical fractures. In neither of these post hoc analyses did the drug affect the serum creatinine concentration. The patients—postmenopausal women—had GFRs as low as 15 mL/min as estimated by the Cockcroft-Gault equation. Similar post hoc data have been published on raloxifene (Evista).64
There are no data on the efficacy (reduction in fracture risk) or safety of any bisphosphonate in patients with GFRs lower than 15 mL/min (stage 5 chronic kidney disease). Nevertheless, the question often arises when fragility fractures occur in this population. Here, only opinion and controversy exist, and we fervently await good science and randomized prospective data.
How to manage renal bone disease after transplantation is a distinctly separate issue in which bisphosphonate use may be even more controversial than in end-stage renal disease.65,66
In my opinion, patients without fractures with stage 5 chronic kidney disease should not be given bisphosphonates or teriparatide offlabel. Treating only on the basis of low bone mineral density and other risk factors seems to be associated with greater risk than benefit.
In stage 5 patients suffering fragility fractures, a bisphosphonate may be considered, but only after renal osteodystrophy has been thoroughly ruled out, which most often requires a bone biopsy.43,67,68 In skilled hands, transiliac bone biopsy is a safe procedure with little morbidity.
If osteoporosis appears to be the cause of the fracture, and if one chooses to use a bisphosphonate and the patient gives his or her informed consent, then I would give half the usual dose and restrict the therapy to no more than 3 years. The reason for halving the dose is based on the known pharmacokinetics of bisphosphonates in people with normal renal function: 50% of a given dose goes to bone and 50% is excreted by the kidney. Furthermore, the dialyzability of bisphosphonates has not been well studied. Limiting the treatment to 3 years is based on the unknown but probably greater bone retention of bisphosphonates when excretion is impaired.
I must emphasize that these approaches are not based on any evidence of efficacy, but rather are considered in extreme cases of often-recurrent fragility fractures in which the fractures per se pose a great risk of morbidity and death. These approaches should be clearly discussed with the patient, undertaken by specialists knowledgeable in complex metabolic bone disease management, and initiated only after the skeletal fragility disorder is well characterized.
SUMMING UP
No consensus exists on the criteria for diagnosing osteoporosis in stage 4 or 5 chronic kidney disease.
In higher-risk patients in stage 1, 2, or 3 chronic kidney disease who have osteoporosis, it appears that any drug approved for osteoporosis can be used, eg, a bisphosphonate, teriparatide, or both.
Considerations for management are far more complex in stage 4 or 5 because of the increased prevalence of other metabolic bone diseases and renal osteodystrophy, and because the World Health Organization criteria cannot be used to diagnose osteoporosis. In stage 5, the differential diagnosis requires careful analysis of a broad range of biochemical markers of bone turnover and, at times, quantitative bone histomorphometry, especially if one is considering using a bisphosphonate. It is unknown if bisphosphonates, by reducing bone turnover in a preexisting low-bone-turnover state, would help or harm bone or would lead to less or more cardiovascular disease. These questions must be addressed by better science and prospective data.
In the future, newer noninvasive radiologic tools to measure microstructure and mineralization of bone promise to help us better understand osteoporosis and renal osteodystrophy in a noninvasive manner.
In clinical practice, at the current time and with current limited knowledge, treatment of osteoporosis in stage 4 or 5 chronic kidney disease is opinion-based. Nevertheless, in very specific clinical cases of severe fragility fractures that, by themselves, may cause disability and death, bisphosphonates should be considered by experts in bone metabolism and, as with any off-label application, after careful informed discussions with the patient.
- Melton LJ. Epidemiology worldwide. Endocrinol Metab Clin North Am 2003; 32:1–13.
- Burge R, Dawson-Hughes B, Solomon DH, Wong JB, King A, Tosteson A. Incidence and economic burden of osteoporosis-related fractures in the United States, 2005–2025. J Bone Miner Res 2007; 22:465–475.
- Barrett-Connor E, Siris ES, Wehren LE, et al. Osteoporosis and fracture risk in women of different ethnic groups. J Bone Miner Res 2005; 20:185–194.
- Assessment of fracture risk and its application to screening for postmenopausal osteoporosis. Report of a WHO Study Group. World Health Organ Tech Rep Ser 1994; 843:1–129.
- Miller PD, Bonnick SL. Clinical application of bone densitometry. In:Favus MJ, editor. Primer on the Metabolic Bone Diseases and Disorders of Mineral Metabolism. 4th ed. American Society for Bone and Mineral Research. Philadelphia, PA: Lippincott Williams & Wilkins; 1999.
- Nickolas TL, Leonard MB, Shane E. Chronic kidney disease and bone fracture: a growing concern. Kidney Int 2008; 74:721–731.
- Gal-Moscovici A, Sprague SM. Osteoporosis and chronic kidney disease. Semin Dial 2007; 20:423–430.
- National Kidney Foundation. Clinical practice guidelines for bone metabolism and disease in chronic kidney disease. Am J Kidney Dis 2003; 42(suppl 3):S1–S201.
- Coresh J, Astor BC, Greene T, Eknoyan G, Levey AS. Prevalence of chronic kidney disease and decreased kidney function in the adult US population: Third National Health and Nutrition Examination Survey. Am J Kidney Dis 2003; 41:1–12.
- Bennett WM. Reporting eGFR. Clin J Am Soc Nephrol 2008; 3:1561–1562.
- Glassock RJ, Winearls C. Screening for CKD with eGFR: doubts and dangers. Clin J Am Soc Nephrol 2008; 3:1563–1568.
- Parfitt AM, Drezner M, Glorieux F, et al. Bone histomorphometry: standardization of nomenclature, symbols, and units. Report of the ASBMR Histomorphometry Nomenclature Committee. J Bone Miner Res 1987; 2:595–610.
- Andress DL, Sherrard DJ. The osteodystrophy of chronic renal failure. In:Schrier RW, editor: Diseases of the Kidney and Urinary Tract. Philadelphia, PA: Lippincott Williams & Wilkins; 2007:2431–2453.
- Moe S, Drueke T, Cunningham J, et al. Definition, evaluation, and classification of renal osteodystrophy: a position statement from Kidney Disease: Improving Global Outcomes (KDIGO). Kidney Int 2006; 69:1945–1953.
- NIH Consensus Development Panel Osteoporosis Prevention Diagnosis and Therapy. Osteoporosis prevention, diagnosis, and therapy. JAMA 2001; 285:785–795.
- Baim S, Binkley N, Bilezikian JP, et al. Official positions of the International Society for Clinical Densitometry and executive summary of the 2007 ISCD Position Development Conference. J Clin Densitom 2008; 11:75–91.
- Siris E, Miller P, Barrett-Connor E, et al. Identification and fracture outcomes of undiagnosed low bone mineral density in postmenopausal women: results from the National Osteoporosis Risk Assessment (NORA). JAMA 2001; 286:2815–2822.
- Schuit SCE, Oei HH, Witteman JC, et al. Fracture incidence and association with bone mineral density in elderly men and women: the Rotterdam Study. Bone 2004; 34:195–202.
- Bouxsein ML. Non-invasive measurements of bone strength: promise and peril. J Musculoskelet Neuronal Interact 2004; 4:404–405.
- Seeman E. Bone quality: the material and structural basis of bone strength. J Bone Miner Metab 2008; 26:1–8.
- Alem AM, Sherrard DJ, Gillen DL, et al. Increased risk of hip fracture among patients with end-stage renal disease. Kidney Int 2000; 58:396–399.
- Ball AM, Gillen DL, Sherrard D, et al. Risk of hip fracture among dialysis and renal transplant recipients. JAMA 2002; 288:3014–3018.
- Jadoul M, Albert JM, Akiba T, et al. Incidence and risk factors for hip or other bone fractures among hemodialysis patients in the Dialysis Outcomes and Practice Patterns Study. Kidney Int 2006; 70:1358–1366.
- Stehman-Breen CO, Sherrard DJ, Alem AM, et al. Risk factors for hip fracture among patients with end-stage renal disease. Kidney Int 2000; 58:2200–2205.
- Wehrli FW, Leonard MB, Saha PK, Gomberg BR. Quantitative highresolution magnetic resonance imaging reveals structural implications of renal osteodystrophy on trabecular and cortical bone. J Magn Reson Imaging 2004; 20:83–89.
- Genant HK, Lang TF, Engelke K, et al. Advances in the noninvasive assessment of bone density, quality, and structure. Calcif Tissue Int 1996; 59(suppl 1):S10–S15.
- Roschger P, Paschalis EP, Fratzl P, Klaushofer K. Bone mineralization density distribution in health and disease. Bone 2008; 42:456–466.
- Miller PD. Bone density and markers of bone turnover in predicting fracture risk and how changes in these measures predict fracture risk reduction. Curr Osteoporos Rep 2005; 3:103–110.
- Miller PD, Hochberg MC, Wehren LE, Ross PD, Wasnich RD. How useful are measures of BMD and bone turnover? Curr Med Res Opin 2005; 21:545–554.
- Chavassieux PM, Delmas PD. Bone remodeling: biochemical markers or bone biopsy? J Bone Miner Res 2006; 21:178–179.
- Garnero P. Biomarkers for osteoporosis management: utility in diagnosis, fracture risk prediction and therapy monitoring. Mol Diagn Ther 2008; 12:157–170.
- Hochberg M, Greenspan S, Wasnich R, Miller P, Thompson D, Ross P. Changes in bone density and turnover explain the reductions in incidence of nonvertebral fractures that occur during treatment with antiresorptive agents. J Clin Endocrinol Metab 2002; 87:1586–1592.
- Bouxsein ML, Delmas PD. Considerations for development of surrogate endpoints for antifracture efficacy of new treatments in osteoporosis: a perspective. J Bone Miner Res 2008; 23:1155–1167.
- Chen P, Satterwhite JH, Licatta AA, et al. Early changes in biochemical markers of bone formation predict BMD response to teriparatide in postmenopausal women with osteoporosis. J Bone Miner Res 2005; 20:962–970.
- Miller PD, Delmas PD, Lindsay R, et al. Early responsiveness of women with osteoporosis to teriparatide after therapy with alendronate or risedronate. J Clin Endocrinol Metab 2008; 93:3785–3793.
- Baim S, Miller PD. Assessing the clinical utility of serum CTX in postmenopausal osteoporosis and its use in predicting risk of osteonecrosis of the jaw. J Bone Miner Res 2009; 24:561–574.
- Miller PD, Lerma EV. Renal bone diseases. In:Kleerekoper M, Siris E, McClung M, editors. The Bone and Mineral Manual—A Practical Guide. 2nd ed. Burlington, MA: Elsevier Academic Press; 2005:127–138.
- Miller PD, Shane E. Management of transplantation renal bone disease: Interplay of bone mineral density and decisions regarding bisphosphonate use. In:Weir MR, editor. Medical Management of Kidney Transplantation. Philadelphia, PA: Lippincott Williams & Wilkins; 2004:359–375.
- Levin A, Bakris GL, Molitch M, et al. Prevalence of abnormal serum vitamin D, PTH, calcium, and phosphorus in patients with chronic kidney disease: results of the study to evaluate early kidney disease. Kidney int 2007; 71:31–38.
- Vassalotti JA, Uribarri J, Chen SC, et al. Trends in mineral metabolism: Kidney Early Evaluation Program (KEEP) and the National Health and Nutrition Examination Survey (NHANES) 1999–2004. Am J Kidney Dis 2008; 51(suppl 2):S56–S68.
- Meier C, Seibel MJ, Kraenzlin ME. Use of bone turnover markers in the real world: are we there yet? J Bone Miner Res 2009; 24:386–388.
- Lehmann G, Ott U, Kaemmerer D, Schuetze J, Wolf G. Bone histomorphometry and biochemical markers of bone turnover in patients with chronic kidney disease stages 3–5. Clin Nephrol 2008; 70:296–305.
- Miller PD. The role of bone biopsy in patients with chronic renal failure. Clin J Am Soc Nephrol 2008; 3(suppl 3):S140–S150.
- Frost HM. Tetracycline-based histological analysis of bone remodeling. Calcif Tissue Res 1969; 3:211–237.
- Hitt O, Jaworski ZF, Shimizu AG, Frost HM. Tissue-level bone formation rates in chronic renal failure, measured by means of tetracycline bone labeling. Can J Physiol Pharmacol 1970; 48:824–828.
- Coen G. Adynamic bone disease: an update and overview. J Nephrol 2005; 18:117–122.
- Parfitt AM. Renal bone disease: a new conceptual framework for the interpretation of bone histomorphometry. Curr Opin Nephrol Hypertens 2003; 12:387–403.
- Brandenburg VM, Floege J. Adynamic bone diseaseùbone and beyond. NDT Plus 2008; 3:135–147. doi:10.1093/ndtplus/sfn040.
- Hruska KA, Saab G, Mathew S, Lund R. Renal osteodystrophy, phosphate homeostasis, and vascular calcification. Semin Dial 2007; 20:309–315.
- Toussaint ND, Elder GJ, Kerr PG. Bisphosphonates in chronic kidney disease; balancing potential benefits and adverse effects on bone and soft tissue. Clin J Am Soc Nephrol 2009; 4:221–233.
- Miller PD. Is there a role for bisphosphonates in chronic kidney disease? Semin Dial 2007; 20:186–190.
- Miller PD. Bisphosphonates: pharmacology and use in the treatment of osteoporosis. In:Marcus R, Feldman D, Nelson DA, Rosen CJ, editors. Osteoporosis. 3rd ed. Boston, MA: Elsevier Academic Press; 2008:1725–1736.
- Russell RG, Watts NB, Ebetino FH, Rogers MJ. Mechanisms of action of bisphosphonates: similarities and differences and their potential influence on clinical efficacy. Osteoporosis Int 2008; 19:733–759.
- Eisman JA, Civitelli R, Adami S, et al. Efficacy and tolerability of intravenous ibandronate injections in postmenopausal osteoporosis: 2-year results from the DIVA study. J Rheumatol 2008; 35:488–497.
- Black DM, Delmas PD, Eastell RR, et al. Once-yearly zoledronic acid for treatment of postmenopausal osteoporosis. N Engl J Med 2007; 356:1809–1822.
- Lewiecki EM, Miller PD. Renal safety of intravenous bisphosphonates in the treatment of osteoporosis. Expert Opin Drug Saf 2007; 6:663–672.
- Miller PD. Anti-resorptives in the management of osteoporosis. Best Pract Res Clin Endocrinol Metab 2008; 22:849–868.
- Perazella MA, Markowitz GS. Bisphosphonate nephrotoxicity. Kidney Int 2008; 74:1385–1393.
- Boonen S, Sellmeyer DE, Lippuner K, et al. Renal safety of annual zoledronic acid infusions in osteoporotic postmenopausal women. Kidney Int 2008; 74:641–648.
- Neer RM, Arnaud CD, Zanchetta JR, et al. Effect of parathyroid hormone (1–34) on fractures and bone mineral density in postmenopausal women with osteoporosis. N Engl J Med 2001; 344:1434–1441.
- Miller PD, Schwartz EN, Chen P, Misurski DA, Krege JH. Teriparatide in postmenopausal women with osteoporosis and mild or moderate renal impairment. Osteoporosis Int 2007; 18:59–68.
- Miller PD, Roux C, Boonen S, Barton I, Dunlap L, Burgio D. Safety and efficacy of risedronate in patients with age-related reduced renal function as estimated by the Cockcroft and Gault method: a pooled analysis of nine clinical trials. J Bone Miner Res 2005; 20:2105–2115.
- Jamal SA, Bauer DC, Ensrud KE, et al. Alendronate treatment in women with normal to severely impaired renal function: an analysis of the fracture intervention trial. J Bone Miner Res 2007; 22:503–508.
- Ishani A, Blackwell T, Jamal SA, Cummings SR, Ensrud KE; MORE Investigators. The effect of raloxifene treatment in postmenopausal women with CKD. J Am Soc Nephrol 2008; 19:1430–1438.
- Coco M, Glicklich D, Faugere MC, et al. Prevention of bone loss in renal transplant recipients: a prospective, randomized trial of intravenous pamidronate. J Am Soc Nephrol 2003; 14:2669–2676.
- Palmer SC, McGregor DO, Strippoli GF. Interventions for preventing bone disease in kidney transplant recipients. Cochrane Database Syst Rev 2007;CD005015.
- Ferreira MA. Diagnosis of renal osteodystrophy: when and how to use biochemical markers and non-invasive methods; when bone biopsy is needed. Nephrol Dial Transplant 2000; 15(suppl 5):8–14.
- Trueba D, Sawaya BP, Mawad H, Malluche HH. Bone biopsy: indications, techniques, and complications. Semin Dial 2003; 16:341–345.
- Melton LJ. Epidemiology worldwide. Endocrinol Metab Clin North Am 2003; 32:1–13.
- Burge R, Dawson-Hughes B, Solomon DH, Wong JB, King A, Tosteson A. Incidence and economic burden of osteoporosis-related fractures in the United States, 2005–2025. J Bone Miner Res 2007; 22:465–475.
- Barrett-Connor E, Siris ES, Wehren LE, et al. Osteoporosis and fracture risk in women of different ethnic groups. J Bone Miner Res 2005; 20:185–194.
- Assessment of fracture risk and its application to screening for postmenopausal osteoporosis. Report of a WHO Study Group. World Health Organ Tech Rep Ser 1994; 843:1–129.
- Miller PD, Bonnick SL. Clinical application of bone densitometry. In:Favus MJ, editor. Primer on the Metabolic Bone Diseases and Disorders of Mineral Metabolism. 4th ed. American Society for Bone and Mineral Research. Philadelphia, PA: Lippincott Williams & Wilkins; 1999.
- Nickolas TL, Leonard MB, Shane E. Chronic kidney disease and bone fracture: a growing concern. Kidney Int 2008; 74:721–731.
- Gal-Moscovici A, Sprague SM. Osteoporosis and chronic kidney disease. Semin Dial 2007; 20:423–430.
- National Kidney Foundation. Clinical practice guidelines for bone metabolism and disease in chronic kidney disease. Am J Kidney Dis 2003; 42(suppl 3):S1–S201.
- Coresh J, Astor BC, Greene T, Eknoyan G, Levey AS. Prevalence of chronic kidney disease and decreased kidney function in the adult US population: Third National Health and Nutrition Examination Survey. Am J Kidney Dis 2003; 41:1–12.
- Bennett WM. Reporting eGFR. Clin J Am Soc Nephrol 2008; 3:1561–1562.
- Glassock RJ, Winearls C. Screening for CKD with eGFR: doubts and dangers. Clin J Am Soc Nephrol 2008; 3:1563–1568.
- Parfitt AM, Drezner M, Glorieux F, et al. Bone histomorphometry: standardization of nomenclature, symbols, and units. Report of the ASBMR Histomorphometry Nomenclature Committee. J Bone Miner Res 1987; 2:595–610.
- Andress DL, Sherrard DJ. The osteodystrophy of chronic renal failure. In:Schrier RW, editor: Diseases of the Kidney and Urinary Tract. Philadelphia, PA: Lippincott Williams & Wilkins; 2007:2431–2453.
- Moe S, Drueke T, Cunningham J, et al. Definition, evaluation, and classification of renal osteodystrophy: a position statement from Kidney Disease: Improving Global Outcomes (KDIGO). Kidney Int 2006; 69:1945–1953.
- NIH Consensus Development Panel Osteoporosis Prevention Diagnosis and Therapy. Osteoporosis prevention, diagnosis, and therapy. JAMA 2001; 285:785–795.
- Baim S, Binkley N, Bilezikian JP, et al. Official positions of the International Society for Clinical Densitometry and executive summary of the 2007 ISCD Position Development Conference. J Clin Densitom 2008; 11:75–91.
- Siris E, Miller P, Barrett-Connor E, et al. Identification and fracture outcomes of undiagnosed low bone mineral density in postmenopausal women: results from the National Osteoporosis Risk Assessment (NORA). JAMA 2001; 286:2815–2822.
- Schuit SCE, Oei HH, Witteman JC, et al. Fracture incidence and association with bone mineral density in elderly men and women: the Rotterdam Study. Bone 2004; 34:195–202.
- Bouxsein ML. Non-invasive measurements of bone strength: promise and peril. J Musculoskelet Neuronal Interact 2004; 4:404–405.
- Seeman E. Bone quality: the material and structural basis of bone strength. J Bone Miner Metab 2008; 26:1–8.
- Alem AM, Sherrard DJ, Gillen DL, et al. Increased risk of hip fracture among patients with end-stage renal disease. Kidney Int 2000; 58:396–399.
- Ball AM, Gillen DL, Sherrard D, et al. Risk of hip fracture among dialysis and renal transplant recipients. JAMA 2002; 288:3014–3018.
- Jadoul M, Albert JM, Akiba T, et al. Incidence and risk factors for hip or other bone fractures among hemodialysis patients in the Dialysis Outcomes and Practice Patterns Study. Kidney Int 2006; 70:1358–1366.
- Stehman-Breen CO, Sherrard DJ, Alem AM, et al. Risk factors for hip fracture among patients with end-stage renal disease. Kidney Int 2000; 58:2200–2205.
- Wehrli FW, Leonard MB, Saha PK, Gomberg BR. Quantitative highresolution magnetic resonance imaging reveals structural implications of renal osteodystrophy on trabecular and cortical bone. J Magn Reson Imaging 2004; 20:83–89.
- Genant HK, Lang TF, Engelke K, et al. Advances in the noninvasive assessment of bone density, quality, and structure. Calcif Tissue Int 1996; 59(suppl 1):S10–S15.
- Roschger P, Paschalis EP, Fratzl P, Klaushofer K. Bone mineralization density distribution in health and disease. Bone 2008; 42:456–466.
- Miller PD. Bone density and markers of bone turnover in predicting fracture risk and how changes in these measures predict fracture risk reduction. Curr Osteoporos Rep 2005; 3:103–110.
- Miller PD, Hochberg MC, Wehren LE, Ross PD, Wasnich RD. How useful are measures of BMD and bone turnover? Curr Med Res Opin 2005; 21:545–554.
- Chavassieux PM, Delmas PD. Bone remodeling: biochemical markers or bone biopsy? J Bone Miner Res 2006; 21:178–179.
- Garnero P. Biomarkers for osteoporosis management: utility in diagnosis, fracture risk prediction and therapy monitoring. Mol Diagn Ther 2008; 12:157–170.
- Hochberg M, Greenspan S, Wasnich R, Miller P, Thompson D, Ross P. Changes in bone density and turnover explain the reductions in incidence of nonvertebral fractures that occur during treatment with antiresorptive agents. J Clin Endocrinol Metab 2002; 87:1586–1592.
- Bouxsein ML, Delmas PD. Considerations for development of surrogate endpoints for antifracture efficacy of new treatments in osteoporosis: a perspective. J Bone Miner Res 2008; 23:1155–1167.
- Chen P, Satterwhite JH, Licatta AA, et al. Early changes in biochemical markers of bone formation predict BMD response to teriparatide in postmenopausal women with osteoporosis. J Bone Miner Res 2005; 20:962–970.
- Miller PD, Delmas PD, Lindsay R, et al. Early responsiveness of women with osteoporosis to teriparatide after therapy with alendronate or risedronate. J Clin Endocrinol Metab 2008; 93:3785–3793.
- Baim S, Miller PD. Assessing the clinical utility of serum CTX in postmenopausal osteoporosis and its use in predicting risk of osteonecrosis of the jaw. J Bone Miner Res 2009; 24:561–574.
- Miller PD, Lerma EV. Renal bone diseases. In:Kleerekoper M, Siris E, McClung M, editors. The Bone and Mineral Manual—A Practical Guide. 2nd ed. Burlington, MA: Elsevier Academic Press; 2005:127–138.
- Miller PD, Shane E. Management of transplantation renal bone disease: Interplay of bone mineral density and decisions regarding bisphosphonate use. In:Weir MR, editor. Medical Management of Kidney Transplantation. Philadelphia, PA: Lippincott Williams & Wilkins; 2004:359–375.
- Levin A, Bakris GL, Molitch M, et al. Prevalence of abnormal serum vitamin D, PTH, calcium, and phosphorus in patients with chronic kidney disease: results of the study to evaluate early kidney disease. Kidney int 2007; 71:31–38.
- Vassalotti JA, Uribarri J, Chen SC, et al. Trends in mineral metabolism: Kidney Early Evaluation Program (KEEP) and the National Health and Nutrition Examination Survey (NHANES) 1999–2004. Am J Kidney Dis 2008; 51(suppl 2):S56–S68.
- Meier C, Seibel MJ, Kraenzlin ME. Use of bone turnover markers in the real world: are we there yet? J Bone Miner Res 2009; 24:386–388.
- Lehmann G, Ott U, Kaemmerer D, Schuetze J, Wolf G. Bone histomorphometry and biochemical markers of bone turnover in patients with chronic kidney disease stages 3–5. Clin Nephrol 2008; 70:296–305.
- Miller PD. The role of bone biopsy in patients with chronic renal failure. Clin J Am Soc Nephrol 2008; 3(suppl 3):S140–S150.
- Frost HM. Tetracycline-based histological analysis of bone remodeling. Calcif Tissue Res 1969; 3:211–237.
- Hitt O, Jaworski ZF, Shimizu AG, Frost HM. Tissue-level bone formation rates in chronic renal failure, measured by means of tetracycline bone labeling. Can J Physiol Pharmacol 1970; 48:824–828.
- Coen G. Adynamic bone disease: an update and overview. J Nephrol 2005; 18:117–122.
- Parfitt AM. Renal bone disease: a new conceptual framework for the interpretation of bone histomorphometry. Curr Opin Nephrol Hypertens 2003; 12:387–403.
- Brandenburg VM, Floege J. Adynamic bone diseaseùbone and beyond. NDT Plus 2008; 3:135–147. doi:10.1093/ndtplus/sfn040.
- Hruska KA, Saab G, Mathew S, Lund R. Renal osteodystrophy, phosphate homeostasis, and vascular calcification. Semin Dial 2007; 20:309–315.
- Toussaint ND, Elder GJ, Kerr PG. Bisphosphonates in chronic kidney disease; balancing potential benefits and adverse effects on bone and soft tissue. Clin J Am Soc Nephrol 2009; 4:221–233.
- Miller PD. Is there a role for bisphosphonates in chronic kidney disease? Semin Dial 2007; 20:186–190.
- Miller PD. Bisphosphonates: pharmacology and use in the treatment of osteoporosis. In:Marcus R, Feldman D, Nelson DA, Rosen CJ, editors. Osteoporosis. 3rd ed. Boston, MA: Elsevier Academic Press; 2008:1725–1736.
- Russell RG, Watts NB, Ebetino FH, Rogers MJ. Mechanisms of action of bisphosphonates: similarities and differences and their potential influence on clinical efficacy. Osteoporosis Int 2008; 19:733–759.
- Eisman JA, Civitelli R, Adami S, et al. Efficacy and tolerability of intravenous ibandronate injections in postmenopausal osteoporosis: 2-year results from the DIVA study. J Rheumatol 2008; 35:488–497.
- Black DM, Delmas PD, Eastell RR, et al. Once-yearly zoledronic acid for treatment of postmenopausal osteoporosis. N Engl J Med 2007; 356:1809–1822.
- Lewiecki EM, Miller PD. Renal safety of intravenous bisphosphonates in the treatment of osteoporosis. Expert Opin Drug Saf 2007; 6:663–672.
- Miller PD. Anti-resorptives in the management of osteoporosis. Best Pract Res Clin Endocrinol Metab 2008; 22:849–868.
- Perazella MA, Markowitz GS. Bisphosphonate nephrotoxicity. Kidney Int 2008; 74:1385–1393.
- Boonen S, Sellmeyer DE, Lippuner K, et al. Renal safety of annual zoledronic acid infusions in osteoporotic postmenopausal women. Kidney Int 2008; 74:641–648.
- Neer RM, Arnaud CD, Zanchetta JR, et al. Effect of parathyroid hormone (1–34) on fractures and bone mineral density in postmenopausal women with osteoporosis. N Engl J Med 2001; 344:1434–1441.
- Miller PD, Schwartz EN, Chen P, Misurski DA, Krege JH. Teriparatide in postmenopausal women with osteoporosis and mild or moderate renal impairment. Osteoporosis Int 2007; 18:59–68.
- Miller PD, Roux C, Boonen S, Barton I, Dunlap L, Burgio D. Safety and efficacy of risedronate in patients with age-related reduced renal function as estimated by the Cockcroft and Gault method: a pooled analysis of nine clinical trials. J Bone Miner Res 2005; 20:2105–2115.
- Jamal SA, Bauer DC, Ensrud KE, et al. Alendronate treatment in women with normal to severely impaired renal function: an analysis of the fracture intervention trial. J Bone Miner Res 2007; 22:503–508.
- Ishani A, Blackwell T, Jamal SA, Cummings SR, Ensrud KE; MORE Investigators. The effect of raloxifene treatment in postmenopausal women with CKD. J Am Soc Nephrol 2008; 19:1430–1438.
- Coco M, Glicklich D, Faugere MC, et al. Prevention of bone loss in renal transplant recipients: a prospective, randomized trial of intravenous pamidronate. J Am Soc Nephrol 2003; 14:2669–2676.
- Palmer SC, McGregor DO, Strippoli GF. Interventions for preventing bone disease in kidney transplant recipients. Cochrane Database Syst Rev 2007;CD005015.
- Ferreira MA. Diagnosis of renal osteodystrophy: when and how to use biochemical markers and non-invasive methods; when bone biopsy is needed. Nephrol Dial Transplant 2000; 15(suppl 5):8–14.
- Trueba D, Sawaya BP, Mawad H, Malluche HH. Bone biopsy: indications, techniques, and complications. Semin Dial 2003; 16:341–345.
KEY POINTS
- If the patient’s glomerular filtration rate (GFR) is at least 30 mL/min/1.73 m2 and if no biochemical test results suggest renal osteodystrophy, osteoporosis can be diagnosed if the T score is less than −2.5 or if the patient has had a fragility fracture. These criteria can also probably be applied, though with less certainty, if the patient’s GFR is as low as 15.
- If the patient’s GFR is less than 15 or if he or she is on dialysis, biochemical profiling often cannot distinguish among the heterogeneous forms of renal bone disease. In some cases of severe chronic kidney disease with fractures, bone biopsy is needed to rule out renal osteodystrophy and to diagnose osteoporosis by exclusion.
- In the author’s opinion, in patients with severe chronic kidney disease and fractures who have “osteoporosis” by exclusion, off-label use of bisphosphonates is an option, but only after very careful consideration.
Treating the renal patient who has a fracture: Opinion vs evidence
Managing bone health in patients with chronic kidney disease presents unique challenges. While the common end point—a fracture—is comparable to that in patients with osteoporosis, the underlying metabolic conditions differ from patient to patient with chronic kidney disease and may be dramatically different from those in patients who have osteoporosis without chronic kidney disease.
Renal osteodystrophy is not osteoporosis
Renal osteodystrophy is not osteoporosis. While osteoporosis in people without kidney disease is defined clinically on the basis of bone mineral density (measured by bone densitometry), renal osteodystrophy is a histologic diagnosis made on bone biopsy: it is a continuum between frankly low-turnoverbone disease—encompassing adynamic bone disease and osteomalacia—and frankly highturnover-bone disease, with severe secondary hyperparathyroid bone disease and osteitis fibrosa. Histologically, there may or may not be low trabecular bone volume or loss of connectivity typical of the bone loss in osteoporosis.
Patients at both ends of the spectrum of bone turnover in renal osteodystrophy may have the same bone mineral density on densitometry. Low bone mineral density may reflect inadequate mineralization (seen in osteomalacia and adynamic bone disease) or increased peritrabecular fibrosis (seen in secondary hyperparathyroid bone disease). High bone mineral density readings may capture extraosseous calcifications, which are very common in chronic kidney disease.
Renal osteodystrophy is part of the syndrome called chronic kidney disease-mineral and bone disease,1 which is not limited to bone fractures but may also affect vascular health. Abnormal calcium deposits in vascular tissue—consistent with calciphylaxis and associated with increased morbidity and mortality rates in chronic kidney disease—may occur with low bone turnover.
The diagnosis of osteoporosis in the general population is based on clinical evidence: the measured bone mineral density is compared with normalized scores. Histologically, the bone of the osteoporotic patient shows osteopenia with increased bone turnover and a shift toward increased bone resorption, resulting in loss of connectivity of the trabeculae, as well as decreased trabecular volume. These conditions are common in advanced age and in certain pathologic states (eg, steroid therapy, metastatic bone disease, Paget disease of bone).
It is well accepted that the risk of fracture in osteoporosis increases as measured bone mineral density decreases. Conversely, increasing bone mineral density has been correlated with fewer fractures. The clinician is often guided by biomarkers of bone metabolism such as urinary N-terminal cross-linked telopeptides of collagen (NTx) in diagnosing and treating bone breakdown.
Can bisphosphonates be used in chronic kidney disease?
Bisphosphonates are antiresorptive agents that bind to the hydroxyapatite of bone. They poison the osteoclast (the bone-resorbing cell), causing its death and thereby halting the resorption of bone. Osteoblasts—the boneforming cells—are presumably not affected, and the bone continues to make osteoid, which is subsequently mineralized. Bone turnover is dramatically decreased. The net effect is increased bone density in people with osteoporosis. The half-life of these agents is years.
In the general population, bisphosphonate therapy has been associated with decreased risk of fragility bone fractures. However, the long-term effects are not yet known. Indeed, jaw necrosis—possibly due to low bone turnover—is being reported with increasing frequency.2 Fractures associated with low bone turnover in patients without chronic kidney disease treated with bisphosphonates longterm are now being reported.3,4
In an article in this issue of the Journal,5 the author advocates the use of bisphosphonate therapy in patients with chronic kidney disease who have low bone mineral density. However, treating patients who have chronic kidney disease on the basis of low bone mineral density with bone-suppressing agents may further depress bone turnover and lead to more extraosseous calcifications as the turnedoff bone is unable to accept serum calcium.6
Further, it is unclear how long “long-term” would be in a patient with advanced chronic kidney disease: Would the half-life of the bisphosphonates be tremendously increased, leading to adverse events sooner? Would adynamic bone disease promptly develop, leading to rampant jaw necrosis and bone fractures? Would vascular calcification flourish?
Bone biomarkers are hard to interpret in chronic kidney disease
In chronic kidney disease, the interpretation of biomarkers of bone metabolism is notoriously unreliable. The usual chemistry values associated with clinical osteoporosis in the general population—ie, elevated levels of urinary NTx, serum C-terminal cross-linked telopeptides of collagen (CTx), osteocalcin, and bone-specific alkaline phosphatase—are not valid in patients with chronic kidney disease, for obvious reasons: with declining renal function, the various markers accumulate in the serum. Urinary NTx does not apply in patients with advanced chronic kidney disease or end-stage renal disease.
How should renal osteodystrophy be treated?
Nephrologists currently focus therapy on reducing hyperphosphatemia (associated with increased morbidity across all stages of chronic kidney disease), replenishing vitamin D as much as possible without causing hyperphosphatemia and hypercalcemia, and suppressing parathyroid hormone secretion.
However, there is not enough evidence on what the goal should be with respect to parathyroid hormone in patients with chronic kidney disease who are not on dialysis. Although in the recent past many believed that parathyroid hormone goals should be 150 to 300 pg/mL in dialysis patients, the latest guidelines suggest that perhaps this goal is too narrow and may lead to more adynamic bone disease. Similarly, there is no consensus on the use of synthetic parathyroid hormone analogues.
Bisphosphonate therapy, particularly with pamidronate (Aredia) and zolendronic acid (Reclast), has been associated with adverse renal effects even in patients without chronic kidney disease. There are no prospective studies of the effects of these agents in patients with depressed renal function.
The patient with chronic kidney disease who has a fracture remains a unique problem for the nephrologist, primary care physician, and subspecialist. Efforts should be concentrated on preventing and treating metabolic bone disease in its entire spectrum, with rational, prospective studies, and should not depend on anecdotal reports. Opinions abound, without adequate evidence to back them up.
- Moe S, Drüeke T, Cunningham J, et al; Kidney Disease: Improving Global Outcomes (KDIGO). Definition, evaluation, and classification of renal osteodystrophy: a position statement from Kidney Disease: Improving Global Outcomes (KDIGO). Kidney Int 2006; 69:1945–1953.
- Rustemeyer J, Bremerich A. Bisphosphonate-associated osteonecrosis of the jaw: what do we currently know? A survey of knowledge given in the recent literature. Clin Oral Investig 2009; Epub ahead of print.
- Armamento-Villareal R, Napoli N, Diemer K, et al. Bone turnover in bone biopsies of patients with low-energy cortical fractures receiving bisphosphonates: a case series. Calcif Tissue Int 2009; 85:37–44.
- Ali T, Jay RH. Spontaneous femoral shaft fracture after long-term alendronate. Age Ageing 2009; Epub ahead of print.
- Miller PD. Fragility fractures in chronic kidney disease: an opinionbased approach. Cleve Clin J Med 2009; 76:713–721.
- Toussaint ND, Elder GJ, Kerr PG. Bisphosphonates in chronic kidney disease; balancing potential benefits and adverse effects on bone and soft tissue. Clin J Am Soc Nephrol 2009; 4:221–233.
Managing bone health in patients with chronic kidney disease presents unique challenges. While the common end point—a fracture—is comparable to that in patients with osteoporosis, the underlying metabolic conditions differ from patient to patient with chronic kidney disease and may be dramatically different from those in patients who have osteoporosis without chronic kidney disease.
Renal osteodystrophy is not osteoporosis
Renal osteodystrophy is not osteoporosis. While osteoporosis in people without kidney disease is defined clinically on the basis of bone mineral density (measured by bone densitometry), renal osteodystrophy is a histologic diagnosis made on bone biopsy: it is a continuum between frankly low-turnoverbone disease—encompassing adynamic bone disease and osteomalacia—and frankly highturnover-bone disease, with severe secondary hyperparathyroid bone disease and osteitis fibrosa. Histologically, there may or may not be low trabecular bone volume or loss of connectivity typical of the bone loss in osteoporosis.
Patients at both ends of the spectrum of bone turnover in renal osteodystrophy may have the same bone mineral density on densitometry. Low bone mineral density may reflect inadequate mineralization (seen in osteomalacia and adynamic bone disease) or increased peritrabecular fibrosis (seen in secondary hyperparathyroid bone disease). High bone mineral density readings may capture extraosseous calcifications, which are very common in chronic kidney disease.
Renal osteodystrophy is part of the syndrome called chronic kidney disease-mineral and bone disease,1 which is not limited to bone fractures but may also affect vascular health. Abnormal calcium deposits in vascular tissue—consistent with calciphylaxis and associated with increased morbidity and mortality rates in chronic kidney disease—may occur with low bone turnover.
The diagnosis of osteoporosis in the general population is based on clinical evidence: the measured bone mineral density is compared with normalized scores. Histologically, the bone of the osteoporotic patient shows osteopenia with increased bone turnover and a shift toward increased bone resorption, resulting in loss of connectivity of the trabeculae, as well as decreased trabecular volume. These conditions are common in advanced age and in certain pathologic states (eg, steroid therapy, metastatic bone disease, Paget disease of bone).
It is well accepted that the risk of fracture in osteoporosis increases as measured bone mineral density decreases. Conversely, increasing bone mineral density has been correlated with fewer fractures. The clinician is often guided by biomarkers of bone metabolism such as urinary N-terminal cross-linked telopeptides of collagen (NTx) in diagnosing and treating bone breakdown.
Can bisphosphonates be used in chronic kidney disease?
Bisphosphonates are antiresorptive agents that bind to the hydroxyapatite of bone. They poison the osteoclast (the bone-resorbing cell), causing its death and thereby halting the resorption of bone. Osteoblasts—the boneforming cells—are presumably not affected, and the bone continues to make osteoid, which is subsequently mineralized. Bone turnover is dramatically decreased. The net effect is increased bone density in people with osteoporosis. The half-life of these agents is years.
In the general population, bisphosphonate therapy has been associated with decreased risk of fragility bone fractures. However, the long-term effects are not yet known. Indeed, jaw necrosis—possibly due to low bone turnover—is being reported with increasing frequency.2 Fractures associated with low bone turnover in patients without chronic kidney disease treated with bisphosphonates longterm are now being reported.3,4
In an article in this issue of the Journal,5 the author advocates the use of bisphosphonate therapy in patients with chronic kidney disease who have low bone mineral density. However, treating patients who have chronic kidney disease on the basis of low bone mineral density with bone-suppressing agents may further depress bone turnover and lead to more extraosseous calcifications as the turnedoff bone is unable to accept serum calcium.6
Further, it is unclear how long “long-term” would be in a patient with advanced chronic kidney disease: Would the half-life of the bisphosphonates be tremendously increased, leading to adverse events sooner? Would adynamic bone disease promptly develop, leading to rampant jaw necrosis and bone fractures? Would vascular calcification flourish?
Bone biomarkers are hard to interpret in chronic kidney disease
In chronic kidney disease, the interpretation of biomarkers of bone metabolism is notoriously unreliable. The usual chemistry values associated with clinical osteoporosis in the general population—ie, elevated levels of urinary NTx, serum C-terminal cross-linked telopeptides of collagen (CTx), osteocalcin, and bone-specific alkaline phosphatase—are not valid in patients with chronic kidney disease, for obvious reasons: with declining renal function, the various markers accumulate in the serum. Urinary NTx does not apply in patients with advanced chronic kidney disease or end-stage renal disease.
How should renal osteodystrophy be treated?
Nephrologists currently focus therapy on reducing hyperphosphatemia (associated with increased morbidity across all stages of chronic kidney disease), replenishing vitamin D as much as possible without causing hyperphosphatemia and hypercalcemia, and suppressing parathyroid hormone secretion.
However, there is not enough evidence on what the goal should be with respect to parathyroid hormone in patients with chronic kidney disease who are not on dialysis. Although in the recent past many believed that parathyroid hormone goals should be 150 to 300 pg/mL in dialysis patients, the latest guidelines suggest that perhaps this goal is too narrow and may lead to more adynamic bone disease. Similarly, there is no consensus on the use of synthetic parathyroid hormone analogues.
Bisphosphonate therapy, particularly with pamidronate (Aredia) and zolendronic acid (Reclast), has been associated with adverse renal effects even in patients without chronic kidney disease. There are no prospective studies of the effects of these agents in patients with depressed renal function.
The patient with chronic kidney disease who has a fracture remains a unique problem for the nephrologist, primary care physician, and subspecialist. Efforts should be concentrated on preventing and treating metabolic bone disease in its entire spectrum, with rational, prospective studies, and should not depend on anecdotal reports. Opinions abound, without adequate evidence to back them up.
Managing bone health in patients with chronic kidney disease presents unique challenges. While the common end point—a fracture—is comparable to that in patients with osteoporosis, the underlying metabolic conditions differ from patient to patient with chronic kidney disease and may be dramatically different from those in patients who have osteoporosis without chronic kidney disease.
Renal osteodystrophy is not osteoporosis
Renal osteodystrophy is not osteoporosis. While osteoporosis in people without kidney disease is defined clinically on the basis of bone mineral density (measured by bone densitometry), renal osteodystrophy is a histologic diagnosis made on bone biopsy: it is a continuum between frankly low-turnoverbone disease—encompassing adynamic bone disease and osteomalacia—and frankly highturnover-bone disease, with severe secondary hyperparathyroid bone disease and osteitis fibrosa. Histologically, there may or may not be low trabecular bone volume or loss of connectivity typical of the bone loss in osteoporosis.
Patients at both ends of the spectrum of bone turnover in renal osteodystrophy may have the same bone mineral density on densitometry. Low bone mineral density may reflect inadequate mineralization (seen in osteomalacia and adynamic bone disease) or increased peritrabecular fibrosis (seen in secondary hyperparathyroid bone disease). High bone mineral density readings may capture extraosseous calcifications, which are very common in chronic kidney disease.
Renal osteodystrophy is part of the syndrome called chronic kidney disease-mineral and bone disease,1 which is not limited to bone fractures but may also affect vascular health. Abnormal calcium deposits in vascular tissue—consistent with calciphylaxis and associated with increased morbidity and mortality rates in chronic kidney disease—may occur with low bone turnover.
The diagnosis of osteoporosis in the general population is based on clinical evidence: the measured bone mineral density is compared with normalized scores. Histologically, the bone of the osteoporotic patient shows osteopenia with increased bone turnover and a shift toward increased bone resorption, resulting in loss of connectivity of the trabeculae, as well as decreased trabecular volume. These conditions are common in advanced age and in certain pathologic states (eg, steroid therapy, metastatic bone disease, Paget disease of bone).
It is well accepted that the risk of fracture in osteoporosis increases as measured bone mineral density decreases. Conversely, increasing bone mineral density has been correlated with fewer fractures. The clinician is often guided by biomarkers of bone metabolism such as urinary N-terminal cross-linked telopeptides of collagen (NTx) in diagnosing and treating bone breakdown.
Can bisphosphonates be used in chronic kidney disease?
Bisphosphonates are antiresorptive agents that bind to the hydroxyapatite of bone. They poison the osteoclast (the bone-resorbing cell), causing its death and thereby halting the resorption of bone. Osteoblasts—the boneforming cells—are presumably not affected, and the bone continues to make osteoid, which is subsequently mineralized. Bone turnover is dramatically decreased. The net effect is increased bone density in people with osteoporosis. The half-life of these agents is years.
In the general population, bisphosphonate therapy has been associated with decreased risk of fragility bone fractures. However, the long-term effects are not yet known. Indeed, jaw necrosis—possibly due to low bone turnover—is being reported with increasing frequency.2 Fractures associated with low bone turnover in patients without chronic kidney disease treated with bisphosphonates longterm are now being reported.3,4
In an article in this issue of the Journal,5 the author advocates the use of bisphosphonate therapy in patients with chronic kidney disease who have low bone mineral density. However, treating patients who have chronic kidney disease on the basis of low bone mineral density with bone-suppressing agents may further depress bone turnover and lead to more extraosseous calcifications as the turnedoff bone is unable to accept serum calcium.6
Further, it is unclear how long “long-term” would be in a patient with advanced chronic kidney disease: Would the half-life of the bisphosphonates be tremendously increased, leading to adverse events sooner? Would adynamic bone disease promptly develop, leading to rampant jaw necrosis and bone fractures? Would vascular calcification flourish?
Bone biomarkers are hard to interpret in chronic kidney disease
In chronic kidney disease, the interpretation of biomarkers of bone metabolism is notoriously unreliable. The usual chemistry values associated with clinical osteoporosis in the general population—ie, elevated levels of urinary NTx, serum C-terminal cross-linked telopeptides of collagen (CTx), osteocalcin, and bone-specific alkaline phosphatase—are not valid in patients with chronic kidney disease, for obvious reasons: with declining renal function, the various markers accumulate in the serum. Urinary NTx does not apply in patients with advanced chronic kidney disease or end-stage renal disease.
How should renal osteodystrophy be treated?
Nephrologists currently focus therapy on reducing hyperphosphatemia (associated with increased morbidity across all stages of chronic kidney disease), replenishing vitamin D as much as possible without causing hyperphosphatemia and hypercalcemia, and suppressing parathyroid hormone secretion.
However, there is not enough evidence on what the goal should be with respect to parathyroid hormone in patients with chronic kidney disease who are not on dialysis. Although in the recent past many believed that parathyroid hormone goals should be 150 to 300 pg/mL in dialysis patients, the latest guidelines suggest that perhaps this goal is too narrow and may lead to more adynamic bone disease. Similarly, there is no consensus on the use of synthetic parathyroid hormone analogues.
Bisphosphonate therapy, particularly with pamidronate (Aredia) and zolendronic acid (Reclast), has been associated with adverse renal effects even in patients without chronic kidney disease. There are no prospective studies of the effects of these agents in patients with depressed renal function.
The patient with chronic kidney disease who has a fracture remains a unique problem for the nephrologist, primary care physician, and subspecialist. Efforts should be concentrated on preventing and treating metabolic bone disease in its entire spectrum, with rational, prospective studies, and should not depend on anecdotal reports. Opinions abound, without adequate evidence to back them up.
- Moe S, Drüeke T, Cunningham J, et al; Kidney Disease: Improving Global Outcomes (KDIGO). Definition, evaluation, and classification of renal osteodystrophy: a position statement from Kidney Disease: Improving Global Outcomes (KDIGO). Kidney Int 2006; 69:1945–1953.
- Rustemeyer J, Bremerich A. Bisphosphonate-associated osteonecrosis of the jaw: what do we currently know? A survey of knowledge given in the recent literature. Clin Oral Investig 2009; Epub ahead of print.
- Armamento-Villareal R, Napoli N, Diemer K, et al. Bone turnover in bone biopsies of patients with low-energy cortical fractures receiving bisphosphonates: a case series. Calcif Tissue Int 2009; 85:37–44.
- Ali T, Jay RH. Spontaneous femoral shaft fracture after long-term alendronate. Age Ageing 2009; Epub ahead of print.
- Miller PD. Fragility fractures in chronic kidney disease: an opinionbased approach. Cleve Clin J Med 2009; 76:713–721.
- Toussaint ND, Elder GJ, Kerr PG. Bisphosphonates in chronic kidney disease; balancing potential benefits and adverse effects on bone and soft tissue. Clin J Am Soc Nephrol 2009; 4:221–233.
- Moe S, Drüeke T, Cunningham J, et al; Kidney Disease: Improving Global Outcomes (KDIGO). Definition, evaluation, and classification of renal osteodystrophy: a position statement from Kidney Disease: Improving Global Outcomes (KDIGO). Kidney Int 2006; 69:1945–1953.
- Rustemeyer J, Bremerich A. Bisphosphonate-associated osteonecrosis of the jaw: what do we currently know? A survey of knowledge given in the recent literature. Clin Oral Investig 2009; Epub ahead of print.
- Armamento-Villareal R, Napoli N, Diemer K, et al. Bone turnover in bone biopsies of patients with low-energy cortical fractures receiving bisphosphonates: a case series. Calcif Tissue Int 2009; 85:37–44.
- Ali T, Jay RH. Spontaneous femoral shaft fracture after long-term alendronate. Age Ageing 2009; Epub ahead of print.
- Miller PD. Fragility fractures in chronic kidney disease: an opinionbased approach. Cleve Clin J Med 2009; 76:713–721.
- Toussaint ND, Elder GJ, Kerr PG. Bisphosphonates in chronic kidney disease; balancing potential benefits and adverse effects on bone and soft tissue. Clin J Am Soc Nephrol 2009; 4:221–233.
Public reporting and pay-for-performance programs in perioperative medicine
Hospital quality measures and rankings are now widely available to the public online, but is public reporting of this information an effective strategy for improving health care? Using a case study of a hospital that suffered negative publicity as a result of a quality report, this article explores the use of public reporting of performance data and pay-for-performance reimbursement strategies to foster quality improvement in the US health care system.
CASE STUDY: A SURGICAL PROGRAM GETS A BAD REPORT―IN THE HEADLINES
In September 2005, The Boston Globe ran a prominent story reporting that the UMass Memorial Medical Center in Worcester, Mass., was abruptly suspending its elective cardiac surgery program.1 The program’s suspension came after state public health officials presented UMass Memorial with a detailed analysis showing that the hospital’s mortality rate for coronary artery bypass graft surgery (CABG) patients was the highest in the state and almost double the average for Massachusetts hospitals.1
Key personnel from UMass Memorial described the events preceding and following the program’s suspension in a journal article published in 2008.2 In 2002, UMass Memorial hired a new chief of cardiothoracic surgery, who resigned in early 2005. A few months after that resignation, state public health officials alerted the hospital to the abovementioned CABG mortality data (from 2002 and 2003), which they said would soon be reported publicly. UMass Memorial then conducted an internal review of its data from the most recent years (2004 and 2005) and found that its risk-adjusted CABG mortality had actually worsened, at which point the hospital voluntarily suspended its cardiac surgery program.2
More news stories arose about UMass Memorial’s program and its problems. The hospital hired consultants and senior surgeons from around the state and New England to completely review its cardiac surgery program. They concluded that “many essential systems were not in place” and made 68 key recommendations, including a complete overhaul of the hospital’s quality-improvement structure. The prior cardiac surgeons departed.2
The cardiac surgery program resumed after a 6-week hiatus, with day-to-day supervision by two senior cardiac surgeons from a Boston teaching hospital. A nationally recognized cardiac surgeon was brought on as chief of cardiac surgery in January 2006. In the 18 months after the program resumed, risk-adjusted CABG mortality rates declined substantially, but patient volume failed to return to presuspension levels and the hospital reported $22 million in lost revenue in fiscal year 2006 as a result of the suspension.2
This case raises a number of questions that help to frame discussion of the benefits and risks of public reporting of hospital quality measures:
- To what extent does public reporting accelerate quality improvement?
- How typical was the subsequent mortality reduction reported by UMass Memorial—ie, can public reporting be expected to improve outcomes?
- Was the effect on patient volume expected—ie, how much does public reporting affect market share?
- Would a pay-for-performance reimbursement model have accelerated improvement?
- Why do public reporting and pay-for-performance programs remain controversial?
- Do patients have a right to know?
WHAT HAS FUELED THE MOVE TOWARD PUBLIC REPORTING?
Drivers of public reporting
Massachusetts is one of a number of states that publicly report outcomes from cardiac surgery and other procedures and processes of care. Three basic factors have helped drive the development of public reporting (and, in some cases, pay-for-performance) programs:
- National policy imperatives designed to improve quality and safety and to reduce costs
- Cultural factors in society, which include consumerism in health care and the desire for transparency
- The growth of information technology and use of the World Wide Web, which has been a huge enabler of public reporting. Public reporting could be done prior to the Web era but would not have reached such a wide audience had the results been released in a book that had to be ordered from a government printing office.
The rationale for public reporting
In theory, how might public reporting and pay-for-performance programs improve quality? Several different mechanisms or factors are likely to be involved:
- Feedback. The basic premise of the National Surgical Quality Improvement Program, to cite one example, is that peer comparison and performance feedback will stimulate quality improvement.
- Reputation. Hospital personnel fear being embarrassed if data show that they are performing poorly compared with other hospitals. Likewise, in recent years we have seen hospitals with the best quality rankings publicly advertise their performance.
- Market share. Here the premise is that patients will tend to select providers with higher quality rankings and shun those with lower rankings.
- Financial incentives. Pay-for-performance programs link payment or reimbursement directly to the desired outcomes and thereby stimulate quality improvement without working through the abovementioned mechanisms.
Approaches to quality measurement
Public reporting of hospital performance requires selection of an approach to measuring quality of care. Generally speaking, measures of health care quality reflect one of three domains of care:
Structural (or environmental) aspects, such as staffing in the intensive care unit (ICU), surgical volume, or availablity of emergency medical responders. An example of a structure-oriented reporting system is the Leapfrog Group’s online posting of hospital ratings based on surgical volumes for high-risk procedures, the degree of computerized order entry implementation, and the presence or absence of various patient safety practices.3
Processes of care, such as whether beta-blockers are prescribed for all patients after a myocardial infarction (MI), or whether thromboprophylaxis measures are ordered for surgical patients in keeping with guideline recommendations. Examples of process-oriented reporting systems include the US Department of Health and Human Services’ Hospital Compare Web site4 and the Commonwealth Fund’s WhyNotTheBest.org site.5
Outcomes of care, such as rates of mortality or complications, or patient satisfaction rates. An example of an outcomes-oriented reporting system is the annual report of institution-specific hospital-acquired infection rates put out by Pennsylvania6 and most other states.
IS THERE EVIDENCE OF BENEFIT?
A consistent effect in spurring quality-improvement efforts
Nearly a dozen published studies have evaluated whether public reporting stimulates quality-improvement activities, and the results have shown fairly consistently that it does. A 2003 study by Hibbard et al is representative of the results.7 This survey-based investigation measured the number of quality-improvement activities in cardiac and obstetric care undertaken by 24 Wisconsin hospitals that were included in an existing public reporting system compared with the number undertaken by 98 other Wisconsin hospitals that received either a private report on their own quality performance (without the information being made public) or no quality report at all. The study found that the hospitals that participated in public reporting were engaged in significantly more quality-improvement activities in both of the clinical areas assessed than were the hospitals receiving private reporting or no reporting.
A mixed effect on patient outcomes
In contrast, the data on whether public reporting improves patient outcomes have so far been mixed. A 2008 systematic review of the literature identified 11 studies that addressed this issue: five studies found that public reporting had a positive effect on patient outcomes, while six studies demonstrated a negative effect or no effect.8 Unfortunately, the methodological quality of most studies was poor: most were before-and-after comparisons without controls.
One of the positive studies in this review examined the effects of New York State’s pioneering institution of provider-specific CABG mortality reports (provider profiling) in 1989.9 The analysis found that between 1987 and 1992 (during which time provider profiling was instituted), unadjusted 30-day mortality rates following bypass surgery declined to a significantly larger degree among New York Medicare patients (33% reduction) than among Medicare patients nationwide (19% reduction) (P < .001).
In contrast, a time-series study from Cleveland Health Quality Choice (CHQC)—an early and innovative public reporting program—exemplifies a case in which public reporting of hospital performance had no discernible effect.10 The study examined trends in 30-day mortality across a range of conditions over a 6-year period for 30 hospitals in the Cleveland area participating in a public reporting system. It found that the hospitals that started out in the worst-performing groups (based on baseline mortality rates) showed no significant change in mortality over time.
DOES PUBLIC REPORTING AFFECT PATIENT CHOICES?
How a high-profile bypass patient chooses a hospital
When former President Bill Clinton developed chest pain and shortness of breath in 2004, he was seen at a small community hospital in Westchester County, N.Y., and then transferred to New York-Presbyterian Hospital/Columbia University Medical Center for bypass surgery.11 Although one would think President Clinton would have chosen the best hospital for CABG in New York, Presbyterian/Columbia’s risk-adjusted mortality rate for CABG was actually about twice the average for New York hospitals and one of the worst in the state, according to the most recent “report card” for New York hospitals available at the time.12
Why did President Clinton choose the hospital he did? Chances are that he, like most other patients, did not base his decision on publicly reported data. His choice probably was heavily influenced by the normal referral patterns of the community hospital where he was first seen.
Surveys show low patient use of data on quality...
The question raised by President Clinton’s case has been formally studied. In 1996, Schneider and Epstein surveyed patients who had recently undergone CABG in Pennsylvania (where surgeon- and hospital-specific mortality rates for cardiac surgery are publicly available) and found that fewer than 1% of patients said that provider ratings had a moderate or major impact on their choice of provider.13
The Kaiser Family Foundation regularly surveys the public about its knowledge and use of publicly available hospital comparison data. In the latest Kaiser survey, conducted in 2008,14 41% of respondents said they believe there are “big differences” in quality among their local hospitals, yet 59% said they would choose a hospital that is familiar to them rather than a higher-rated facility. These findings may be explained, in part, by a lack of awareness that data on hospital quality are available: only 7% of survey participants said they had seen and used information comparing the quality of hospitals to make health care decisions in the prior year, and only 6% said they had seen and used information comparing physicians.
...But a trend toward greater acceptance
Although consumers’ use of publicly reported quality data remains low, their recognition of the value of such data has grown over time. Kaiser has conducted similar public surveys dating back to 1996, and the period from 1996 to 2008 saw a substantial decrease (from 72% to 59%) in the percentage of Americans who would choose a hospital based on familiarity more than on quality ratings. Similarly, the percentage of Americans who would prefer a surgeon with high quality ratings over a surgeon who has treated friends or family more than doubled from 1996 (20%) to 2008 (47%).14
What effect on market share?
Studies on the effects that public reporting has on hospital market share have been limited.
Schneider and Epstein surveyed cardiologists in Pennsylvania in 1995 and found that 87% of them said the state’s public reporting of surgeon- and hospital-specific mortality rates for CABG had no influence or minimal influence on their referral recommendations.15
Similarly, a review of New York State’s public reporting system for CABG 15 years after its launch found that hospital performance was not associated with a subsequent change in market share, not even among those hospitals with the highest mortality rate in a given year.16 Interestingly, however, this review also showed that surgeons in the bottom performance quartile were four times as likely as other surgeons to leave practice in the year following their poor report, which is one of the most prominent outcomes associated with provider profiling reported to date.
PAY-FOR-PERFORMANCE PROGRAMS
Evidence on the impact of pay-for-performance programs in the hospital setting is even more limited than that for public reporting.
Some evidence has come from the CMS/Premier Hospital Quality Incentive Demonstration, a pay-for-performance collaboration between the Centers for Medicare and Medicaid Services (CMS) and Premier, Inc., a nationwide alliance of hospitals that promotes best practices.17 The demonstration calls for hospitals that rank in the top quintile or decile for performance to receive a 1% or 2% Medicare payment bonus for five clinical focus areas: cardiac surgery, hip and knee surgery, pneumonia, heart failure, and acute MI. Performance ratings are based primarily on process measures as well as a few clinical outcome measures. Results from the first 21 months of the demonstration showed a consistent improvement in the hospitals’ composite quality scores in each of the five clinical areas.17
It is important to recognize, however, that this improvement occurred against the backdrop of broad national adoption of public reporting of hospital quality data, which makes it difficult to tease out how much of the improvement was truly attributable to pay-for-performance, especially in the absence of a control group.
To address this question, my colleagues and I evaluated adherence to quality measures over a 2-year period at 613 hospitals participating in a national public reporting initiative,18 including 207 hospitals that simultaneously took part in the CMS/Premier Hospital Quality Incentive Demonstration’s pay-for-performance program described above. We found that the hospitals participating in both public reporting and the pay-for-performance initiative achieved only modestly greater improvements in quality than did the hospitals engaged solely in public reporting; the difference amounted to only about a 1% improvement in process measures per year.
In another controlled study, Glickman et al compared quality improvement in the management of acute MI between 54 hospitals in a CMS pay-for-performance pilot project and 446 control hospitals without pay-for-performance incentives.19 They found that the pay-for-performance hospitals achieved a statistically significantly greater degree of improvement compared with control hospitals on two of six process-of-care measures (use of aspirin at discharge and smoking-cessation counseling) but not on the composite process-of-care measure. There was no significant difference between the groups in improvements in in-hospital mortality.
Why have the effects of pay-for-performance initiatives so far been so limited? It may be that the bonuses are too small and that public reporting is already effective at stimulating quality improvement, so that the incremental benefit of adding financial incentives is small. In the case of my group’s study,18 another possible factor was that the hospitals’ baseline performance on the quality measures assessed was already high—approaching or exceeding 90% on 5 of the 10 measures—thereby limiting our power to detect differences between the groups.
CONTROVERSIES AND CHALLENGES
Many issues continue to surround public reporting and pay-for-performance programs:
- Are the measures used to evaluate health care systems suitable and evidence-based? Do they truly reflect the quality of care that providers are giving?
- Do the programs encourage “teaching to the test” rather than stimulating real and comprehensive improvement? Do they make the system prone to misuse or overuse of measured services?
- How much of the variation in hospital outcomes can be explained by the current process-of-care measures?
- Should quality be measured by outcomes or processes? Outcomes matter more to patients, but they require risk adjustment to ensure valid comparisons, and risk adjustment can be difficult and expensive to conduct.
- How much is chance a factor in apparent performance differences between hospitals?
- How much is patient selection a factor? Might public reporting lead to “cherry-picking” of low-risk patients and thereby reduce access to care for other patients?
Unidirectional measures can lead to misuse, overuse
In 2003, the Infectious Diseases Society of America updated its guidelines on community-acquired pneumonia to recommend that patients receive antibiotics within 4 hours of hospital admission. This recommendation was widely adopted as an incentive-linked performance measure by CMS and other third-party payers. Kanwar et al studied the impact of this guidelines-based incentive in a pre/post study at one large teaching hospital.20 They found that while significantly more patients received antibiotics in a timely fashion after publication of the guidelines (2005) versus before the guidelines (2003), almost one-third of patients receiving antibiotics in 2005 had normal chest radiographs and thus were not appropriate candidates for therapy. Moreover, significantly fewer patients in 2005 had a final diagnosis of pneumonia at discharge, and there was no difference between the two periods in rates of mortality or ICU transfer. The researchers concluded that linking the quality indicator of early antibiotic use to financial incentives may lead to misdiagnosis of pneumonia and inappropriate antibiotic use.
Of course, antibiotic timing is not the only quality measure subject to overuse or misuse; other measures pose similar risks, including prophylaxis for deep vein thrombosis, glycemic control measures, and target immunization rates.
More-nuanced measures needed
We must also consider how well reported quality measures actually reflect our objectives. For example, an evaluation of 962 hospitals’ performance in managing acute MI found that the publicly reported core process measures for acute MI (beta-blocker and aspirin at admission and discharge, ACE inhibitor at discharge, smoking-cessation counseling, timely reperfusion) together explained only 6% of the variance among the hospitals in risk-adjusted 30-day mortality.21 This underscores how complicated the factors affecting mortality are, and how existing process measures have only begun to scratch the surface.
How much of a role does chance play?
Another issue is the role of chance and our limited power to detect real differences in outcomes, as illustrated by an analysis by Dimick et al of all discharges from a nationally representative sample of nearly 1,000 hospitals.22 The objective was to determine whether the seven operations for which mortality is advocated as a quality indicator by the Agency for Healthcare Research and Quality are performed often enough to reliably identify hospitals with increased mortality rates. The researchers found that only for one of the seven procedures—CABG—is there sufficient caseload over a 3-year period at the majority of US hospitals to accurately detect a mortality rate twice the national average.
Although CMS is highly committed to public reporting, the comparative mortality data available on its Hospital Compare Web site are not very useful for driving consumer choice or motivating hospitals to improve. For example, of the nearly 4,500 US hospitals that reported data on 30-day mortality from MI, only 17 hospitals were considered to be better than the national average and only 7 were considered worse than the national average.4
CASE REVISITED: LESSONS FROM THE UMASS MEMORIAL EXPERIENCE
Returning to our case study, what can the UMass Memorial experience teach us, and how well does it reflect the literature about the usefulness of public reporting?
Did public reporting accelerate quality improvement efforts? Yes. Reporting led to the suspension of cardiac surgery and substantive reorganization, which is consistent with the literature.
Was the mortality reduction typical? No. An optimist’s view would be that the drastic actions spurred by the media coverage had strong effects. A skeptic might say that perhaps UMass Memorial did some “cherry-picking” of patients, or that they got better at coding procedures in a way that reflected more favorably on the hospital.
Were the declines in patient volumes predictable? No. So far, the data suggest that public reporting has its greatest effects on providers rather than on institutions. This may change, however, with the introduction of tiered copayments, whereby patients are asked to pay more if they get their care from lower rated institutions.
Would financial incentives have accelerated improvement? It is too early to tell. The evidence for pay-for-performance programs is limited, and the benefits demonstrated so far have been modest. But in many ways the alternative is worse: our current system of financing and paying for hospital care offers no financial incentives to hospitals for investing in the personnel or systems required to achieve better outcomes—and instead rewards (through supplemental payments) adverse outcomes.
Did prospective patients have a right to know? Despite the limitations of public reporting, one of the most compelling arguments in its favor is that patients at UMass Memorial had the right to know about the program’s outcomes. This alone may ultimately justify the expense and efforts involved. Transparency and accountability are core values of open democratic societies, and US society relies on public reporting in many other realms: the National Highway Traffic Safety Administration publicizes crash test ratings, the Securities and Exchange Commission enforces public reporting by financial institutions, and the Federal Aviation Administration reports on airline safety, timeliness of flights, and lost baggage rates.
FUTURE DIRECTIONS
In the future, we can expect more measurement and reporting of health care factors that patients care most about, such as clinical outcomes and the patient experience. It is likely that public reporting and pay-for-performance programs will address a broader range of conditions and comprise a larger number of measures. CMS has outlined plans to increase the number of publicly reported measures to more than 70 by 2010 and more than 100 by 2011. My hope is that this expansion of data, along with improved data synthesis and presentation, will foster greater use of publicly reported data. Further, the continued evolution of the Web and social networking sites is very likely to enhance public awareness of hospital performance and change the ways in which patients use these data.
DISCUSSION
Question from the audience: I’m concerned about what seems to be a unilateral effort to improve quality. There are many components of health care delivery beyond those you’ve described, including the efforts of patients, insurers, employers, and the government. The reality is that patients don’t plan for illness, insurance companies often deny care, more and more employers are providing less coverage or no coverage, and Medicare is on the road to insolvency. Is the battle for quality winnable when all these other components of delivery are failing?
Dr. Lindenauer: You make good points. But from the standpoint of professionalism, I think we have a compelling duty to constantly strive to improve the quality of care in our hospitals and practices. I have presented strategies for potentially accelerating improvements that providers are trying to make anyway. Public reporting and financial incentives are likely to be with us for a while, and their use is likely to grow. But as you said, they address only part of the problem confronting American health care.
Question from the audience: For the savvy health care consumer, is there one particular Web site for hospital or provider comparisons that you would especially recommend? Do you actually recommend using such Web sites to patients before they undergo certain procedures?
Dr. Lindenauer: I think the Hospital Compare site from the Department of Health and Human Services is the key Web site. The California Hospital Assessment and Reporting Taskforce (CHART) has a good site, and the Commonwealth Fund’s WhyNotTheBest.org is an interesting newcomer.
However, even the most ardent advocates for public reporting wouldn’t say the information available today is sufficient for making decisions. There’s still an important role for getting recommendations from other doctors who are familiar with local hospitals and providers.
I’m optimistic that the changes that are coming to these Web sites will provide a better user experience and make it harder to ignore the results of public reporting. Today we can say, “Hospital A is better at discharge instructions or smoking cessation counseling.” But we all can appreciate how weak those kinds of measures are because their implementation is subject to local interpretations. Once risk-adjusted outcomes and more-meaningful process measures are available, I’d be surprised if more patients weren’t willing to base their decisions on published comparisons.
- Kowalczyk L, Smith S. Hospital halts heart surgeries due to deaths: high rate cited at Worcester facility. The Boston Globe. September 22, 2005.
- Ettinger WH, Hylka SM, Phillips RA, Harrison LH Jr, Cyr JA, Sussman AJ. When things go wrong: the impact of being a statistical outlier in publicly reported coronary artery bypass graft surgery mortality data. Am J Med Qual 2008; 23:90–95.
- Leapfrog hospital quality ratings. The Leapfrog Group Web site. http://www.leapfroggroup.org/cp. Accessed June 10, 2009.
- Hospital Compare: a quality tool provided by Medicare. U.S. Department of Health & Human Services Web site. http://www.hospitalcompare.hhs.gov. Accessed June 10, 2009.
- Why Not the Best (Beta): A Health Care Quality Improvement Resource. The Commonwealth Fund. http://www.WhyNotTheBest.org. Accessed May 6, 2009.
- Hospital-acquired infections in Pennsylvania. Pennsylvania Health Care Cost Containment Council Web site. http://www.phc4.org. Accessed April 6, 2009.
- Hibbard JH, Stockard J, Tusler M. Does publicizing hospital performance stimulate quality improvement efforts? Health Aff (Millwood) 2003; 22:84–94.
- Fung CH, Lim YW, Mattke S, Damberg C, Shekelle PG. Systematic review: the evidence that publishing patient care performance data improves quality of care. Ann Intern Med 2008; 148:111–123.
- Peterson ED, DeLong ER, Jollis JG, et al. The effects of New York’s bypass surgery provider profiling on access to care and patient outcomes in the elderly. J Am Coll Cardiol 1998; 32:993–999.
- Baker DW, Einstadter D, Thomas C, et al. The effect of publicly reporting hospital performance on market share and risk-adjusted mortality at high-mortality hospitals. Med Care 2003; 41:729–740.
- Graylock J. After chest pains, Clinton set to undergo bypass surgery. USA Today. September 3, 2004.
- Adult Cardiac Surgery in New York State, 1999–2001. Albany, NY: New York State Department of Health; April 2004. http://www.health.state.ny.us/nysdoh/heart/pdf/1999-2001_cabg.pdf. Accessed June 10, 2009.
- Schneider EC, Epstein AM. Use of public performance reports: a survey of patients undergoing cardiac surgery. JAMA 1998; 279:1638–1642.
- The Henry J. Kaiser Family Foundation. 2008 Update on Consumers’ Views of Patient Safety and Quality Information: Summary & Chartpack; October 2008. http://www.kff.org/kaiserpolls/upload/7819.pdf. Accessed June 10, 2009.
- Schneider EC, Epstein AM. Influence of cardiac-surgery performance reports on referral practices and access to care: a survey of cardiovascular specialists. N Engl J Med 1996; 335:251–256.
- Jha AK, Epstein AM. The predictive accuracy of the New York State coronary artery bypass surgery report-card system. Health Aff (Millwood) 2006; 25:844–855.
- Remus D. Pay for performance: CMS/Premier Hospital Quality Incentive Demonstration Project—year 1 results, December 2005. PowerPoint presentation available at: http://www.premierinc.com/quality-safety/tools-services/p4p/hqi/results/index.jsp. Accessed June 10, 2009.
- Lindenauer PK, Remus D, Roman S, et al. Public reporting and pay for performance in hospital quality improvement. N Engl J Med 2007; 356:486–496.
- Glickman SW, Ou FS, DeLong ER, et al. Pay for performance, quality of care, and outcomes in acute myocardial infarction. JAMA 2007; 297:2373–2380.
- Kanwar M, Brar N, Khatib R, Fakih MG. Misdiagnosis of community-acquired pneumonia and inappropriate utilization of antibiotics: side effects of the 4-h antibiotic administration rule. Chest 2007; 131:1865–1869.
- Bradley EH, Herrin J, Elbel B, et al. Hospital quality for acute myocardial infarction: correlation among process measures and relationship with short-term mortality. JAMA 2006; 296:72–78.
- Dimick JB, Welch HG, Birkmeyer JD. Surgical mortality as an indicator of hospital quality: the problem with small sample size. JAMA 2004; 292:847–851.
Hospital quality measures and rankings are now widely available to the public online, but is public reporting of this information an effective strategy for improving health care? Using a case study of a hospital that suffered negative publicity as a result of a quality report, this article explores the use of public reporting of performance data and pay-for-performance reimbursement strategies to foster quality improvement in the US health care system.
CASE STUDY: A SURGICAL PROGRAM GETS A BAD REPORT―IN THE HEADLINES
In September 2005, The Boston Globe ran a prominent story reporting that the UMass Memorial Medical Center in Worcester, Mass., was abruptly suspending its elective cardiac surgery program.1 The program’s suspension came after state public health officials presented UMass Memorial with a detailed analysis showing that the hospital’s mortality rate for coronary artery bypass graft surgery (CABG) patients was the highest in the state and almost double the average for Massachusetts hospitals.1
Key personnel from UMass Memorial described the events preceding and following the program’s suspension in a journal article published in 2008.2 In 2002, UMass Memorial hired a new chief of cardiothoracic surgery, who resigned in early 2005. A few months after that resignation, state public health officials alerted the hospital to the abovementioned CABG mortality data (from 2002 and 2003), which they said would soon be reported publicly. UMass Memorial then conducted an internal review of its data from the most recent years (2004 and 2005) and found that its risk-adjusted CABG mortality had actually worsened, at which point the hospital voluntarily suspended its cardiac surgery program.2
More news stories arose about UMass Memorial’s program and its problems. The hospital hired consultants and senior surgeons from around the state and New England to completely review its cardiac surgery program. They concluded that “many essential systems were not in place” and made 68 key recommendations, including a complete overhaul of the hospital’s quality-improvement structure. The prior cardiac surgeons departed.2
The cardiac surgery program resumed after a 6-week hiatus, with day-to-day supervision by two senior cardiac surgeons from a Boston teaching hospital. A nationally recognized cardiac surgeon was brought on as chief of cardiac surgery in January 2006. In the 18 months after the program resumed, risk-adjusted CABG mortality rates declined substantially, but patient volume failed to return to presuspension levels and the hospital reported $22 million in lost revenue in fiscal year 2006 as a result of the suspension.2
This case raises a number of questions that help to frame discussion of the benefits and risks of public reporting of hospital quality measures:
- To what extent does public reporting accelerate quality improvement?
- How typical was the subsequent mortality reduction reported by UMass Memorial—ie, can public reporting be expected to improve outcomes?
- Was the effect on patient volume expected—ie, how much does public reporting affect market share?
- Would a pay-for-performance reimbursement model have accelerated improvement?
- Why do public reporting and pay-for-performance programs remain controversial?
- Do patients have a right to know?
WHAT HAS FUELED THE MOVE TOWARD PUBLIC REPORTING?
Drivers of public reporting
Massachusetts is one of a number of states that publicly report outcomes from cardiac surgery and other procedures and processes of care. Three basic factors have helped drive the development of public reporting (and, in some cases, pay-for-performance) programs:
- National policy imperatives designed to improve quality and safety and to reduce costs
- Cultural factors in society, which include consumerism in health care and the desire for transparency
- The growth of information technology and use of the World Wide Web, which has been a huge enabler of public reporting. Public reporting could be done prior to the Web era but would not have reached such a wide audience had the results been released in a book that had to be ordered from a government printing office.
The rationale for public reporting
In theory, how might public reporting and pay-for-performance programs improve quality? Several different mechanisms or factors are likely to be involved:
- Feedback. The basic premise of the National Surgical Quality Improvement Program, to cite one example, is that peer comparison and performance feedback will stimulate quality improvement.
- Reputation. Hospital personnel fear being embarrassed if data show that they are performing poorly compared with other hospitals. Likewise, in recent years we have seen hospitals with the best quality rankings publicly advertise their performance.
- Market share. Here the premise is that patients will tend to select providers with higher quality rankings and shun those with lower rankings.
- Financial incentives. Pay-for-performance programs link payment or reimbursement directly to the desired outcomes and thereby stimulate quality improvement without working through the abovementioned mechanisms.
Approaches to quality measurement
Public reporting of hospital performance requires selection of an approach to measuring quality of care. Generally speaking, measures of health care quality reflect one of three domains of care:
Structural (or environmental) aspects, such as staffing in the intensive care unit (ICU), surgical volume, or availablity of emergency medical responders. An example of a structure-oriented reporting system is the Leapfrog Group’s online posting of hospital ratings based on surgical volumes for high-risk procedures, the degree of computerized order entry implementation, and the presence or absence of various patient safety practices.3
Processes of care, such as whether beta-blockers are prescribed for all patients after a myocardial infarction (MI), or whether thromboprophylaxis measures are ordered for surgical patients in keeping with guideline recommendations. Examples of process-oriented reporting systems include the US Department of Health and Human Services’ Hospital Compare Web site4 and the Commonwealth Fund’s WhyNotTheBest.org site.5
Outcomes of care, such as rates of mortality or complications, or patient satisfaction rates. An example of an outcomes-oriented reporting system is the annual report of institution-specific hospital-acquired infection rates put out by Pennsylvania6 and most other states.
IS THERE EVIDENCE OF BENEFIT?
A consistent effect in spurring quality-improvement efforts
Nearly a dozen published studies have evaluated whether public reporting stimulates quality-improvement activities, and the results have shown fairly consistently that it does. A 2003 study by Hibbard et al is representative of the results.7 This survey-based investigation measured the number of quality-improvement activities in cardiac and obstetric care undertaken by 24 Wisconsin hospitals that were included in an existing public reporting system compared with the number undertaken by 98 other Wisconsin hospitals that received either a private report on their own quality performance (without the information being made public) or no quality report at all. The study found that the hospitals that participated in public reporting were engaged in significantly more quality-improvement activities in both of the clinical areas assessed than were the hospitals receiving private reporting or no reporting.
A mixed effect on patient outcomes
In contrast, the data on whether public reporting improves patient outcomes have so far been mixed. A 2008 systematic review of the literature identified 11 studies that addressed this issue: five studies found that public reporting had a positive effect on patient outcomes, while six studies demonstrated a negative effect or no effect.8 Unfortunately, the methodological quality of most studies was poor: most were before-and-after comparisons without controls.
One of the positive studies in this review examined the effects of New York State’s pioneering institution of provider-specific CABG mortality reports (provider profiling) in 1989.9 The analysis found that between 1987 and 1992 (during which time provider profiling was instituted), unadjusted 30-day mortality rates following bypass surgery declined to a significantly larger degree among New York Medicare patients (33% reduction) than among Medicare patients nationwide (19% reduction) (P < .001).
In contrast, a time-series study from Cleveland Health Quality Choice (CHQC)—an early and innovative public reporting program—exemplifies a case in which public reporting of hospital performance had no discernible effect.10 The study examined trends in 30-day mortality across a range of conditions over a 6-year period for 30 hospitals in the Cleveland area participating in a public reporting system. It found that the hospitals that started out in the worst-performing groups (based on baseline mortality rates) showed no significant change in mortality over time.
DOES PUBLIC REPORTING AFFECT PATIENT CHOICES?
How a high-profile bypass patient chooses a hospital
When former President Bill Clinton developed chest pain and shortness of breath in 2004, he was seen at a small community hospital in Westchester County, N.Y., and then transferred to New York-Presbyterian Hospital/Columbia University Medical Center for bypass surgery.11 Although one would think President Clinton would have chosen the best hospital for CABG in New York, Presbyterian/Columbia’s risk-adjusted mortality rate for CABG was actually about twice the average for New York hospitals and one of the worst in the state, according to the most recent “report card” for New York hospitals available at the time.12
Why did President Clinton choose the hospital he did? Chances are that he, like most other patients, did not base his decision on publicly reported data. His choice probably was heavily influenced by the normal referral patterns of the community hospital where he was first seen.
Surveys show low patient use of data on quality...
The question raised by President Clinton’s case has been formally studied. In 1996, Schneider and Epstein surveyed patients who had recently undergone CABG in Pennsylvania (where surgeon- and hospital-specific mortality rates for cardiac surgery are publicly available) and found that fewer than 1% of patients said that provider ratings had a moderate or major impact on their choice of provider.13
The Kaiser Family Foundation regularly surveys the public about its knowledge and use of publicly available hospital comparison data. In the latest Kaiser survey, conducted in 2008,14 41% of respondents said they believe there are “big differences” in quality among their local hospitals, yet 59% said they would choose a hospital that is familiar to them rather than a higher-rated facility. These findings may be explained, in part, by a lack of awareness that data on hospital quality are available: only 7% of survey participants said they had seen and used information comparing the quality of hospitals to make health care decisions in the prior year, and only 6% said they had seen and used information comparing physicians.
...But a trend toward greater acceptance
Although consumers’ use of publicly reported quality data remains low, their recognition of the value of such data has grown over time. Kaiser has conducted similar public surveys dating back to 1996, and the period from 1996 to 2008 saw a substantial decrease (from 72% to 59%) in the percentage of Americans who would choose a hospital based on familiarity more than on quality ratings. Similarly, the percentage of Americans who would prefer a surgeon with high quality ratings over a surgeon who has treated friends or family more than doubled from 1996 (20%) to 2008 (47%).14
What effect on market share?
Studies on the effects that public reporting has on hospital market share have been limited.
Schneider and Epstein surveyed cardiologists in Pennsylvania in 1995 and found that 87% of them said the state’s public reporting of surgeon- and hospital-specific mortality rates for CABG had no influence or minimal influence on their referral recommendations.15
Similarly, a review of New York State’s public reporting system for CABG 15 years after its launch found that hospital performance was not associated with a subsequent change in market share, not even among those hospitals with the highest mortality rate in a given year.16 Interestingly, however, this review also showed that surgeons in the bottom performance quartile were four times as likely as other surgeons to leave practice in the year following their poor report, which is one of the most prominent outcomes associated with provider profiling reported to date.
PAY-FOR-PERFORMANCE PROGRAMS
Evidence on the impact of pay-for-performance programs in the hospital setting is even more limited than that for public reporting.
Some evidence has come from the CMS/Premier Hospital Quality Incentive Demonstration, a pay-for-performance collaboration between the Centers for Medicare and Medicaid Services (CMS) and Premier, Inc., a nationwide alliance of hospitals that promotes best practices.17 The demonstration calls for hospitals that rank in the top quintile or decile for performance to receive a 1% or 2% Medicare payment bonus for five clinical focus areas: cardiac surgery, hip and knee surgery, pneumonia, heart failure, and acute MI. Performance ratings are based primarily on process measures as well as a few clinical outcome measures. Results from the first 21 months of the demonstration showed a consistent improvement in the hospitals’ composite quality scores in each of the five clinical areas.17
It is important to recognize, however, that this improvement occurred against the backdrop of broad national adoption of public reporting of hospital quality data, which makes it difficult to tease out how much of the improvement was truly attributable to pay-for-performance, especially in the absence of a control group.
To address this question, my colleagues and I evaluated adherence to quality measures over a 2-year period at 613 hospitals participating in a national public reporting initiative,18 including 207 hospitals that simultaneously took part in the CMS/Premier Hospital Quality Incentive Demonstration’s pay-for-performance program described above. We found that the hospitals participating in both public reporting and the pay-for-performance initiative achieved only modestly greater improvements in quality than did the hospitals engaged solely in public reporting; the difference amounted to only about a 1% improvement in process measures per year.
In another controlled study, Glickman et al compared quality improvement in the management of acute MI between 54 hospitals in a CMS pay-for-performance pilot project and 446 control hospitals without pay-for-performance incentives.19 They found that the pay-for-performance hospitals achieved a statistically significantly greater degree of improvement compared with control hospitals on two of six process-of-care measures (use of aspirin at discharge and smoking-cessation counseling) but not on the composite process-of-care measure. There was no significant difference between the groups in improvements in in-hospital mortality.
Why have the effects of pay-for-performance initiatives so far been so limited? It may be that the bonuses are too small and that public reporting is already effective at stimulating quality improvement, so that the incremental benefit of adding financial incentives is small. In the case of my group’s study,18 another possible factor was that the hospitals’ baseline performance on the quality measures assessed was already high—approaching or exceeding 90% on 5 of the 10 measures—thereby limiting our power to detect differences between the groups.
CONTROVERSIES AND CHALLENGES
Many issues continue to surround public reporting and pay-for-performance programs:
- Are the measures used to evaluate health care systems suitable and evidence-based? Do they truly reflect the quality of care that providers are giving?
- Do the programs encourage “teaching to the test” rather than stimulating real and comprehensive improvement? Do they make the system prone to misuse or overuse of measured services?
- How much of the variation in hospital outcomes can be explained by the current process-of-care measures?
- Should quality be measured by outcomes or processes? Outcomes matter more to patients, but they require risk adjustment to ensure valid comparisons, and risk adjustment can be difficult and expensive to conduct.
- How much is chance a factor in apparent performance differences between hospitals?
- How much is patient selection a factor? Might public reporting lead to “cherry-picking” of low-risk patients and thereby reduce access to care for other patients?
Unidirectional measures can lead to misuse, overuse
In 2003, the Infectious Diseases Society of America updated its guidelines on community-acquired pneumonia to recommend that patients receive antibiotics within 4 hours of hospital admission. This recommendation was widely adopted as an incentive-linked performance measure by CMS and other third-party payers. Kanwar et al studied the impact of this guidelines-based incentive in a pre/post study at one large teaching hospital.20 They found that while significantly more patients received antibiotics in a timely fashion after publication of the guidelines (2005) versus before the guidelines (2003), almost one-third of patients receiving antibiotics in 2005 had normal chest radiographs and thus were not appropriate candidates for therapy. Moreover, significantly fewer patients in 2005 had a final diagnosis of pneumonia at discharge, and there was no difference between the two periods in rates of mortality or ICU transfer. The researchers concluded that linking the quality indicator of early antibiotic use to financial incentives may lead to misdiagnosis of pneumonia and inappropriate antibiotic use.
Of course, antibiotic timing is not the only quality measure subject to overuse or misuse; other measures pose similar risks, including prophylaxis for deep vein thrombosis, glycemic control measures, and target immunization rates.
More-nuanced measures needed
We must also consider how well reported quality measures actually reflect our objectives. For example, an evaluation of 962 hospitals’ performance in managing acute MI found that the publicly reported core process measures for acute MI (beta-blocker and aspirin at admission and discharge, ACE inhibitor at discharge, smoking-cessation counseling, timely reperfusion) together explained only 6% of the variance among the hospitals in risk-adjusted 30-day mortality.21 This underscores how complicated the factors affecting mortality are, and how existing process measures have only begun to scratch the surface.
How much of a role does chance play?
Another issue is the role of chance and our limited power to detect real differences in outcomes, as illustrated by an analysis by Dimick et al of all discharges from a nationally representative sample of nearly 1,000 hospitals.22 The objective was to determine whether the seven operations for which mortality is advocated as a quality indicator by the Agency for Healthcare Research and Quality are performed often enough to reliably identify hospitals with increased mortality rates. The researchers found that only for one of the seven procedures—CABG—is there sufficient caseload over a 3-year period at the majority of US hospitals to accurately detect a mortality rate twice the national average.
Although CMS is highly committed to public reporting, the comparative mortality data available on its Hospital Compare Web site are not very useful for driving consumer choice or motivating hospitals to improve. For example, of the nearly 4,500 US hospitals that reported data on 30-day mortality from MI, only 17 hospitals were considered to be better than the national average and only 7 were considered worse than the national average.4
CASE REVISITED: LESSONS FROM THE UMASS MEMORIAL EXPERIENCE
Returning to our case study, what can the UMass Memorial experience teach us, and how well does it reflect the literature about the usefulness of public reporting?
Did public reporting accelerate quality improvement efforts? Yes. Reporting led to the suspension of cardiac surgery and substantive reorganization, which is consistent with the literature.
Was the mortality reduction typical? No. An optimist’s view would be that the drastic actions spurred by the media coverage had strong effects. A skeptic might say that perhaps UMass Memorial did some “cherry-picking” of patients, or that they got better at coding procedures in a way that reflected more favorably on the hospital.
Were the declines in patient volumes predictable? No. So far, the data suggest that public reporting has its greatest effects on providers rather than on institutions. This may change, however, with the introduction of tiered copayments, whereby patients are asked to pay more if they get their care from lower rated institutions.
Would financial incentives have accelerated improvement? It is too early to tell. The evidence for pay-for-performance programs is limited, and the benefits demonstrated so far have been modest. But in many ways the alternative is worse: our current system of financing and paying for hospital care offers no financial incentives to hospitals for investing in the personnel or systems required to achieve better outcomes—and instead rewards (through supplemental payments) adverse outcomes.
Did prospective patients have a right to know? Despite the limitations of public reporting, one of the most compelling arguments in its favor is that patients at UMass Memorial had the right to know about the program’s outcomes. This alone may ultimately justify the expense and efforts involved. Transparency and accountability are core values of open democratic societies, and US society relies on public reporting in many other realms: the National Highway Traffic Safety Administration publicizes crash test ratings, the Securities and Exchange Commission enforces public reporting by financial institutions, and the Federal Aviation Administration reports on airline safety, timeliness of flights, and lost baggage rates.
FUTURE DIRECTIONS
In the future, we can expect more measurement and reporting of health care factors that patients care most about, such as clinical outcomes and the patient experience. It is likely that public reporting and pay-for-performance programs will address a broader range of conditions and comprise a larger number of measures. CMS has outlined plans to increase the number of publicly reported measures to more than 70 by 2010 and more than 100 by 2011. My hope is that this expansion of data, along with improved data synthesis and presentation, will foster greater use of publicly reported data. Further, the continued evolution of the Web and social networking sites is very likely to enhance public awareness of hospital performance and change the ways in which patients use these data.
DISCUSSION
Question from the audience: I’m concerned about what seems to be a unilateral effort to improve quality. There are many components of health care delivery beyond those you’ve described, including the efforts of patients, insurers, employers, and the government. The reality is that patients don’t plan for illness, insurance companies often deny care, more and more employers are providing less coverage or no coverage, and Medicare is on the road to insolvency. Is the battle for quality winnable when all these other components of delivery are failing?
Dr. Lindenauer: You make good points. But from the standpoint of professionalism, I think we have a compelling duty to constantly strive to improve the quality of care in our hospitals and practices. I have presented strategies for potentially accelerating improvements that providers are trying to make anyway. Public reporting and financial incentives are likely to be with us for a while, and their use is likely to grow. But as you said, they address only part of the problem confronting American health care.
Question from the audience: For the savvy health care consumer, is there one particular Web site for hospital or provider comparisons that you would especially recommend? Do you actually recommend using such Web sites to patients before they undergo certain procedures?
Dr. Lindenauer: I think the Hospital Compare site from the Department of Health and Human Services is the key Web site. The California Hospital Assessment and Reporting Taskforce (CHART) has a good site, and the Commonwealth Fund’s WhyNotTheBest.org is an interesting newcomer.
However, even the most ardent advocates for public reporting wouldn’t say the information available today is sufficient for making decisions. There’s still an important role for getting recommendations from other doctors who are familiar with local hospitals and providers.
I’m optimistic that the changes that are coming to these Web sites will provide a better user experience and make it harder to ignore the results of public reporting. Today we can say, “Hospital A is better at discharge instructions or smoking cessation counseling.” But we all can appreciate how weak those kinds of measures are because their implementation is subject to local interpretations. Once risk-adjusted outcomes and more-meaningful process measures are available, I’d be surprised if more patients weren’t willing to base their decisions on published comparisons.
Hospital quality measures and rankings are now widely available to the public online, but is public reporting of this information an effective strategy for improving health care? Using a case study of a hospital that suffered negative publicity as a result of a quality report, this article explores the use of public reporting of performance data and pay-for-performance reimbursement strategies to foster quality improvement in the US health care system.
CASE STUDY: A SURGICAL PROGRAM GETS A BAD REPORT―IN THE HEADLINES
In September 2005, The Boston Globe ran a prominent story reporting that the UMass Memorial Medical Center in Worcester, Mass., was abruptly suspending its elective cardiac surgery program.1 The program’s suspension came after state public health officials presented UMass Memorial with a detailed analysis showing that the hospital’s mortality rate for coronary artery bypass graft surgery (CABG) patients was the highest in the state and almost double the average for Massachusetts hospitals.1
Key personnel from UMass Memorial described the events preceding and following the program’s suspension in a journal article published in 2008.2 In 2002, UMass Memorial hired a new chief of cardiothoracic surgery, who resigned in early 2005. A few months after that resignation, state public health officials alerted the hospital to the abovementioned CABG mortality data (from 2002 and 2003), which they said would soon be reported publicly. UMass Memorial then conducted an internal review of its data from the most recent years (2004 and 2005) and found that its risk-adjusted CABG mortality had actually worsened, at which point the hospital voluntarily suspended its cardiac surgery program.2
More news stories arose about UMass Memorial’s program and its problems. The hospital hired consultants and senior surgeons from around the state and New England to completely review its cardiac surgery program. They concluded that “many essential systems were not in place” and made 68 key recommendations, including a complete overhaul of the hospital’s quality-improvement structure. The prior cardiac surgeons departed.2
The cardiac surgery program resumed after a 6-week hiatus, with day-to-day supervision by two senior cardiac surgeons from a Boston teaching hospital. A nationally recognized cardiac surgeon was brought on as chief of cardiac surgery in January 2006. In the 18 months after the program resumed, risk-adjusted CABG mortality rates declined substantially, but patient volume failed to return to presuspension levels and the hospital reported $22 million in lost revenue in fiscal year 2006 as a result of the suspension.2
This case raises a number of questions that help to frame discussion of the benefits and risks of public reporting of hospital quality measures:
- To what extent does public reporting accelerate quality improvement?
- How typical was the subsequent mortality reduction reported by UMass Memorial—ie, can public reporting be expected to improve outcomes?
- Was the effect on patient volume expected—ie, how much does public reporting affect market share?
- Would a pay-for-performance reimbursement model have accelerated improvement?
- Why do public reporting and pay-for-performance programs remain controversial?
- Do patients have a right to know?
WHAT HAS FUELED THE MOVE TOWARD PUBLIC REPORTING?
Drivers of public reporting
Massachusetts is one of a number of states that publicly report outcomes from cardiac surgery and other procedures and processes of care. Three basic factors have helped drive the development of public reporting (and, in some cases, pay-for-performance) programs:
- National policy imperatives designed to improve quality and safety and to reduce costs
- Cultural factors in society, which include consumerism in health care and the desire for transparency
- The growth of information technology and use of the World Wide Web, which has been a huge enabler of public reporting. Public reporting could be done prior to the Web era but would not have reached such a wide audience had the results been released in a book that had to be ordered from a government printing office.
The rationale for public reporting
In theory, how might public reporting and pay-for-performance programs improve quality? Several different mechanisms or factors are likely to be involved:
- Feedback. The basic premise of the National Surgical Quality Improvement Program, to cite one example, is that peer comparison and performance feedback will stimulate quality improvement.
- Reputation. Hospital personnel fear being embarrassed if data show that they are performing poorly compared with other hospitals. Likewise, in recent years we have seen hospitals with the best quality rankings publicly advertise their performance.
- Market share. Here the premise is that patients will tend to select providers with higher quality rankings and shun those with lower rankings.
- Financial incentives. Pay-for-performance programs link payment or reimbursement directly to the desired outcomes and thereby stimulate quality improvement without working through the abovementioned mechanisms.
Approaches to quality measurement
Public reporting of hospital performance requires selection of an approach to measuring quality of care. Generally speaking, measures of health care quality reflect one of three domains of care:
Structural (or environmental) aspects, such as staffing in the intensive care unit (ICU), surgical volume, or availablity of emergency medical responders. An example of a structure-oriented reporting system is the Leapfrog Group’s online posting of hospital ratings based on surgical volumes for high-risk procedures, the degree of computerized order entry implementation, and the presence or absence of various patient safety practices.3
Processes of care, such as whether beta-blockers are prescribed for all patients after a myocardial infarction (MI), or whether thromboprophylaxis measures are ordered for surgical patients in keeping with guideline recommendations. Examples of process-oriented reporting systems include the US Department of Health and Human Services’ Hospital Compare Web site4 and the Commonwealth Fund’s WhyNotTheBest.org site.5
Outcomes of care, such as rates of mortality or complications, or patient satisfaction rates. An example of an outcomes-oriented reporting system is the annual report of institution-specific hospital-acquired infection rates put out by Pennsylvania6 and most other states.
IS THERE EVIDENCE OF BENEFIT?
A consistent effect in spurring quality-improvement efforts
Nearly a dozen published studies have evaluated whether public reporting stimulates quality-improvement activities, and the results have shown fairly consistently that it does. A 2003 study by Hibbard et al is representative of the results.7 This survey-based investigation measured the number of quality-improvement activities in cardiac and obstetric care undertaken by 24 Wisconsin hospitals that were included in an existing public reporting system compared with the number undertaken by 98 other Wisconsin hospitals that received either a private report on their own quality performance (without the information being made public) or no quality report at all. The study found that the hospitals that participated in public reporting were engaged in significantly more quality-improvement activities in both of the clinical areas assessed than were the hospitals receiving private reporting or no reporting.
A mixed effect on patient outcomes
In contrast, the data on whether public reporting improves patient outcomes have so far been mixed. A 2008 systematic review of the literature identified 11 studies that addressed this issue: five studies found that public reporting had a positive effect on patient outcomes, while six studies demonstrated a negative effect or no effect.8 Unfortunately, the methodological quality of most studies was poor: most were before-and-after comparisons without controls.
One of the positive studies in this review examined the effects of New York State’s pioneering institution of provider-specific CABG mortality reports (provider profiling) in 1989.9 The analysis found that between 1987 and 1992 (during which time provider profiling was instituted), unadjusted 30-day mortality rates following bypass surgery declined to a significantly larger degree among New York Medicare patients (33% reduction) than among Medicare patients nationwide (19% reduction) (P < .001).
In contrast, a time-series study from Cleveland Health Quality Choice (CHQC)—an early and innovative public reporting program—exemplifies a case in which public reporting of hospital performance had no discernible effect.10 The study examined trends in 30-day mortality across a range of conditions over a 6-year period for 30 hospitals in the Cleveland area participating in a public reporting system. It found that the hospitals that started out in the worst-performing groups (based on baseline mortality rates) showed no significant change in mortality over time.
DOES PUBLIC REPORTING AFFECT PATIENT CHOICES?
How a high-profile bypass patient chooses a hospital
When former President Bill Clinton developed chest pain and shortness of breath in 2004, he was seen at a small community hospital in Westchester County, N.Y., and then transferred to New York-Presbyterian Hospital/Columbia University Medical Center for bypass surgery.11 Although one would think President Clinton would have chosen the best hospital for CABG in New York, Presbyterian/Columbia’s risk-adjusted mortality rate for CABG was actually about twice the average for New York hospitals and one of the worst in the state, according to the most recent “report card” for New York hospitals available at the time.12
Why did President Clinton choose the hospital he did? Chances are that he, like most other patients, did not base his decision on publicly reported data. His choice probably was heavily influenced by the normal referral patterns of the community hospital where he was first seen.
Surveys show low patient use of data on quality...
The question raised by President Clinton’s case has been formally studied. In 1996, Schneider and Epstein surveyed patients who had recently undergone CABG in Pennsylvania (where surgeon- and hospital-specific mortality rates for cardiac surgery are publicly available) and found that fewer than 1% of patients said that provider ratings had a moderate or major impact on their choice of provider.13
The Kaiser Family Foundation regularly surveys the public about its knowledge and use of publicly available hospital comparison data. In the latest Kaiser survey, conducted in 2008,14 41% of respondents said they believe there are “big differences” in quality among their local hospitals, yet 59% said they would choose a hospital that is familiar to them rather than a higher-rated facility. These findings may be explained, in part, by a lack of awareness that data on hospital quality are available: only 7% of survey participants said they had seen and used information comparing the quality of hospitals to make health care decisions in the prior year, and only 6% said they had seen and used information comparing physicians.
...But a trend toward greater acceptance
Although consumers’ use of publicly reported quality data remains low, their recognition of the value of such data has grown over time. Kaiser has conducted similar public surveys dating back to 1996, and the period from 1996 to 2008 saw a substantial decrease (from 72% to 59%) in the percentage of Americans who would choose a hospital based on familiarity more than on quality ratings. Similarly, the percentage of Americans who would prefer a surgeon with high quality ratings over a surgeon who has treated friends or family more than doubled from 1996 (20%) to 2008 (47%).14
What effect on market share?
Studies on the effects that public reporting has on hospital market share have been limited.
Schneider and Epstein surveyed cardiologists in Pennsylvania in 1995 and found that 87% of them said the state’s public reporting of surgeon- and hospital-specific mortality rates for CABG had no influence or minimal influence on their referral recommendations.15
Similarly, a review of New York State’s public reporting system for CABG 15 years after its launch found that hospital performance was not associated with a subsequent change in market share, not even among those hospitals with the highest mortality rate in a given year.16 Interestingly, however, this review also showed that surgeons in the bottom performance quartile were four times as likely as other surgeons to leave practice in the year following their poor report, which is one of the most prominent outcomes associated with provider profiling reported to date.
PAY-FOR-PERFORMANCE PROGRAMS
Evidence on the impact of pay-for-performance programs in the hospital setting is even more limited than that for public reporting.
Some evidence has come from the CMS/Premier Hospital Quality Incentive Demonstration, a pay-for-performance collaboration between the Centers for Medicare and Medicaid Services (CMS) and Premier, Inc., a nationwide alliance of hospitals that promotes best practices.17 The demonstration calls for hospitals that rank in the top quintile or decile for performance to receive a 1% or 2% Medicare payment bonus for five clinical focus areas: cardiac surgery, hip and knee surgery, pneumonia, heart failure, and acute MI. Performance ratings are based primarily on process measures as well as a few clinical outcome measures. Results from the first 21 months of the demonstration showed a consistent improvement in the hospitals’ composite quality scores in each of the five clinical areas.17
It is important to recognize, however, that this improvement occurred against the backdrop of broad national adoption of public reporting of hospital quality data, which makes it difficult to tease out how much of the improvement was truly attributable to pay-for-performance, especially in the absence of a control group.
To address this question, my colleagues and I evaluated adherence to quality measures over a 2-year period at 613 hospitals participating in a national public reporting initiative,18 including 207 hospitals that simultaneously took part in the CMS/Premier Hospital Quality Incentive Demonstration’s pay-for-performance program described above. We found that the hospitals participating in both public reporting and the pay-for-performance initiative achieved only modestly greater improvements in quality than did the hospitals engaged solely in public reporting; the difference amounted to only about a 1% improvement in process measures per year.
In another controlled study, Glickman et al compared quality improvement in the management of acute MI between 54 hospitals in a CMS pay-for-performance pilot project and 446 control hospitals without pay-for-performance incentives.19 They found that the pay-for-performance hospitals achieved a statistically significantly greater degree of improvement compared with control hospitals on two of six process-of-care measures (use of aspirin at discharge and smoking-cessation counseling) but not on the composite process-of-care measure. There was no significant difference between the groups in improvements in in-hospital mortality.
Why have the effects of pay-for-performance initiatives so far been so limited? It may be that the bonuses are too small and that public reporting is already effective at stimulating quality improvement, so that the incremental benefit of adding financial incentives is small. In the case of my group’s study,18 another possible factor was that the hospitals’ baseline performance on the quality measures assessed was already high—approaching or exceeding 90% on 5 of the 10 measures—thereby limiting our power to detect differences between the groups.
CONTROVERSIES AND CHALLENGES
Many issues continue to surround public reporting and pay-for-performance programs:
- Are the measures used to evaluate health care systems suitable and evidence-based? Do they truly reflect the quality of care that providers are giving?
- Do the programs encourage “teaching to the test” rather than stimulating real and comprehensive improvement? Do they make the system prone to misuse or overuse of measured services?
- How much of the variation in hospital outcomes can be explained by the current process-of-care measures?
- Should quality be measured by outcomes or processes? Outcomes matter more to patients, but they require risk adjustment to ensure valid comparisons, and risk adjustment can be difficult and expensive to conduct.
- How much is chance a factor in apparent performance differences between hospitals?
- How much is patient selection a factor? Might public reporting lead to “cherry-picking” of low-risk patients and thereby reduce access to care for other patients?
Unidirectional measures can lead to misuse, overuse
In 2003, the Infectious Diseases Society of America updated its guidelines on community-acquired pneumonia to recommend that patients receive antibiotics within 4 hours of hospital admission. This recommendation was widely adopted as an incentive-linked performance measure by CMS and other third-party payers. Kanwar et al studied the impact of this guidelines-based incentive in a pre/post study at one large teaching hospital.20 They found that while significantly more patients received antibiotics in a timely fashion after publication of the guidelines (2005) versus before the guidelines (2003), almost one-third of patients receiving antibiotics in 2005 had normal chest radiographs and thus were not appropriate candidates for therapy. Moreover, significantly fewer patients in 2005 had a final diagnosis of pneumonia at discharge, and there was no difference between the two periods in rates of mortality or ICU transfer. The researchers concluded that linking the quality indicator of early antibiotic use to financial incentives may lead to misdiagnosis of pneumonia and inappropriate antibiotic use.
Of course, antibiotic timing is not the only quality measure subject to overuse or misuse; other measures pose similar risks, including prophylaxis for deep vein thrombosis, glycemic control measures, and target immunization rates.
More-nuanced measures needed
We must also consider how well reported quality measures actually reflect our objectives. For example, an evaluation of 962 hospitals’ performance in managing acute MI found that the publicly reported core process measures for acute MI (beta-blocker and aspirin at admission and discharge, ACE inhibitor at discharge, smoking-cessation counseling, timely reperfusion) together explained only 6% of the variance among the hospitals in risk-adjusted 30-day mortality.21 This underscores how complicated the factors affecting mortality are, and how existing process measures have only begun to scratch the surface.
How much of a role does chance play?
Another issue is the role of chance and our limited power to detect real differences in outcomes, as illustrated by an analysis by Dimick et al of all discharges from a nationally representative sample of nearly 1,000 hospitals.22 The objective was to determine whether the seven operations for which mortality is advocated as a quality indicator by the Agency for Healthcare Research and Quality are performed often enough to reliably identify hospitals with increased mortality rates. The researchers found that only for one of the seven procedures—CABG—is there sufficient caseload over a 3-year period at the majority of US hospitals to accurately detect a mortality rate twice the national average.
Although CMS is highly committed to public reporting, the comparative mortality data available on its Hospital Compare Web site are not very useful for driving consumer choice or motivating hospitals to improve. For example, of the nearly 4,500 US hospitals that reported data on 30-day mortality from MI, only 17 hospitals were considered to be better than the national average and only 7 were considered worse than the national average.4
CASE REVISITED: LESSONS FROM THE UMASS MEMORIAL EXPERIENCE
Returning to our case study, what can the UMass Memorial experience teach us, and how well does it reflect the literature about the usefulness of public reporting?
Did public reporting accelerate quality improvement efforts? Yes. Reporting led to the suspension of cardiac surgery and substantive reorganization, which is consistent with the literature.
Was the mortality reduction typical? No. An optimist’s view would be that the drastic actions spurred by the media coverage had strong effects. A skeptic might say that perhaps UMass Memorial did some “cherry-picking” of patients, or that they got better at coding procedures in a way that reflected more favorably on the hospital.
Were the declines in patient volumes predictable? No. So far, the data suggest that public reporting has its greatest effects on providers rather than on institutions. This may change, however, with the introduction of tiered copayments, whereby patients are asked to pay more if they get their care from lower rated institutions.
Would financial incentives have accelerated improvement? It is too early to tell. The evidence for pay-for-performance programs is limited, and the benefits demonstrated so far have been modest. But in many ways the alternative is worse: our current system of financing and paying for hospital care offers no financial incentives to hospitals for investing in the personnel or systems required to achieve better outcomes—and instead rewards (through supplemental payments) adverse outcomes.
Did prospective patients have a right to know? Despite the limitations of public reporting, one of the most compelling arguments in its favor is that patients at UMass Memorial had the right to know about the program’s outcomes. This alone may ultimately justify the expense and efforts involved. Transparency and accountability are core values of open democratic societies, and US society relies on public reporting in many other realms: the National Highway Traffic Safety Administration publicizes crash test ratings, the Securities and Exchange Commission enforces public reporting by financial institutions, and the Federal Aviation Administration reports on airline safety, timeliness of flights, and lost baggage rates.
FUTURE DIRECTIONS
In the future, we can expect more measurement and reporting of health care factors that patients care most about, such as clinical outcomes and the patient experience. It is likely that public reporting and pay-for-performance programs will address a broader range of conditions and comprise a larger number of measures. CMS has outlined plans to increase the number of publicly reported measures to more than 70 by 2010 and more than 100 by 2011. My hope is that this expansion of data, along with improved data synthesis and presentation, will foster greater use of publicly reported data. Further, the continued evolution of the Web and social networking sites is very likely to enhance public awareness of hospital performance and change the ways in which patients use these data.
DISCUSSION
Question from the audience: I’m concerned about what seems to be a unilateral effort to improve quality. There are many components of health care delivery beyond those you’ve described, including the efforts of patients, insurers, employers, and the government. The reality is that patients don’t plan for illness, insurance companies often deny care, more and more employers are providing less coverage or no coverage, and Medicare is on the road to insolvency. Is the battle for quality winnable when all these other components of delivery are failing?
Dr. Lindenauer: You make good points. But from the standpoint of professionalism, I think we have a compelling duty to constantly strive to improve the quality of care in our hospitals and practices. I have presented strategies for potentially accelerating improvements that providers are trying to make anyway. Public reporting and financial incentives are likely to be with us for a while, and their use is likely to grow. But as you said, they address only part of the problem confronting American health care.
Question from the audience: For the savvy health care consumer, is there one particular Web site for hospital or provider comparisons that you would especially recommend? Do you actually recommend using such Web sites to patients before they undergo certain procedures?
Dr. Lindenauer: I think the Hospital Compare site from the Department of Health and Human Services is the key Web site. The California Hospital Assessment and Reporting Taskforce (CHART) has a good site, and the Commonwealth Fund’s WhyNotTheBest.org is an interesting newcomer.
However, even the most ardent advocates for public reporting wouldn’t say the information available today is sufficient for making decisions. There’s still an important role for getting recommendations from other doctors who are familiar with local hospitals and providers.
I’m optimistic that the changes that are coming to these Web sites will provide a better user experience and make it harder to ignore the results of public reporting. Today we can say, “Hospital A is better at discharge instructions or smoking cessation counseling.” But we all can appreciate how weak those kinds of measures are because their implementation is subject to local interpretations. Once risk-adjusted outcomes and more-meaningful process measures are available, I’d be surprised if more patients weren’t willing to base their decisions on published comparisons.
- Kowalczyk L, Smith S. Hospital halts heart surgeries due to deaths: high rate cited at Worcester facility. The Boston Globe. September 22, 2005.
- Ettinger WH, Hylka SM, Phillips RA, Harrison LH Jr, Cyr JA, Sussman AJ. When things go wrong: the impact of being a statistical outlier in publicly reported coronary artery bypass graft surgery mortality data. Am J Med Qual 2008; 23:90–95.
- Leapfrog hospital quality ratings. The Leapfrog Group Web site. http://www.leapfroggroup.org/cp. Accessed June 10, 2009.
- Hospital Compare: a quality tool provided by Medicare. U.S. Department of Health & Human Services Web site. http://www.hospitalcompare.hhs.gov. Accessed June 10, 2009.
- Why Not the Best (Beta): A Health Care Quality Improvement Resource. The Commonwealth Fund. http://www.WhyNotTheBest.org. Accessed May 6, 2009.
- Hospital-acquired infections in Pennsylvania. Pennsylvania Health Care Cost Containment Council Web site. http://www.phc4.org. Accessed April 6, 2009.
- Hibbard JH, Stockard J, Tusler M. Does publicizing hospital performance stimulate quality improvement efforts? Health Aff (Millwood) 2003; 22:84–94.
- Fung CH, Lim YW, Mattke S, Damberg C, Shekelle PG. Systematic review: the evidence that publishing patient care performance data improves quality of care. Ann Intern Med 2008; 148:111–123.
- Peterson ED, DeLong ER, Jollis JG, et al. The effects of New York’s bypass surgery provider profiling on access to care and patient outcomes in the elderly. J Am Coll Cardiol 1998; 32:993–999.
- Baker DW, Einstadter D, Thomas C, et al. The effect of publicly reporting hospital performance on market share and risk-adjusted mortality at high-mortality hospitals. Med Care 2003; 41:729–740.
- Graylock J. After chest pains, Clinton set to undergo bypass surgery. USA Today. September 3, 2004.
- Adult Cardiac Surgery in New York State, 1999–2001. Albany, NY: New York State Department of Health; April 2004. http://www.health.state.ny.us/nysdoh/heart/pdf/1999-2001_cabg.pdf. Accessed June 10, 2009.
- Schneider EC, Epstein AM. Use of public performance reports: a survey of patients undergoing cardiac surgery. JAMA 1998; 279:1638–1642.
- The Henry J. Kaiser Family Foundation. 2008 Update on Consumers’ Views of Patient Safety and Quality Information: Summary & Chartpack; October 2008. http://www.kff.org/kaiserpolls/upload/7819.pdf. Accessed June 10, 2009.
- Schneider EC, Epstein AM. Influence of cardiac-surgery performance reports on referral practices and access to care: a survey of cardiovascular specialists. N Engl J Med 1996; 335:251–256.
- Jha AK, Epstein AM. The predictive accuracy of the New York State coronary artery bypass surgery report-card system. Health Aff (Millwood) 2006; 25:844–855.
- Remus D. Pay for performance: CMS/Premier Hospital Quality Incentive Demonstration Project—year 1 results, December 2005. PowerPoint presentation available at: http://www.premierinc.com/quality-safety/tools-services/p4p/hqi/results/index.jsp. Accessed June 10, 2009.
- Lindenauer PK, Remus D, Roman S, et al. Public reporting and pay for performance in hospital quality improvement. N Engl J Med 2007; 356:486–496.
- Glickman SW, Ou FS, DeLong ER, et al. Pay for performance, quality of care, and outcomes in acute myocardial infarction. JAMA 2007; 297:2373–2380.
- Kanwar M, Brar N, Khatib R, Fakih MG. Misdiagnosis of community-acquired pneumonia and inappropriate utilization of antibiotics: side effects of the 4-h antibiotic administration rule. Chest 2007; 131:1865–1869.
- Bradley EH, Herrin J, Elbel B, et al. Hospital quality for acute myocardial infarction: correlation among process measures and relationship with short-term mortality. JAMA 2006; 296:72–78.
- Dimick JB, Welch HG, Birkmeyer JD. Surgical mortality as an indicator of hospital quality: the problem with small sample size. JAMA 2004; 292:847–851.
- Kowalczyk L, Smith S. Hospital halts heart surgeries due to deaths: high rate cited at Worcester facility. The Boston Globe. September 22, 2005.
- Ettinger WH, Hylka SM, Phillips RA, Harrison LH Jr, Cyr JA, Sussman AJ. When things go wrong: the impact of being a statistical outlier in publicly reported coronary artery bypass graft surgery mortality data. Am J Med Qual 2008; 23:90–95.
- Leapfrog hospital quality ratings. The Leapfrog Group Web site. http://www.leapfroggroup.org/cp. Accessed June 10, 2009.
- Hospital Compare: a quality tool provided by Medicare. U.S. Department of Health & Human Services Web site. http://www.hospitalcompare.hhs.gov. Accessed June 10, 2009.
- Why Not the Best (Beta): A Health Care Quality Improvement Resource. The Commonwealth Fund. http://www.WhyNotTheBest.org. Accessed May 6, 2009.
- Hospital-acquired infections in Pennsylvania. Pennsylvania Health Care Cost Containment Council Web site. http://www.phc4.org. Accessed April 6, 2009.
- Hibbard JH, Stockard J, Tusler M. Does publicizing hospital performance stimulate quality improvement efforts? Health Aff (Millwood) 2003; 22:84–94.
- Fung CH, Lim YW, Mattke S, Damberg C, Shekelle PG. Systematic review: the evidence that publishing patient care performance data improves quality of care. Ann Intern Med 2008; 148:111–123.
- Peterson ED, DeLong ER, Jollis JG, et al. The effects of New York’s bypass surgery provider profiling on access to care and patient outcomes in the elderly. J Am Coll Cardiol 1998; 32:993–999.
- Baker DW, Einstadter D, Thomas C, et al. The effect of publicly reporting hospital performance on market share and risk-adjusted mortality at high-mortality hospitals. Med Care 2003; 41:729–740.
- Graylock J. After chest pains, Clinton set to undergo bypass surgery. USA Today. September 3, 2004.
- Adult Cardiac Surgery in New York State, 1999–2001. Albany, NY: New York State Department of Health; April 2004. http://www.health.state.ny.us/nysdoh/heart/pdf/1999-2001_cabg.pdf. Accessed June 10, 2009.
- Schneider EC, Epstein AM. Use of public performance reports: a survey of patients undergoing cardiac surgery. JAMA 1998; 279:1638–1642.
- The Henry J. Kaiser Family Foundation. 2008 Update on Consumers’ Views of Patient Safety and Quality Information: Summary & Chartpack; October 2008. http://www.kff.org/kaiserpolls/upload/7819.pdf. Accessed June 10, 2009.
- Schneider EC, Epstein AM. Influence of cardiac-surgery performance reports on referral practices and access to care: a survey of cardiovascular specialists. N Engl J Med 1996; 335:251–256.
- Jha AK, Epstein AM. The predictive accuracy of the New York State coronary artery bypass surgery report-card system. Health Aff (Millwood) 2006; 25:844–855.
- Remus D. Pay for performance: CMS/Premier Hospital Quality Incentive Demonstration Project—year 1 results, December 2005. PowerPoint presentation available at: http://www.premierinc.com/quality-safety/tools-services/p4p/hqi/results/index.jsp. Accessed June 10, 2009.
- Lindenauer PK, Remus D, Roman S, et al. Public reporting and pay for performance in hospital quality improvement. N Engl J Med 2007; 356:486–496.
- Glickman SW, Ou FS, DeLong ER, et al. Pay for performance, quality of care, and outcomes in acute myocardial infarction. JAMA 2007; 297:2373–2380.
- Kanwar M, Brar N, Khatib R, Fakih MG. Misdiagnosis of community-acquired pneumonia and inappropriate utilization of antibiotics: side effects of the 4-h antibiotic administration rule. Chest 2007; 131:1865–1869.
- Bradley EH, Herrin J, Elbel B, et al. Hospital quality for acute myocardial infarction: correlation among process measures and relationship with short-term mortality. JAMA 2006; 296:72–78.
- Dimick JB, Welch HG, Birkmeyer JD. Surgical mortality as an indicator of hospital quality: the problem with small sample size. JAMA 2004; 292:847–851.
KEY POINTS
- Public reporting programs have expanded in recent years, driven by national policy imperatives to improve safety, increased demands for transparency, patient “consumerism,” and the growth of information technology.
- Hospital-based pay-for-performance programs have had only a minor impact on quality so far, possibly because financial incentives have been small and much of the programs’ potential benefit may be preempted by existing public reporting efforts.
- These programs have considerable potential to accelerate improvement in quality but are limited by a need for more-nuanced process measures and better risk-adjustment methods.
- These programs may lead to unintended consequences such as misuse or overuse of measured services, “cherry-picking” of low-risk patients, or misclassification of providers.
- Continued growth of the Internet and social-networking sites will likely enhance and change the way patients use and share information about the quality of health care.
Cardiac risk stratification for noncardiac surgery
In patients undergoing noncardiac surgery, preoperative intervention for a cardiac condition is rarely needed simply to reduce the risk of the surgery unless such intervention is indicated separate from the preoperative context.
This is the overriding message of the 2007 guidelines on perioperative cardiovascular evaluation and care for noncardiac surgery issued by the American College of Cardiology (ACC) and American Heart Association (AHA),1 for which I was privileged to chair the writing committee. This article outlines current best practices in cardiac risk stratification for noncardiac surgery, highlighting key recommendations from the ACC/AHA 2007 perioperative guidelines.
PURPOSE OF THE PREOPERATIVE CARDIAC EVALUATION
Provide clinical judgment, not clearance for surgery
A proper cardiac evaluation prior to noncardiac surgery involves a comprehensive patient assessment that draws on clinical findings, the clinical experience of the consulting physician (typically a cardiologist or internist), and an assessment of the literature. The purpose is not to give medical clearance for surgery but rather to provide informed clinical judgment to the anesthesiologist and the surgical team in terms of the following1:
- The patient’s current medical status
- Recommendations regarding the management and risk of cardiac problems during the perioperative period
- The patient’s clinical risk profile, to assist with treatment decisions that may affect short- or long-term cardiac outcomes.
Order tests only when results may change management
The consulting physician’s clinical judgment is critical in determining the need to order any specific tests. In general, a test to further define cardiac risk is valid only when its results could change the planned management and lead to a specific intervention. Potential interventions that may result from knowledge gained through testing include:
- Delaying the operation because of unstable symptoms
- Coronary revascularization
- Attempting medical optimization before surgery
- Involving additional specialists or providers in the patient’s perioperative care
- Modification of intraoperative monitoring
- Modification of postoperative monitoring
- Modification of the surgical location, particularly when the procedure is scheduled for an ambulatory surgical center.
The cardiac evaluation should result in an estimation of cardiac risk. If the consulting physician’s estimation of risk is not clearly above or below the threshold for a potential intervention, then further testing may be indicated to further define the need for interventions (ie, reaching the threshold for action).
WHAT TO WORRY ABOUT FIRST: HIGH-RISK CONDITIONS THAT REQUIRE EVALUATION AND TREATMENT
In a recommendation categorized as a Class I, Level B endorsement,* the ACC/AHA 2007 perioperative guidelines specify four active cardiac conditions for which an evaluation and treatment are required before noncardiac surgery1:
- Unstable coronary syndromes, including unstable or severe angina or recent myocardial infarction (MI). These syndromes should be the first and most important consideration. Unstable angina is a hypercoagulable state, as is recent MI. The hypercoagulability of these conditions is compounded by the hypercoagulability induced by the perioperative setting itself. As a result, the rate of perioperative MI or death in the setting of unstable angina is as high as 28%.2 In the case of unstable coronary syndromes, delaying surgery is appropriate if the risks of the surgery are deemed greater than its potential benefits.
- Decompensated heart failure, defined as New York Heart Association functional class IV disease or worsening or new-onset heart failure.
- Significant arrhythmias, defined as high-grade or Mobitz II atrioventricular block, third-degree atrioventricular heart block, symptomatic ventricular arrhythmias, supraventricular arrhythmias with uncontrolled ventricular rate, symptomatic bradycardia, and newly recognized ventricular tachycardia.
- Severe valvular disease, defined as severe aortic stenosis and symptomatic mitral stenosis.
(*The ACC/AHA 2007 perioperative guidelines make recommendations by classifying the magnitude of benefit versus risk [I = the intervention should be undertaken; IIa = the intervention is reasonable to undertake; IIb = the intervention may be considered; III = the intervention should not be undertaken] and assigning a level of supporting evidence [A = highest level of evidence; B = limited evidence; C = very limited evidence].)
CARDIAC RISK STRATIFICATION: INITIAL PATIENT ASSESSMENT
Clinical risk factors and functional capacity
The Revised Cardiac Risk Index of Lee et al3 remains the general paradigm for stratifying cardiac risk before noncardiac surgery. This validated index consists of six independent predictors of cardiac complications:
- High-risk surgery (intraperitoneal, intrathoracic, or suprainguinal vascular procedures)
- Ischemic heart disease
- History of congestive heart failure
- History of cerebrovascular disease
- Insulin therapy for diabetes mellitus
- Preoperative creatinine level greater than 2.0 mg/dL.
The more predictors a patient has, the greater the risk of perioperative complications. Thus, the Revised Cardiac Risk Index is a good tool for establishing a baseline risk level for use in determining whether a preoperative or perioperative intervention is likely to make a difference in the patient’s surgical outcome. For the purpose of the algorithmic approach to testing, the surgical procedure is not considered a risk factor. Additionally, type 2 diabetes mellitus is also considered a risk factor.
Another important determinant of risk is the patient’s functional capacity. A study of 600 patients undergoing major noncardiac procedures found that poor self-reported exercise capacity, defined as an inability to walk four blocks or climb two flights of stairs, was associated with significantly more perioperative complications than was good exercise capacity.4 Simple instruments such as the Duke Activity Status Index5 can be used to estimate the patient’s functional capacity.
Procedure-specific risk
In addition to patient-specific factors, surgery-specific cardiac risk can be important, especially in patients with more than two clinical risk factors. The ACC/AHA 2007 perioperative guidelines identify three categories of surgery-specific risk1:
- Vascular surgery (the highest-risk category and also the most extensively studied), which has been associated with cardiac morbidity rates of greater than 5% in many reports. Examples include aortic and other major vascular surgery, as well as peripheral vascular surgery.
- Intermediate-risk surgery, for which reported cardiac morbidity rates range from 1% to 5%. Examples include intraperitoneal and intrathoracic procedures, carotid endarterectomy, head and neck surgery, orthopedic surgery, and prostate surgery.
- Low-risk surgery, for which reported cardiac morbidity rates are generally below 1%. Examples include endoscopic and superficial procedures, cataract surgery, breast surgery, and ambulatory surgery. Patients undergoing these procedures do not generally require further preoperative cardiac testing.1
Of course, some variability exists within each risk level as a result of institutional differences in surgical volume and expertise as well as in preoperative evaluation and other processes of care. Endovascular surgery is considered intermediate risk from a perioperative perspective but is in the same risk category as vascular surgery from a 1-year perspective.
Risk stratification promotes good perioperative outcomes
Appropriate risk stratification can make the day of surgery among the safest times for patients undergoing outpatient procedures. A retrospective analysis of Medicare claims from the late 1990s for more than 500,000 elderly patients undergoing low-risk procedures in various outpatient settings found that the mortality rate was only 1 in 50,000 on the day of surgery but increased substantially over the following 7 days and 30 days.6 This was likely a reflection of the diligence applied to managing patient-specific risk factors before proceeding to outpatient surgery.
HEART RATE CONTROL
Chronic beta-blockade can obviate need for cardiac testing
The DECREASE (Dutch Echocardiographic Cardiac Risk Evaluation Applying Strees Echo) II trial assessed the value of cardiac testing before major vascular surgery in intermediate-risk patients (ie, with one or two cardiac risk factors) receiving chronic beta-blocker therapy begun 7 to 30 days prior to surgery.7 Among the study’s 770 intermediate-risk patients, the primary outcome—cardiac death or MI at 30 days—was no different between those randomized to receive stress testing or no stress testing. The investigators concluded that cardiac testing can safely be omitted in intermediate-risk patients if beta-blockers are used with the aim of tight heart rate control.
Continue ongoing beta-blocker therapy, start in select high-risk patients
The ACC/AHA 2007 perioperative guidelines recommend continuing beta-blocker therapy in patients who are already receiving these agents (Class I, Level C). For patients not already taking beta-blockers, their initiation is recommended in those undergoing vascular surgery who have ischemia on preoperative testing (Class I, Level B). The guidelines designate beta-blockers as “probably” recommended (Class IIa, Level B) for several other patient subgroups with high cardiac risk, mainly in the setting of vascular surgery.1
Notably, the guidelines were written before publication of the Perioperative Ischemic Evaluation (POISE),8 which questioned the risk/benefit profile of perioperative beta-blockade in patients with or at high risk of atherosclerotic disease (see the Poldermans–Devereaux debate on page S84 of this supplement), and therefore may require revision (an update is scheduled for release in November 2009).
LIMITED ROLE FOR CORONARY REVASCULARIZATION
Until recently, no randomized trials had assessed the benefit of prophylactic coronary revascularization to reduce the perioperative risk of noncardiac surgery. The first large such trial was the Coronary Artery Revascularization Prophylaxis (CARP) study, which randomized 510 patients scheduled for major elective vascular surgery to undergo or not undergo coronary artery revascularization before the procedure.9 The study found that revascularization failed to affect any outcome measure, including mortality or the development of MI, out to 6 years of follow-up. Notably, the CARP population consisted mostly of patients with single-, double-, or mild triple-vessel coronary artery disease, so the study was limited in that it did not include patients with strong indications for coronary artery bypass graft surgery (CABG).7
A reanalysis of the CARP results by the type of revascularization procedure—CABG or percutaneous coronary intervention (PCI)—revealed that patients undergoing CABG had lower rates of death, MI, and additional revascularization procedures compared with those undergoing PCI, despite the presumably more extensive disease of the CABG recipients.10
Benefit apparently limited to left main disease
Further analysis of patients in the CARP trial who underwent coronary angiography found that one subgroup—patients with left main disease—did experience an improvement in survival with preoperative coronary revascularization.11
In a subsequent randomized pilot study, Poldermans et al found no advantage to preoperative coronary revascularization among patients with extensive ischemia who underwent major vascular surgery.12 While this study was not adequately sized to definitively address the value of preoperative revascularization in these high-risk patients, its results are consistent with those of the CARP trial.
In a retrospective cohort study of patients who underwent noncardiac surgery, Posner and colleagues found that rates of adverse cardiac outcomes among patients who had recent PCI (≤ 90 days before surgery) were similar to rates among matched controls with nonrevascularized coronary disease.13 Patients who had had remote PCI (> 90 days before surgery) had a lower risk of poor outcomes than did matched controls with nonrevascularized disease, but had a higher risk than did controls without coronary disease.13
PATIENTS WITH CORONARY STENTS: STENT TYPE AND TIME SINCE PLACEMENT ARE KEY
The lack of benefit from prophylactic PCI prior to noncardiac surgery also applies to PCI procedures that involve coronary stent placement. For instance, a propensity-score analysis found no benefit from prophylactic PCI (using stents in the vast majority of cases) in patients with coronary artery disease in terms of adverse coronary events or death following aortic surgery.14
In patients who have undergone prior PCI, noncardiac surgery poses special challenges, especially in relation to stents. Restenosis is a particular concern with the use of bare-metal stents, and development of stent thrombosis is a particular risk with the use of drug-eluting stents.15 The use of drug-eluting stents requires intensive antiplatelet therapy for at least 1 year following stent implantation to prevent stent thrombosis.16
Time interval to surgery after bare-metal stent placement
The effect of prior PCI with bare-metal stents on outcomes following noncardiac surgery was examined in a recent large retrospective study by Nuttall et al.17 The incidence of major cardiac events was found to be lowest when noncardiac surgery was performed more than 90 days after PCI with bare-metal stents. Using patients who had a greater than 90-day interval before surgery as the reference group, propensity analysis showed that performing surgery within 30 days of PCI was associated with an odds ratio of 3.6 for major cardiac events. The odds ratio was reduced to 1.6 when surgery was performed 31 to 90 days after PCI. These findings suggest that 30 days may be an ideal minimum time interval, from a risk/benefit standpoint, between PCI with bare-metal stents and noncardiac surgery.
Time interval to surgery after drug-eluting stent placement
A recent retrospective study by Rabbitts et al examined patients who had noncardiac surgery after prior PCI with drug-eluting stents, focusing on the relationship between the timing of the procedures and major cardiac events during hospitalization for the surgery.18 Although the frequency of major cardiac events was not statistically significantly associated with the time between stent placement and surgery, the frequency was lowest—3.3%—when surgery followed drug-eluting stent placement by more than 365 days (versus rates of 5.7% to 6.4% for various intervals of less than 365 days).
ACC/AHA recommendations
Timing of antiplatelet interruption
Results from a prospectively maintained Dutch registry19 are consistent with the findings reviewed above: patients who underwent noncardiac surgery less than 30 days after bare-metal stent implantation or less than 6 months after drug-eluting stent implantation (early surgery group) had a significantly elevated rate of major cardiac events compared with patients in whom the interval between stenting and noncardiac surgery was longer (late surgery group). Notably, this report also found that the rate of major cardiac events within the early surgery group was significantly higher in patients whose antiplatelet therapy was discontinued during the preoperative period than in those whose antiplatelet therapy was not stopped.19
A hypercoagulable state develops within 7 to 10 days after interruption of antiplatelet therapy, at which time the patient is vulnerable to thrombosis. In general, surgery should not proceed during this time without antiplatelet coverage.
From my perspective, giving ketorolac or aspirin the morning of surgery may be beneficial for patients whose antiplatelet therapy has been stopped 7 to 10 days previously, although no data from randomized trials exist to support this practice. Theoretically, it is reasonable to stop antiplatelet therapy 4 to 5 days before surgery in patients with an increased risk of bleeding without exposing them to the hypercoagulability that would set in if therapy were stopped earlier.
A FRAMEWORK FOR CARDIAC EVALUATION
The following are among the algorithm’s key recommendations:
- Patients requiring urgent noncardiac surgery should proceed to the operating room with perioperative surveillance (Class I, Level C).
- Patients with active cardiac conditions who are undergoing nonurgent surgery should be evaluated and treated per ACC/AHA guidelines before proceeding to the operating room is considered (Class I, Level B).
- Patients scheduled for a low-risk procedure can proceed to surgery without testing (Class I, Level B).
- Patients scheduled for intermediate-risk surgery or vascular surgery are to be assessed by functional capacity and clinical risk factors. Proceeding with planned surgery is appropriate in patients with good functional capacity (Class IIa, Level B). In patients with poor or unknown functional capacity undergoing vascular surgery who have three or more clinical risk factors, testing should be considered if the results would change management (Class IIa, Level B).
- Patients with one or more clinical risk factors undergoing intermediate-risk surgery and those with fewer than three clinical risk factors undergoing vascular surgery may proceed to planned surgery with control of heart rate to diminish the stress response perioperatively (Class IIa, Level B), or they may undergo noninvasive testing, but only if the results would change management (Class IIb, Level B).
- Patients undergoing intermediate-risk or vascular surgery who have poor or unknown functional capacity but no clinical risk factors may proceed to surgery without testing (Class I, Level B).
DISCUSSION
Question from the audience: The POISE study showed a 30% reduction in nonfatal MI with routine perioperative beta-blockade but an overall increase in mortality. Since most MIs occur immediately postoperatively and sepsis occurs a bit later, would you consider continuing beta-blocker therapy for a few days to prevent an MI but then stopping it before sepsis develops?
Dr. Fleisher: I’ve had discussions with sepsis experts about the link between beta-blocker therapy and sepsis and death in POISE, and the belief is that beta-blockers do not cause sepsis. I think that a septic patient on acute high-dose beta-blocker therapy can’t respond appropriately because of an inability to increase cardiac output. I believe we should titrate beta-blockers more closely. Information on preoperative dose titration in POISE is not available because of the way the trial was designed. Sepsis developed in only 53 of the 8,351 patients randomized in the study.
I would not start an acute beta-blocker protocol just to get a patient through surgery. I would start a perioperative hemodynamic protocol with the goal of maintaining the patient’s heart rate at lower than 80 beats per minute. Because I don’t believe that beta-blockers cause sepsis, if I initiated a beta-blocker preoperatively, I would not stop it at 2 days.
Question from the audience: Is there a time period during which a patient with a bare-metal stent could have back surgery or knee replacement surgery while not on aspirin?
Dr. Fleisher: The guidelines say that if a patient is on aspirin, it should be continued indefinitely. The issue is one of risk versus benefit. For back surgery, if bleeding is a concern, stopping aspirin for 6 or 7 days after the 30-day period following PCI is not unreasonable, but I would not stop it during the first 30 days following PCI.
Question from the audience: I don’t assess for vascular surgery but rather for the Whipple procedure [radical pancreatoduodenectomy], and I use the Revised Cardiac Risk Index to assess the number of risk factors. I believe the Whipple procedure is a high-risk operation, but it appears to be considered an intermediate-risk operation by the ACC/AHA guidelines. Is my approach to risk assessment appropriate?
Dr. Fleisher: If the rates of morbidity and mortality with the Whipple procedure are low at your institution, you might risk worsening your outcomes by applying someone else’s paradigms to your institution. There’s a big difference in risk between a surgeon who does a Whipple in 5 hours with 0.5 to 1.0 U of blood loss and a surgeon who does a 12-hour Whipple with 20 U of blood loss, necessitating a stay in the intensive care unit for multiple days. You need to consider the risk associated with your institution and specifically with the surgeon.
Question from the audience: Peripheral vascular disease is considered a coronary heart disease risk equivalent, so why is it not one of the criteria in the Revised Cardiac Risk Index?
Dr. Fleisher: The criteria are not hard and fast. The index was devised at one institution, Brigham and Women’s Hospital, in about 4,000 patients, and it has been used differently. It assigns 1 point to ischemic heart disease. It would not be inappropriate to assume that any atherosclerotic class of disease is equivalent to ischemic heart disease for risk purposes.
Question from the audience: You mentioned a 4-day window for withholding clopidogrel. Do you factor into the decision the duration of therapy? Some cardiologists go beyond the 1-year recommendation to continue clopidogrel after stenting because they believe there is still benefit.
Dr. Fleisher: The key is to confer with the cardiologist who implanted the stent, who knows the stenosis for which the stent was implanted. A problem we’ve had for years is that a practitioner will stop the antiplatelet agent without having spoken to the surgeon or the anesthesiologist. As an anesthesiologist, I need to know that someone has done a risk/benefit assessment of whether to continue antiplatelet agents in a given patient.
Question from the audience: The Revised Cardiac Risk Index of Lee et al3 includes the type of surgery in its total point system while the ACC/AHA guidelines do not. Can you explain the discrepancy?
Dr. Fleisher: We on the writing committee for the ACC/AHA 2007 perioperative guidelines made a decision to pull out the type of surgery and use the other five risk factors of Lee et al. It was a consensus of the committee because we believed that the complexity of the surgery itself is a separate consideration for risk. That’s why we included the medical risk factors and considered the surgical factors separately.
- Fleisher LA, Beckman JA, Brown KA, et al. ACC/AHA 2007 guidelines on perioperative cardiovascular evaluation and care for noncardiac surgery: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines [published correction appears in J Am Coll Cardiol 2008; 52:794–797]. J Am Coll Cardiol 2007; 50:1707–1732.
- Shah KB, Kleinman BS, Rao TLK, Jacobs HK, Mestan K, Schaafsma M. Angina and other risk factors in patients with cardiac diseases undergoing noncardiac operations. Anesth Analg 1990; 70:240–247.
- Lee TH, Marcantonio ER, Mangione CM, et al. Derivation and prospective validation of a simple index for prediction of cardiac risk of major noncardiac surgery. Circulation 1999; 100:1043–1049.
- Reilly DF, McNeely MJ, Doerner D, et al. Self-reported exercise tolerance and the risk of serious perioperative complications. Arch Intern Med 1999; 159:2185–2192.
- Nelson CL, Herndon JE, Mark DB, et al. Relation of clinical and angiographic factors to functional capacity as measured by the Duke Activity Status Index. Am J Cardiol 1991; 68:973–975.
- Fleisher LA, Pasternak LR, Herbert R, Anderson GF. Inpatient hospital admission and death after outpatient surgery in elderly patients: importance of patient and system characteristics and location of care. Arch Surg 2004; 139:67–72.
- Poldermans D, Bax JJ, Schouten O, et al. Should major vascular surgery be delayed because of preoperative cardiac testing in intermediate-risk patients receiving beta-blocker therapy with tight heart rate control? J Am Coll Cardiol 2006; 48:964–969.
- POISE Study Group, Devereaux PJ, Yang H, Yusuf S, et al. Effects of extended-release metoprolol succinate in patients undergoing non-cardiac surgery (POISE trial): a randomized controlled trial. Lancet 2008; 371:1839–1847.
- McFalls EO, Ward HB, Moritz TE, et al. Coronary-artery revascularization before elective major vascular surgery. N Engl J Med 2004; 351:2795–2804.
- Ward HB, Kelly RF, Thottapurathu L, et al. Coronary artery bypass grafting is superior to percutaneous coronary intervention in prevention of perioperative myocardial infarctions during subsequent vascular surgery. Ann Thorac Surg 2006; 82:795–801.
- Garcia S, Moritz TE, Ward HB, et al. Usefulness of revascularization of patients with multivessel coronary artery disease before elective vascular surgery for abdominal aortic and peripheral occlusive disease. Am J Cardiol 2008; 102:809–813.
- Poldermans D, Schouten O, Vidakovic R, et al. A clinical randomized trial to evaluate the safety of a noninvasive approach in high-risk patients undergoing major vascular surgery: the DECREASE-V Pilot Study. J Am Coll Cardiol 2007; 49:1763–1769.
- Posner KL, Van Norman GA, Chan V. Adverse cardiac outcomes after noncardiac surgery in patients with prior percutaneous transluminal coronary angioplasty. Anesth Analg 1999; 89:553–560.
- Godet G, Riou B, Bertrand M, et al. Does preoperative coronary angioplasty improve perioperative cardiac outcome? Anesthesiology 2005; 102:739–746.
- Shuchman M. Debating the risks of drug-eluting stents. N Engl J Med 2007; 356:325–328.
- King SB III, Smith SC Jr, Hirshfeld JW Jr, et al. ACC/AHA/SCAI. 2007 focused update of the ACC/AHA/SCAI 2005 guideline update for percutaneous coronary intervention: a report of the American College of Cardiology/American Heart Association Task Force on Practice guidelines. J Am Coll Cardiol 2008; 51:172–209.
- Nuttall GA, Brown MJ, Stombaugh JW, et al. Time and cardiac risk of surgery after bare-metal stent percutaneous coronary intervention. Anesthesiology 2008; 109:588–595.
- Rabbitts JA, Nuttall GA, Brown MJ, et al. Cardiac risk of noncardiac surgery after percutaneous coronary intervention with drug-eluting stents. Anesthesiology 2008; 109:596–604.
- Schouten O, van Domburg RT, Bax JJ, et al. Noncardiac surgery after coronary stenting: early surgery and interruption of antiplatelet therapy are associated with an increase in major adverse cardiac events. J Am Coll Cardiol 2007; 49:122–124.
- Correction to Fleisher et al, J Am Coll Cardiol 2007; 50:1707–1732. J Am Coll Cardiol 2008; 52:794–797.
In patients undergoing noncardiac surgery, preoperative intervention for a cardiac condition is rarely needed simply to reduce the risk of the surgery unless such intervention is indicated separate from the preoperative context.
This is the overriding message of the 2007 guidelines on perioperative cardiovascular evaluation and care for noncardiac surgery issued by the American College of Cardiology (ACC) and American Heart Association (AHA),1 for which I was privileged to chair the writing committee. This article outlines current best practices in cardiac risk stratification for noncardiac surgery, highlighting key recommendations from the ACC/AHA 2007 perioperative guidelines.
PURPOSE OF THE PREOPERATIVE CARDIAC EVALUATION
Provide clinical judgment, not clearance for surgery
A proper cardiac evaluation prior to noncardiac surgery involves a comprehensive patient assessment that draws on clinical findings, the clinical experience of the consulting physician (typically a cardiologist or internist), and an assessment of the literature. The purpose is not to give medical clearance for surgery but rather to provide informed clinical judgment to the anesthesiologist and the surgical team in terms of the following1:
- The patient’s current medical status
- Recommendations regarding the management and risk of cardiac problems during the perioperative period
- The patient’s clinical risk profile, to assist with treatment decisions that may affect short- or long-term cardiac outcomes.
Order tests only when results may change management
The consulting physician’s clinical judgment is critical in determining the need to order any specific tests. In general, a test to further define cardiac risk is valid only when its results could change the planned management and lead to a specific intervention. Potential interventions that may result from knowledge gained through testing include:
- Delaying the operation because of unstable symptoms
- Coronary revascularization
- Attempting medical optimization before surgery
- Involving additional specialists or providers in the patient’s perioperative care
- Modification of intraoperative monitoring
- Modification of postoperative monitoring
- Modification of the surgical location, particularly when the procedure is scheduled for an ambulatory surgical center.
The cardiac evaluation should result in an estimation of cardiac risk. If the consulting physician’s estimation of risk is not clearly above or below the threshold for a potential intervention, then further testing may be indicated to further define the need for interventions (ie, reaching the threshold for action).
WHAT TO WORRY ABOUT FIRST: HIGH-RISK CONDITIONS THAT REQUIRE EVALUATION AND TREATMENT
In a recommendation categorized as a Class I, Level B endorsement,* the ACC/AHA 2007 perioperative guidelines specify four active cardiac conditions for which an evaluation and treatment are required before noncardiac surgery1:
- Unstable coronary syndromes, including unstable or severe angina or recent myocardial infarction (MI). These syndromes should be the first and most important consideration. Unstable angina is a hypercoagulable state, as is recent MI. The hypercoagulability of these conditions is compounded by the hypercoagulability induced by the perioperative setting itself. As a result, the rate of perioperative MI or death in the setting of unstable angina is as high as 28%.2 In the case of unstable coronary syndromes, delaying surgery is appropriate if the risks of the surgery are deemed greater than its potential benefits.
- Decompensated heart failure, defined as New York Heart Association functional class IV disease or worsening or new-onset heart failure.
- Significant arrhythmias, defined as high-grade or Mobitz II atrioventricular block, third-degree atrioventricular heart block, symptomatic ventricular arrhythmias, supraventricular arrhythmias with uncontrolled ventricular rate, symptomatic bradycardia, and newly recognized ventricular tachycardia.
- Severe valvular disease, defined as severe aortic stenosis and symptomatic mitral stenosis.
(*The ACC/AHA 2007 perioperative guidelines make recommendations by classifying the magnitude of benefit versus risk [I = the intervention should be undertaken; IIa = the intervention is reasonable to undertake; IIb = the intervention may be considered; III = the intervention should not be undertaken] and assigning a level of supporting evidence [A = highest level of evidence; B = limited evidence; C = very limited evidence].)
CARDIAC RISK STRATIFICATION: INITIAL PATIENT ASSESSMENT
Clinical risk factors and functional capacity
The Revised Cardiac Risk Index of Lee et al3 remains the general paradigm for stratifying cardiac risk before noncardiac surgery. This validated index consists of six independent predictors of cardiac complications:
- High-risk surgery (intraperitoneal, intrathoracic, or suprainguinal vascular procedures)
- Ischemic heart disease
- History of congestive heart failure
- History of cerebrovascular disease
- Insulin therapy for diabetes mellitus
- Preoperative creatinine level greater than 2.0 mg/dL.
The more predictors a patient has, the greater the risk of perioperative complications. Thus, the Revised Cardiac Risk Index is a good tool for establishing a baseline risk level for use in determining whether a preoperative or perioperative intervention is likely to make a difference in the patient’s surgical outcome. For the purpose of the algorithmic approach to testing, the surgical procedure is not considered a risk factor. Additionally, type 2 diabetes mellitus is also considered a risk factor.
Another important determinant of risk is the patient’s functional capacity. A study of 600 patients undergoing major noncardiac procedures found that poor self-reported exercise capacity, defined as an inability to walk four blocks or climb two flights of stairs, was associated with significantly more perioperative complications than was good exercise capacity.4 Simple instruments such as the Duke Activity Status Index5 can be used to estimate the patient’s functional capacity.
Procedure-specific risk
In addition to patient-specific factors, surgery-specific cardiac risk can be important, especially in patients with more than two clinical risk factors. The ACC/AHA 2007 perioperative guidelines identify three categories of surgery-specific risk1:
- Vascular surgery (the highest-risk category and also the most extensively studied), which has been associated with cardiac morbidity rates of greater than 5% in many reports. Examples include aortic and other major vascular surgery, as well as peripheral vascular surgery.
- Intermediate-risk surgery, for which reported cardiac morbidity rates range from 1% to 5%. Examples include intraperitoneal and intrathoracic procedures, carotid endarterectomy, head and neck surgery, orthopedic surgery, and prostate surgery.
- Low-risk surgery, for which reported cardiac morbidity rates are generally below 1%. Examples include endoscopic and superficial procedures, cataract surgery, breast surgery, and ambulatory surgery. Patients undergoing these procedures do not generally require further preoperative cardiac testing.1
Of course, some variability exists within each risk level as a result of institutional differences in surgical volume and expertise as well as in preoperative evaluation and other processes of care. Endovascular surgery is considered intermediate risk from a perioperative perspective but is in the same risk category as vascular surgery from a 1-year perspective.
Risk stratification promotes good perioperative outcomes
Appropriate risk stratification can make the day of surgery among the safest times for patients undergoing outpatient procedures. A retrospective analysis of Medicare claims from the late 1990s for more than 500,000 elderly patients undergoing low-risk procedures in various outpatient settings found that the mortality rate was only 1 in 50,000 on the day of surgery but increased substantially over the following 7 days and 30 days.6 This was likely a reflection of the diligence applied to managing patient-specific risk factors before proceeding to outpatient surgery.
HEART RATE CONTROL
Chronic beta-blockade can obviate need for cardiac testing
The DECREASE (Dutch Echocardiographic Cardiac Risk Evaluation Applying Strees Echo) II trial assessed the value of cardiac testing before major vascular surgery in intermediate-risk patients (ie, with one or two cardiac risk factors) receiving chronic beta-blocker therapy begun 7 to 30 days prior to surgery.7 Among the study’s 770 intermediate-risk patients, the primary outcome—cardiac death or MI at 30 days—was no different between those randomized to receive stress testing or no stress testing. The investigators concluded that cardiac testing can safely be omitted in intermediate-risk patients if beta-blockers are used with the aim of tight heart rate control.
Continue ongoing beta-blocker therapy, start in select high-risk patients
The ACC/AHA 2007 perioperative guidelines recommend continuing beta-blocker therapy in patients who are already receiving these agents (Class I, Level C). For patients not already taking beta-blockers, their initiation is recommended in those undergoing vascular surgery who have ischemia on preoperative testing (Class I, Level B). The guidelines designate beta-blockers as “probably” recommended (Class IIa, Level B) for several other patient subgroups with high cardiac risk, mainly in the setting of vascular surgery.1
Notably, the guidelines were written before publication of the Perioperative Ischemic Evaluation (POISE),8 which questioned the risk/benefit profile of perioperative beta-blockade in patients with or at high risk of atherosclerotic disease (see the Poldermans–Devereaux debate on page S84 of this supplement), and therefore may require revision (an update is scheduled for release in November 2009).
LIMITED ROLE FOR CORONARY REVASCULARIZATION
Until recently, no randomized trials had assessed the benefit of prophylactic coronary revascularization to reduce the perioperative risk of noncardiac surgery. The first large such trial was the Coronary Artery Revascularization Prophylaxis (CARP) study, which randomized 510 patients scheduled for major elective vascular surgery to undergo or not undergo coronary artery revascularization before the procedure.9 The study found that revascularization failed to affect any outcome measure, including mortality or the development of MI, out to 6 years of follow-up. Notably, the CARP population consisted mostly of patients with single-, double-, or mild triple-vessel coronary artery disease, so the study was limited in that it did not include patients with strong indications for coronary artery bypass graft surgery (CABG).7
A reanalysis of the CARP results by the type of revascularization procedure—CABG or percutaneous coronary intervention (PCI)—revealed that patients undergoing CABG had lower rates of death, MI, and additional revascularization procedures compared with those undergoing PCI, despite the presumably more extensive disease of the CABG recipients.10
Benefit apparently limited to left main disease
Further analysis of patients in the CARP trial who underwent coronary angiography found that one subgroup—patients with left main disease—did experience an improvement in survival with preoperative coronary revascularization.11
In a subsequent randomized pilot study, Poldermans et al found no advantage to preoperative coronary revascularization among patients with extensive ischemia who underwent major vascular surgery.12 While this study was not adequately sized to definitively address the value of preoperative revascularization in these high-risk patients, its results are consistent with those of the CARP trial.
In a retrospective cohort study of patients who underwent noncardiac surgery, Posner and colleagues found that rates of adverse cardiac outcomes among patients who had recent PCI (≤ 90 days before surgery) were similar to rates among matched controls with nonrevascularized coronary disease.13 Patients who had had remote PCI (> 90 days before surgery) had a lower risk of poor outcomes than did matched controls with nonrevascularized disease, but had a higher risk than did controls without coronary disease.13
PATIENTS WITH CORONARY STENTS: STENT TYPE AND TIME SINCE PLACEMENT ARE KEY
The lack of benefit from prophylactic PCI prior to noncardiac surgery also applies to PCI procedures that involve coronary stent placement. For instance, a propensity-score analysis found no benefit from prophylactic PCI (using stents in the vast majority of cases) in patients with coronary artery disease in terms of adverse coronary events or death following aortic surgery.14
In patients who have undergone prior PCI, noncardiac surgery poses special challenges, especially in relation to stents. Restenosis is a particular concern with the use of bare-metal stents, and development of stent thrombosis is a particular risk with the use of drug-eluting stents.15 The use of drug-eluting stents requires intensive antiplatelet therapy for at least 1 year following stent implantation to prevent stent thrombosis.16
Time interval to surgery after bare-metal stent placement
The effect of prior PCI with bare-metal stents on outcomes following noncardiac surgery was examined in a recent large retrospective study by Nuttall et al.17 The incidence of major cardiac events was found to be lowest when noncardiac surgery was performed more than 90 days after PCI with bare-metal stents. Using patients who had a greater than 90-day interval before surgery as the reference group, propensity analysis showed that performing surgery within 30 days of PCI was associated with an odds ratio of 3.6 for major cardiac events. The odds ratio was reduced to 1.6 when surgery was performed 31 to 90 days after PCI. These findings suggest that 30 days may be an ideal minimum time interval, from a risk/benefit standpoint, between PCI with bare-metal stents and noncardiac surgery.
Time interval to surgery after drug-eluting stent placement
A recent retrospective study by Rabbitts et al examined patients who had noncardiac surgery after prior PCI with drug-eluting stents, focusing on the relationship between the timing of the procedures and major cardiac events during hospitalization for the surgery.18 Although the frequency of major cardiac events was not statistically significantly associated with the time between stent placement and surgery, the frequency was lowest—3.3%—when surgery followed drug-eluting stent placement by more than 365 days (versus rates of 5.7% to 6.4% for various intervals of less than 365 days).
ACC/AHA recommendations
Timing of antiplatelet interruption
Results from a prospectively maintained Dutch registry19 are consistent with the findings reviewed above: patients who underwent noncardiac surgery less than 30 days after bare-metal stent implantation or less than 6 months after drug-eluting stent implantation (early surgery group) had a significantly elevated rate of major cardiac events compared with patients in whom the interval between stenting and noncardiac surgery was longer (late surgery group). Notably, this report also found that the rate of major cardiac events within the early surgery group was significantly higher in patients whose antiplatelet therapy was discontinued during the preoperative period than in those whose antiplatelet therapy was not stopped.19
A hypercoagulable state develops within 7 to 10 days after interruption of antiplatelet therapy, at which time the patient is vulnerable to thrombosis. In general, surgery should not proceed during this time without antiplatelet coverage.
From my perspective, giving ketorolac or aspirin the morning of surgery may be beneficial for patients whose antiplatelet therapy has been stopped 7 to 10 days previously, although no data from randomized trials exist to support this practice. Theoretically, it is reasonable to stop antiplatelet therapy 4 to 5 days before surgery in patients with an increased risk of bleeding without exposing them to the hypercoagulability that would set in if therapy were stopped earlier.
A FRAMEWORK FOR CARDIAC EVALUATION
The following are among the algorithm’s key recommendations:
- Patients requiring urgent noncardiac surgery should proceed to the operating room with perioperative surveillance (Class I, Level C).
- Patients with active cardiac conditions who are undergoing nonurgent surgery should be evaluated and treated per ACC/AHA guidelines before proceeding to the operating room is considered (Class I, Level B).
- Patients scheduled for a low-risk procedure can proceed to surgery without testing (Class I, Level B).
- Patients scheduled for intermediate-risk surgery or vascular surgery are to be assessed by functional capacity and clinical risk factors. Proceeding with planned surgery is appropriate in patients with good functional capacity (Class IIa, Level B). In patients with poor or unknown functional capacity undergoing vascular surgery who have three or more clinical risk factors, testing should be considered if the results would change management (Class IIa, Level B).
- Patients with one or more clinical risk factors undergoing intermediate-risk surgery and those with fewer than three clinical risk factors undergoing vascular surgery may proceed to planned surgery with control of heart rate to diminish the stress response perioperatively (Class IIa, Level B), or they may undergo noninvasive testing, but only if the results would change management (Class IIb, Level B).
- Patients undergoing intermediate-risk or vascular surgery who have poor or unknown functional capacity but no clinical risk factors may proceed to surgery without testing (Class I, Level B).
DISCUSSION
Question from the audience: The POISE study showed a 30% reduction in nonfatal MI with routine perioperative beta-blockade but an overall increase in mortality. Since most MIs occur immediately postoperatively and sepsis occurs a bit later, would you consider continuing beta-blocker therapy for a few days to prevent an MI but then stopping it before sepsis develops?
Dr. Fleisher: I’ve had discussions with sepsis experts about the link between beta-blocker therapy and sepsis and death in POISE, and the belief is that beta-blockers do not cause sepsis. I think that a septic patient on acute high-dose beta-blocker therapy can’t respond appropriately because of an inability to increase cardiac output. I believe we should titrate beta-blockers more closely. Information on preoperative dose titration in POISE is not available because of the way the trial was designed. Sepsis developed in only 53 of the 8,351 patients randomized in the study.
I would not start an acute beta-blocker protocol just to get a patient through surgery. I would start a perioperative hemodynamic protocol with the goal of maintaining the patient’s heart rate at lower than 80 beats per minute. Because I don’t believe that beta-blockers cause sepsis, if I initiated a beta-blocker preoperatively, I would not stop it at 2 days.
Question from the audience: Is there a time period during which a patient with a bare-metal stent could have back surgery or knee replacement surgery while not on aspirin?
Dr. Fleisher: The guidelines say that if a patient is on aspirin, it should be continued indefinitely. The issue is one of risk versus benefit. For back surgery, if bleeding is a concern, stopping aspirin for 6 or 7 days after the 30-day period following PCI is not unreasonable, but I would not stop it during the first 30 days following PCI.
Question from the audience: I don’t assess for vascular surgery but rather for the Whipple procedure [radical pancreatoduodenectomy], and I use the Revised Cardiac Risk Index to assess the number of risk factors. I believe the Whipple procedure is a high-risk operation, but it appears to be considered an intermediate-risk operation by the ACC/AHA guidelines. Is my approach to risk assessment appropriate?
Dr. Fleisher: If the rates of morbidity and mortality with the Whipple procedure are low at your institution, you might risk worsening your outcomes by applying someone else’s paradigms to your institution. There’s a big difference in risk between a surgeon who does a Whipple in 5 hours with 0.5 to 1.0 U of blood loss and a surgeon who does a 12-hour Whipple with 20 U of blood loss, necessitating a stay in the intensive care unit for multiple days. You need to consider the risk associated with your institution and specifically with the surgeon.
Question from the audience: Peripheral vascular disease is considered a coronary heart disease risk equivalent, so why is it not one of the criteria in the Revised Cardiac Risk Index?
Dr. Fleisher: The criteria are not hard and fast. The index was devised at one institution, Brigham and Women’s Hospital, in about 4,000 patients, and it has been used differently. It assigns 1 point to ischemic heart disease. It would not be inappropriate to assume that any atherosclerotic class of disease is equivalent to ischemic heart disease for risk purposes.
Question from the audience: You mentioned a 4-day window for withholding clopidogrel. Do you factor into the decision the duration of therapy? Some cardiologists go beyond the 1-year recommendation to continue clopidogrel after stenting because they believe there is still benefit.
Dr. Fleisher: The key is to confer with the cardiologist who implanted the stent, who knows the stenosis for which the stent was implanted. A problem we’ve had for years is that a practitioner will stop the antiplatelet agent without having spoken to the surgeon or the anesthesiologist. As an anesthesiologist, I need to know that someone has done a risk/benefit assessment of whether to continue antiplatelet agents in a given patient.
Question from the audience: The Revised Cardiac Risk Index of Lee et al3 includes the type of surgery in its total point system while the ACC/AHA guidelines do not. Can you explain the discrepancy?
Dr. Fleisher: We on the writing committee for the ACC/AHA 2007 perioperative guidelines made a decision to pull out the type of surgery and use the other five risk factors of Lee et al. It was a consensus of the committee because we believed that the complexity of the surgery itself is a separate consideration for risk. That’s why we included the medical risk factors and considered the surgical factors separately.
In patients undergoing noncardiac surgery, preoperative intervention for a cardiac condition is rarely needed simply to reduce the risk of the surgery unless such intervention is indicated separate from the preoperative context.
This is the overriding message of the 2007 guidelines on perioperative cardiovascular evaluation and care for noncardiac surgery issued by the American College of Cardiology (ACC) and American Heart Association (AHA),1 for which I was privileged to chair the writing committee. This article outlines current best practices in cardiac risk stratification for noncardiac surgery, highlighting key recommendations from the ACC/AHA 2007 perioperative guidelines.
PURPOSE OF THE PREOPERATIVE CARDIAC EVALUATION
Provide clinical judgment, not clearance for surgery
A proper cardiac evaluation prior to noncardiac surgery involves a comprehensive patient assessment that draws on clinical findings, the clinical experience of the consulting physician (typically a cardiologist or internist), and an assessment of the literature. The purpose is not to give medical clearance for surgery but rather to provide informed clinical judgment to the anesthesiologist and the surgical team in terms of the following1:
- The patient’s current medical status
- Recommendations regarding the management and risk of cardiac problems during the perioperative period
- The patient’s clinical risk profile, to assist with treatment decisions that may affect short- or long-term cardiac outcomes.
Order tests only when results may change management
The consulting physician’s clinical judgment is critical in determining the need to order any specific tests. In general, a test to further define cardiac risk is valid only when its results could change the planned management and lead to a specific intervention. Potential interventions that may result from knowledge gained through testing include:
- Delaying the operation because of unstable symptoms
- Coronary revascularization
- Attempting medical optimization before surgery
- Involving additional specialists or providers in the patient’s perioperative care
- Modification of intraoperative monitoring
- Modification of postoperative monitoring
- Modification of the surgical location, particularly when the procedure is scheduled for an ambulatory surgical center.
The cardiac evaluation should result in an estimation of cardiac risk. If the consulting physician’s estimation of risk is not clearly above or below the threshold for a potential intervention, then further testing may be indicated to further define the need for interventions (ie, reaching the threshold for action).
WHAT TO WORRY ABOUT FIRST: HIGH-RISK CONDITIONS THAT REQUIRE EVALUATION AND TREATMENT
In a recommendation categorized as a Class I, Level B endorsement,* the ACC/AHA 2007 perioperative guidelines specify four active cardiac conditions for which an evaluation and treatment are required before noncardiac surgery1:
- Unstable coronary syndromes, including unstable or severe angina or recent myocardial infarction (MI). These syndromes should be the first and most important consideration. Unstable angina is a hypercoagulable state, as is recent MI. The hypercoagulability of these conditions is compounded by the hypercoagulability induced by the perioperative setting itself. As a result, the rate of perioperative MI or death in the setting of unstable angina is as high as 28%.2 In the case of unstable coronary syndromes, delaying surgery is appropriate if the risks of the surgery are deemed greater than its potential benefits.
- Decompensated heart failure, defined as New York Heart Association functional class IV disease or worsening or new-onset heart failure.
- Significant arrhythmias, defined as high-grade or Mobitz II atrioventricular block, third-degree atrioventricular heart block, symptomatic ventricular arrhythmias, supraventricular arrhythmias with uncontrolled ventricular rate, symptomatic bradycardia, and newly recognized ventricular tachycardia.
- Severe valvular disease, defined as severe aortic stenosis and symptomatic mitral stenosis.
(*The ACC/AHA 2007 perioperative guidelines make recommendations by classifying the magnitude of benefit versus risk [I = the intervention should be undertaken; IIa = the intervention is reasonable to undertake; IIb = the intervention may be considered; III = the intervention should not be undertaken] and assigning a level of supporting evidence [A = highest level of evidence; B = limited evidence; C = very limited evidence].)
CARDIAC RISK STRATIFICATION: INITIAL PATIENT ASSESSMENT
Clinical risk factors and functional capacity
The Revised Cardiac Risk Index of Lee et al3 remains the general paradigm for stratifying cardiac risk before noncardiac surgery. This validated index consists of six independent predictors of cardiac complications:
- High-risk surgery (intraperitoneal, intrathoracic, or suprainguinal vascular procedures)
- Ischemic heart disease
- History of congestive heart failure
- History of cerebrovascular disease
- Insulin therapy for diabetes mellitus
- Preoperative creatinine level greater than 2.0 mg/dL.
The more predictors a patient has, the greater the risk of perioperative complications. Thus, the Revised Cardiac Risk Index is a good tool for establishing a baseline risk level for use in determining whether a preoperative or perioperative intervention is likely to make a difference in the patient’s surgical outcome. For the purpose of the algorithmic approach to testing, the surgical procedure is not considered a risk factor. Additionally, type 2 diabetes mellitus is also considered a risk factor.
Another important determinant of risk is the patient’s functional capacity. A study of 600 patients undergoing major noncardiac procedures found that poor self-reported exercise capacity, defined as an inability to walk four blocks or climb two flights of stairs, was associated with significantly more perioperative complications than was good exercise capacity.4 Simple instruments such as the Duke Activity Status Index5 can be used to estimate the patient’s functional capacity.
Procedure-specific risk
In addition to patient-specific factors, surgery-specific cardiac risk can be important, especially in patients with more than two clinical risk factors. The ACC/AHA 2007 perioperative guidelines identify three categories of surgery-specific risk1:
- Vascular surgery (the highest-risk category and also the most extensively studied), which has been associated with cardiac morbidity rates of greater than 5% in many reports. Examples include aortic and other major vascular surgery, as well as peripheral vascular surgery.
- Intermediate-risk surgery, for which reported cardiac morbidity rates range from 1% to 5%. Examples include intraperitoneal and intrathoracic procedures, carotid endarterectomy, head and neck surgery, orthopedic surgery, and prostate surgery.
- Low-risk surgery, for which reported cardiac morbidity rates are generally below 1%. Examples include endoscopic and superficial procedures, cataract surgery, breast surgery, and ambulatory surgery. Patients undergoing these procedures do not generally require further preoperative cardiac testing.1
Of course, some variability exists within each risk level as a result of institutional differences in surgical volume and expertise as well as in preoperative evaluation and other processes of care. Endovascular surgery is considered intermediate risk from a perioperative perspective but is in the same risk category as vascular surgery from a 1-year perspective.
Risk stratification promotes good perioperative outcomes
Appropriate risk stratification can make the day of surgery among the safest times for patients undergoing outpatient procedures. A retrospective analysis of Medicare claims from the late 1990s for more than 500,000 elderly patients undergoing low-risk procedures in various outpatient settings found that the mortality rate was only 1 in 50,000 on the day of surgery but increased substantially over the following 7 days and 30 days.6 This was likely a reflection of the diligence applied to managing patient-specific risk factors before proceeding to outpatient surgery.
HEART RATE CONTROL
Chronic beta-blockade can obviate need for cardiac testing
The DECREASE (Dutch Echocardiographic Cardiac Risk Evaluation Applying Strees Echo) II trial assessed the value of cardiac testing before major vascular surgery in intermediate-risk patients (ie, with one or two cardiac risk factors) receiving chronic beta-blocker therapy begun 7 to 30 days prior to surgery.7 Among the study’s 770 intermediate-risk patients, the primary outcome—cardiac death or MI at 30 days—was no different between those randomized to receive stress testing or no stress testing. The investigators concluded that cardiac testing can safely be omitted in intermediate-risk patients if beta-blockers are used with the aim of tight heart rate control.
Continue ongoing beta-blocker therapy, start in select high-risk patients
The ACC/AHA 2007 perioperative guidelines recommend continuing beta-blocker therapy in patients who are already receiving these agents (Class I, Level C). For patients not already taking beta-blockers, their initiation is recommended in those undergoing vascular surgery who have ischemia on preoperative testing (Class I, Level B). The guidelines designate beta-blockers as “probably” recommended (Class IIa, Level B) for several other patient subgroups with high cardiac risk, mainly in the setting of vascular surgery.1
Notably, the guidelines were written before publication of the Perioperative Ischemic Evaluation (POISE),8 which questioned the risk/benefit profile of perioperative beta-blockade in patients with or at high risk of atherosclerotic disease (see the Poldermans–Devereaux debate on page S84 of this supplement), and therefore may require revision (an update is scheduled for release in November 2009).
LIMITED ROLE FOR CORONARY REVASCULARIZATION
Until recently, no randomized trials had assessed the benefit of prophylactic coronary revascularization to reduce the perioperative risk of noncardiac surgery. The first large such trial was the Coronary Artery Revascularization Prophylaxis (CARP) study, which randomized 510 patients scheduled for major elective vascular surgery to undergo or not undergo coronary artery revascularization before the procedure.9 The study found that revascularization failed to affect any outcome measure, including mortality or the development of MI, out to 6 years of follow-up. Notably, the CARP population consisted mostly of patients with single-, double-, or mild triple-vessel coronary artery disease, so the study was limited in that it did not include patients with strong indications for coronary artery bypass graft surgery (CABG).7
A reanalysis of the CARP results by the type of revascularization procedure—CABG or percutaneous coronary intervention (PCI)—revealed that patients undergoing CABG had lower rates of death, MI, and additional revascularization procedures compared with those undergoing PCI, despite the presumably more extensive disease of the CABG recipients.10
Benefit apparently limited to left main disease
Further analysis of patients in the CARP trial who underwent coronary angiography found that one subgroup—patients with left main disease—did experience an improvement in survival with preoperative coronary revascularization.11
In a subsequent randomized pilot study, Poldermans et al found no advantage to preoperative coronary revascularization among patients with extensive ischemia who underwent major vascular surgery.12 While this study was not adequately sized to definitively address the value of preoperative revascularization in these high-risk patients, its results are consistent with those of the CARP trial.
In a retrospective cohort study of patients who underwent noncardiac surgery, Posner and colleagues found that rates of adverse cardiac outcomes among patients who had recent PCI (≤ 90 days before surgery) were similar to rates among matched controls with nonrevascularized coronary disease.13 Patients who had had remote PCI (> 90 days before surgery) had a lower risk of poor outcomes than did matched controls with nonrevascularized disease, but had a higher risk than did controls without coronary disease.13
PATIENTS WITH CORONARY STENTS: STENT TYPE AND TIME SINCE PLACEMENT ARE KEY
The lack of benefit from prophylactic PCI prior to noncardiac surgery also applies to PCI procedures that involve coronary stent placement. For instance, a propensity-score analysis found no benefit from prophylactic PCI (using stents in the vast majority of cases) in patients with coronary artery disease in terms of adverse coronary events or death following aortic surgery.14
In patients who have undergone prior PCI, noncardiac surgery poses special challenges, especially in relation to stents. Restenosis is a particular concern with the use of bare-metal stents, and development of stent thrombosis is a particular risk with the use of drug-eluting stents.15 The use of drug-eluting stents requires intensive antiplatelet therapy for at least 1 year following stent implantation to prevent stent thrombosis.16
Time interval to surgery after bare-metal stent placement
The effect of prior PCI with bare-metal stents on outcomes following noncardiac surgery was examined in a recent large retrospective study by Nuttall et al.17 The incidence of major cardiac events was found to be lowest when noncardiac surgery was performed more than 90 days after PCI with bare-metal stents. Using patients who had a greater than 90-day interval before surgery as the reference group, propensity analysis showed that performing surgery within 30 days of PCI was associated with an odds ratio of 3.6 for major cardiac events. The odds ratio was reduced to 1.6 when surgery was performed 31 to 90 days after PCI. These findings suggest that 30 days may be an ideal minimum time interval, from a risk/benefit standpoint, between PCI with bare-metal stents and noncardiac surgery.
Time interval to surgery after drug-eluting stent placement
A recent retrospective study by Rabbitts et al examined patients who had noncardiac surgery after prior PCI with drug-eluting stents, focusing on the relationship between the timing of the procedures and major cardiac events during hospitalization for the surgery.18 Although the frequency of major cardiac events was not statistically significantly associated with the time between stent placement and surgery, the frequency was lowest—3.3%—when surgery followed drug-eluting stent placement by more than 365 days (versus rates of 5.7% to 6.4% for various intervals of less than 365 days).
ACC/AHA recommendations
Timing of antiplatelet interruption
Results from a prospectively maintained Dutch registry19 are consistent with the findings reviewed above: patients who underwent noncardiac surgery less than 30 days after bare-metal stent implantation or less than 6 months after drug-eluting stent implantation (early surgery group) had a significantly elevated rate of major cardiac events compared with patients in whom the interval between stenting and noncardiac surgery was longer (late surgery group). Notably, this report also found that the rate of major cardiac events within the early surgery group was significantly higher in patients whose antiplatelet therapy was discontinued during the preoperative period than in those whose antiplatelet therapy was not stopped.19
A hypercoagulable state develops within 7 to 10 days after interruption of antiplatelet therapy, at which time the patient is vulnerable to thrombosis. In general, surgery should not proceed during this time without antiplatelet coverage.
From my perspective, giving ketorolac or aspirin the morning of surgery may be beneficial for patients whose antiplatelet therapy has been stopped 7 to 10 days previously, although no data from randomized trials exist to support this practice. Theoretically, it is reasonable to stop antiplatelet therapy 4 to 5 days before surgery in patients with an increased risk of bleeding without exposing them to the hypercoagulability that would set in if therapy were stopped earlier.
A FRAMEWORK FOR CARDIAC EVALUATION
The following are among the algorithm’s key recommendations:
- Patients requiring urgent noncardiac surgery should proceed to the operating room with perioperative surveillance (Class I, Level C).
- Patients with active cardiac conditions who are undergoing nonurgent surgery should be evaluated and treated per ACC/AHA guidelines before proceeding to the operating room is considered (Class I, Level B).
- Patients scheduled for a low-risk procedure can proceed to surgery without testing (Class I, Level B).
- Patients scheduled for intermediate-risk surgery or vascular surgery are to be assessed by functional capacity and clinical risk factors. Proceeding with planned surgery is appropriate in patients with good functional capacity (Class IIa, Level B). In patients with poor or unknown functional capacity undergoing vascular surgery who have three or more clinical risk factors, testing should be considered if the results would change management (Class IIa, Level B).
- Patients with one or more clinical risk factors undergoing intermediate-risk surgery and those with fewer than three clinical risk factors undergoing vascular surgery may proceed to planned surgery with control of heart rate to diminish the stress response perioperatively (Class IIa, Level B), or they may undergo noninvasive testing, but only if the results would change management (Class IIb, Level B).
- Patients undergoing intermediate-risk or vascular surgery who have poor or unknown functional capacity but no clinical risk factors may proceed to surgery without testing (Class I, Level B).
DISCUSSION
Question from the audience: The POISE study showed a 30% reduction in nonfatal MI with routine perioperative beta-blockade but an overall increase in mortality. Since most MIs occur immediately postoperatively and sepsis occurs a bit later, would you consider continuing beta-blocker therapy for a few days to prevent an MI but then stopping it before sepsis develops?
Dr. Fleisher: I’ve had discussions with sepsis experts about the link between beta-blocker therapy and sepsis and death in POISE, and the belief is that beta-blockers do not cause sepsis. I think that a septic patient on acute high-dose beta-blocker therapy can’t respond appropriately because of an inability to increase cardiac output. I believe we should titrate beta-blockers more closely. Information on preoperative dose titration in POISE is not available because of the way the trial was designed. Sepsis developed in only 53 of the 8,351 patients randomized in the study.
I would not start an acute beta-blocker protocol just to get a patient through surgery. I would start a perioperative hemodynamic protocol with the goal of maintaining the patient’s heart rate at lower than 80 beats per minute. Because I don’t believe that beta-blockers cause sepsis, if I initiated a beta-blocker preoperatively, I would not stop it at 2 days.
Question from the audience: Is there a time period during which a patient with a bare-metal stent could have back surgery or knee replacement surgery while not on aspirin?
Dr. Fleisher: The guidelines say that if a patient is on aspirin, it should be continued indefinitely. The issue is one of risk versus benefit. For back surgery, if bleeding is a concern, stopping aspirin for 6 or 7 days after the 30-day period following PCI is not unreasonable, but I would not stop it during the first 30 days following PCI.
Question from the audience: I don’t assess for vascular surgery but rather for the Whipple procedure [radical pancreatoduodenectomy], and I use the Revised Cardiac Risk Index to assess the number of risk factors. I believe the Whipple procedure is a high-risk operation, but it appears to be considered an intermediate-risk operation by the ACC/AHA guidelines. Is my approach to risk assessment appropriate?
Dr. Fleisher: If the rates of morbidity and mortality with the Whipple procedure are low at your institution, you might risk worsening your outcomes by applying someone else’s paradigms to your institution. There’s a big difference in risk between a surgeon who does a Whipple in 5 hours with 0.5 to 1.0 U of blood loss and a surgeon who does a 12-hour Whipple with 20 U of blood loss, necessitating a stay in the intensive care unit for multiple days. You need to consider the risk associated with your institution and specifically with the surgeon.
Question from the audience: Peripheral vascular disease is considered a coronary heart disease risk equivalent, so why is it not one of the criteria in the Revised Cardiac Risk Index?
Dr. Fleisher: The criteria are not hard and fast. The index was devised at one institution, Brigham and Women’s Hospital, in about 4,000 patients, and it has been used differently. It assigns 1 point to ischemic heart disease. It would not be inappropriate to assume that any atherosclerotic class of disease is equivalent to ischemic heart disease for risk purposes.
Question from the audience: You mentioned a 4-day window for withholding clopidogrel. Do you factor into the decision the duration of therapy? Some cardiologists go beyond the 1-year recommendation to continue clopidogrel after stenting because they believe there is still benefit.
Dr. Fleisher: The key is to confer with the cardiologist who implanted the stent, who knows the stenosis for which the stent was implanted. A problem we’ve had for years is that a practitioner will stop the antiplatelet agent without having spoken to the surgeon or the anesthesiologist. As an anesthesiologist, I need to know that someone has done a risk/benefit assessment of whether to continue antiplatelet agents in a given patient.
Question from the audience: The Revised Cardiac Risk Index of Lee et al3 includes the type of surgery in its total point system while the ACC/AHA guidelines do not. Can you explain the discrepancy?
Dr. Fleisher: We on the writing committee for the ACC/AHA 2007 perioperative guidelines made a decision to pull out the type of surgery and use the other five risk factors of Lee et al. It was a consensus of the committee because we believed that the complexity of the surgery itself is a separate consideration for risk. That’s why we included the medical risk factors and considered the surgical factors separately.
- Fleisher LA, Beckman JA, Brown KA, et al. ACC/AHA 2007 guidelines on perioperative cardiovascular evaluation and care for noncardiac surgery: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines [published correction appears in J Am Coll Cardiol 2008; 52:794–797]. J Am Coll Cardiol 2007; 50:1707–1732.
- Shah KB, Kleinman BS, Rao TLK, Jacobs HK, Mestan K, Schaafsma M. Angina and other risk factors in patients with cardiac diseases undergoing noncardiac operations. Anesth Analg 1990; 70:240–247.
- Lee TH, Marcantonio ER, Mangione CM, et al. Derivation and prospective validation of a simple index for prediction of cardiac risk of major noncardiac surgery. Circulation 1999; 100:1043–1049.
- Reilly DF, McNeely MJ, Doerner D, et al. Self-reported exercise tolerance and the risk of serious perioperative complications. Arch Intern Med 1999; 159:2185–2192.
- Nelson CL, Herndon JE, Mark DB, et al. Relation of clinical and angiographic factors to functional capacity as measured by the Duke Activity Status Index. Am J Cardiol 1991; 68:973–975.
- Fleisher LA, Pasternak LR, Herbert R, Anderson GF. Inpatient hospital admission and death after outpatient surgery in elderly patients: importance of patient and system characteristics and location of care. Arch Surg 2004; 139:67–72.
- Poldermans D, Bax JJ, Schouten O, et al. Should major vascular surgery be delayed because of preoperative cardiac testing in intermediate-risk patients receiving beta-blocker therapy with tight heart rate control? J Am Coll Cardiol 2006; 48:964–969.
- POISE Study Group, Devereaux PJ, Yang H, Yusuf S, et al. Effects of extended-release metoprolol succinate in patients undergoing non-cardiac surgery (POISE trial): a randomized controlled trial. Lancet 2008; 371:1839–1847.
- McFalls EO, Ward HB, Moritz TE, et al. Coronary-artery revascularization before elective major vascular surgery. N Engl J Med 2004; 351:2795–2804.
- Ward HB, Kelly RF, Thottapurathu L, et al. Coronary artery bypass grafting is superior to percutaneous coronary intervention in prevention of perioperative myocardial infarctions during subsequent vascular surgery. Ann Thorac Surg 2006; 82:795–801.
- Garcia S, Moritz TE, Ward HB, et al. Usefulness of revascularization of patients with multivessel coronary artery disease before elective vascular surgery for abdominal aortic and peripheral occlusive disease. Am J Cardiol 2008; 102:809–813.
- Poldermans D, Schouten O, Vidakovic R, et al. A clinical randomized trial to evaluate the safety of a noninvasive approach in high-risk patients undergoing major vascular surgery: the DECREASE-V Pilot Study. J Am Coll Cardiol 2007; 49:1763–1769.
- Posner KL, Van Norman GA, Chan V. Adverse cardiac outcomes after noncardiac surgery in patients with prior percutaneous transluminal coronary angioplasty. Anesth Analg 1999; 89:553–560.
- Godet G, Riou B, Bertrand M, et al. Does preoperative coronary angioplasty improve perioperative cardiac outcome? Anesthesiology 2005; 102:739–746.
- Shuchman M. Debating the risks of drug-eluting stents. N Engl J Med 2007; 356:325–328.
- King SB III, Smith SC Jr, Hirshfeld JW Jr, et al. ACC/AHA/SCAI. 2007 focused update of the ACC/AHA/SCAI 2005 guideline update for percutaneous coronary intervention: a report of the American College of Cardiology/American Heart Association Task Force on Practice guidelines. J Am Coll Cardiol 2008; 51:172–209.
- Nuttall GA, Brown MJ, Stombaugh JW, et al. Time and cardiac risk of surgery after bare-metal stent percutaneous coronary intervention. Anesthesiology 2008; 109:588–595.
- Rabbitts JA, Nuttall GA, Brown MJ, et al. Cardiac risk of noncardiac surgery after percutaneous coronary intervention with drug-eluting stents. Anesthesiology 2008; 109:596–604.
- Schouten O, van Domburg RT, Bax JJ, et al. Noncardiac surgery after coronary stenting: early surgery and interruption of antiplatelet therapy are associated with an increase in major adverse cardiac events. J Am Coll Cardiol 2007; 49:122–124.
- Correction to Fleisher et al, J Am Coll Cardiol 2007; 50:1707–1732. J Am Coll Cardiol 2008; 52:794–797.
- Fleisher LA, Beckman JA, Brown KA, et al. ACC/AHA 2007 guidelines on perioperative cardiovascular evaluation and care for noncardiac surgery: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines [published correction appears in J Am Coll Cardiol 2008; 52:794–797]. J Am Coll Cardiol 2007; 50:1707–1732.
- Shah KB, Kleinman BS, Rao TLK, Jacobs HK, Mestan K, Schaafsma M. Angina and other risk factors in patients with cardiac diseases undergoing noncardiac operations. Anesth Analg 1990; 70:240–247.
- Lee TH, Marcantonio ER, Mangione CM, et al. Derivation and prospective validation of a simple index for prediction of cardiac risk of major noncardiac surgery. Circulation 1999; 100:1043–1049.
- Reilly DF, McNeely MJ, Doerner D, et al. Self-reported exercise tolerance and the risk of serious perioperative complications. Arch Intern Med 1999; 159:2185–2192.
- Nelson CL, Herndon JE, Mark DB, et al. Relation of clinical and angiographic factors to functional capacity as measured by the Duke Activity Status Index. Am J Cardiol 1991; 68:973–975.
- Fleisher LA, Pasternak LR, Herbert R, Anderson GF. Inpatient hospital admission and death after outpatient surgery in elderly patients: importance of patient and system characteristics and location of care. Arch Surg 2004; 139:67–72.
- Poldermans D, Bax JJ, Schouten O, et al. Should major vascular surgery be delayed because of preoperative cardiac testing in intermediate-risk patients receiving beta-blocker therapy with tight heart rate control? J Am Coll Cardiol 2006; 48:964–969.
- POISE Study Group, Devereaux PJ, Yang H, Yusuf S, et al. Effects of extended-release metoprolol succinate in patients undergoing non-cardiac surgery (POISE trial): a randomized controlled trial. Lancet 2008; 371:1839–1847.
- McFalls EO, Ward HB, Moritz TE, et al. Coronary-artery revascularization before elective major vascular surgery. N Engl J Med 2004; 351:2795–2804.
- Ward HB, Kelly RF, Thottapurathu L, et al. Coronary artery bypass grafting is superior to percutaneous coronary intervention in prevention of perioperative myocardial infarctions during subsequent vascular surgery. Ann Thorac Surg 2006; 82:795–801.
- Garcia S, Moritz TE, Ward HB, et al. Usefulness of revascularization of patients with multivessel coronary artery disease before elective vascular surgery for abdominal aortic and peripheral occlusive disease. Am J Cardiol 2008; 102:809–813.
- Poldermans D, Schouten O, Vidakovic R, et al. A clinical randomized trial to evaluate the safety of a noninvasive approach in high-risk patients undergoing major vascular surgery: the DECREASE-V Pilot Study. J Am Coll Cardiol 2007; 49:1763–1769.
- Posner KL, Van Norman GA, Chan V. Adverse cardiac outcomes after noncardiac surgery in patients with prior percutaneous transluminal coronary angioplasty. Anesth Analg 1999; 89:553–560.
- Godet G, Riou B, Bertrand M, et al. Does preoperative coronary angioplasty improve perioperative cardiac outcome? Anesthesiology 2005; 102:739–746.
- Shuchman M. Debating the risks of drug-eluting stents. N Engl J Med 2007; 356:325–328.
- King SB III, Smith SC Jr, Hirshfeld JW Jr, et al. ACC/AHA/SCAI. 2007 focused update of the ACC/AHA/SCAI 2005 guideline update for percutaneous coronary intervention: a report of the American College of Cardiology/American Heart Association Task Force on Practice guidelines. J Am Coll Cardiol 2008; 51:172–209.
- Nuttall GA, Brown MJ, Stombaugh JW, et al. Time and cardiac risk of surgery after bare-metal stent percutaneous coronary intervention. Anesthesiology 2008; 109:588–595.
- Rabbitts JA, Nuttall GA, Brown MJ, et al. Cardiac risk of noncardiac surgery after percutaneous coronary intervention with drug-eluting stents. Anesthesiology 2008; 109:596–604.
- Schouten O, van Domburg RT, Bax JJ, et al. Noncardiac surgery after coronary stenting: early surgery and interruption of antiplatelet therapy are associated with an increase in major adverse cardiac events. J Am Coll Cardiol 2007; 49:122–124.
- Correction to Fleisher et al, J Am Coll Cardiol 2007; 50:1707–1732. J Am Coll Cardiol 2008; 52:794–797.
KEY POINTS
- In addition to patient-specific factors, preoperative cardiac assessment should account for the risk of cardiac morbidity related to the procedure itself. Vascular surgery confers the highest risk, with reported rates of cardiac morbidity often greater than 5%.
- Continuation of chronic beta-blocker therapy is prudent during the perioperative period.
- Coronary revascularization prior to noncardiac surgery is generally indicated only in unstable patients and in patients with left main disease.
- Nonurgent noncardiac surgery should be delayed for at least 30 days after PCI using a bare-metal stent and for at least 365 days after PCI using a drug-eluting stent.
- Discontinuing antiplatelet therapy in patients with coronary stents may induce a hypercoagulable state within approximately 7 to 10 days.
Perioperative care of the elderly patient: An update*
Acute hospital care is fast becoming acute geriatric care: people aged 65 years or older are only 13% of the population but account for 44% of days of care in nonfederal hospitals and 38% of discharges.1 In general, the elderly have longer hospital stays, incur greater costs, and have a higher risk of adverse outcomes than do their younger counterparts.2
Among the most common surgical procedures for patients older than 65 are percutaneous coronary intervention with stenting, coronary artery bypass graft surgery, and open reduction internal fixation for hip fracture; the latter is the most common operation in patients aged 85 years or older.3
Elderly patients frequently pose many challenges perioperatively that are not often seen in younger patients. Dementia, frailty, impaired ability to care for oneself, and malnourishment may be present at baseline and are likely to worsen postoperatively. The elderly are at increased risk of acute delirium and cognitive impairment postoperatively, which often complicates recovery and discharge placement.
This article uses a case study to review perioperative problems commonly encountered in elderly surgical patients, particularly those undergoing hip surgery. As the case is presented, I will review strategies to assess risks and prevent and mitigate postoperative cognitive dysfunction and other barriers to recovery.
CASE: AN 82-YEAR-OLD WOMAN WITH HIP FRACTURE
An 82-year-old woman is admitted to undergo open reduction internal fixation for hip fracture. She has a history of osteoarthritis, systolic hypertension, and visual impairment (20/70). Her medications include a beta-blocker, a thiazide diuretic, analgesics as needed, and a multivitamin. She was independent in all activities of daily living before the fracture. She is a social drinker and does not smoke. She has no known cardiovascular, lung, or renal disease.
Her laboratory test results are as follows:
- Blood urea nitrogen (BUN), 24 mg/dL
- Creatinine, 1.0 mg/dL
- Hemoglobin, 12.8 g/dL
- Albumin, 3.8 gm/dL
- Normal levels of thyroid-stimulating hormone and vitamin B12.
Thus, her lab results are normal except for the BUN:creatinine ratio being a bit high, at 24:1 (normal is 10:1, with ratios greater than 18:1 being associated with an increased risk of delirium4).
ASSESSING COGNITIVE RISK: POSTOPERATIVE COGNITIVE DYSFUNCTION VS DELIRIUM
Question: Which of these statements about this patient is most correct?
A. She is at high risk (> 40%) of postoperative cognitive dysfunction
B. Her risk of postoperative delirium is 5% to 10%
C. Postoperative delirium cannot be prevented
D. Preoperative haloperidol (1.5 mg/day for 3 days) will reduce the risk of delirium by 25%
The best answer is A. Postoperative cognitive dysfunction is different from delirium, though it is part of a spectrum of cognitive impairment that may occur after surgery and even persist for a prolonged period. The patient’s risk of postoperative delirium is actually a bit higher than 10% (see “Estimating the risk of delirium” below). Some evidence shows that postoperative delirium can be prevented, at least in hip fracture patients. Kalisvaart et al found that preoperative treatment with low-dose haloperidol reduced the duration and severity of delirium in elderly patients following hip surgery but did not reduce its incidence.5
Cognitive dysfunction often follows surgery
Postoperative cognitive dysfunction has long been recognized and was first described in patients after cardiac surgery, especially following coronary artery bypass graft procedures. In the last several years, we have recognized that it also occurs in patients who undergo noncardiac surgery. Postoperative cognitive dysfunction, which may persist for weeks to months, may not be obvious but can be detected by standard neuropsychological testing.6
Postoperative cognitive dysfunction is different from the “emergence delirium” that may immediately follow surgery and that is often associated with the wearing off of anesthesia. It is also distinct from “incident delirium,” which sometimes occurs over the first few postoperative days (discussed below).
Postoperative dysfunction is especially persistent in the elderly
A recent study found cognitive dysfunction to be common at hospital discharge after major noncardiac surgery in adults of all ages: rates at discharge were 36.6% in patients aged 18 to 39 years, 30.4% in those aged 40 to 59, and 41.4% in those 60 or older.7 Notably, however, the oldest group was most likely to have persistent symptoms. Three months after surgery, 12.7% of patients aged 60 or older continued to have postoperative cognitive dysfunction, which was more than double the rates in the young and middle-aged patient groups (5.7% and 5.6%, respectively).7
Although the cause of postoperative cognitive dysfunction is not well understood, predisposing factors in addition to advanced age include metabolic problems, lower educational level, and previous cerebral vascular accident.7 When elective surgery is considered by elderly patients, the decision should take into account their risk of postoperative cognitive dysfunction and the impact it may have on their quality of life.
PREDICTING AND PREVENTING DELIRIUM
Delirium is easily recognized
Delirium is a common complication of surgery. Unlike postoperative cognitive dysfunction, delirium is easy to detect clinically. It is a disorder of attention and cognition and classically presents as an acute change in mental status accompanied by the following8:
- Fluctuation in awareness
- Memory impairment
- Inattention (inability to stay on task, distractibility)
- Disorganized or illogical thinking
- Altered level of consciousness—ie, hyperalertness (agitation, pulling out intravenous lines, etc) or hypoalertness (“quiet delirium”).
Estimating the risk of delirium
Marcantonio and colleagues developed a model to predict the likelihood that delirium will develop in patients undergoing elective surgery.9 The model assigns points to various risk factors as follows:
- Age ≥ 70 years (1 point)
- History of alcohol abuse (1 point)
- Baseline cognitive impairment (1 point)
- Severe physical impairment (reduced ability to walk or perform daily activities) (1 point)
- Abnormal preoperative blood levels of electrolytes or glucose (1 point)
- Noncardiac thoracic surgery (1 point)
- Abdominal aortic aneurysm surgery (2 points).
The study to validate this model found that a score of 0 points is associated with only a 2% risk of developing postoperative delirium. A score of 3 or more points is associated with a 50% risk of postoperative delirium. A score of 1 or 2 points (as for the patient in our case study) is associated with an 11% risk, according to this Marcantonio model.9
Additionally, well-designed cohort studies of medical patients10 have identified four major independent predictors of incident delirium:
- Severe illness (eg, high fever, complicated infections)
- Baseline dementia
- Dehydration (high BUN:creatinine ratio)
- Sensory impairments (particularly visual).
Kalisvaart et al conducted a prospective cohort study to determine whether these risk factors in medical patients are applicable to elderly patients undergoing hip surgery.11 They found that the incidence of delirium was low (4%) in hip surgery patients with none of these factors, increased to 11% in patients with one or two of these factors, and increased to 37% in patients with three or four factors. These findings suggest that hip surgery patients (like our case patient) may be at greater risk of postoperative delirium than is reflected in the Marcantonio model discussed above,9 which was validated in a study of patients undergoing elective (not emergent) surgery.
Several drug classes raise dementia risk
Anticholinergic medications and other drugs with anticholinergic properties, ie, benzodiazepines and the opioid agent meperidine, also raise the risk for delirium. In general, the older an elderly patient is, the less appropriate these agents are. Many drugs that are not typically recognized as anticholinergics may have potent anticholinergic activity, including tricyclic antidepressants, first-generation antihistamines (eg, diphenhydramine), and high-dose H2-receptor blockers (particularly cimetidine); these agents too should be avoided in elderly patients.12
Strategies to reduce postoperative delirium risk
How can we lower the risk of postoperative delirium in elderly hip fracture patients? Marcantonio et al13 randomized 126 patients undergoing hip fracture repair to receive usual care alone or supplemented with the following additional measures:
- Supplemental oxygen during surgery
- Optimization of electrolytes and blood glucose preoperatively
- Discontinuation of high-risk medications
- Adequate nutritional intake (by parenteral route if necessary)
- Encouragement to get out of bed on the first postoperative day
- Treatment of severe pain.
The incidence of delirium was reduced from 50% in the usual-care group to 32% in the intervention group, and the incidence of severe delirium was reduced even more, from 29% to 12%, respectively.13
OTHER BEST PRACTICES IN PERIOPERATIVE HIP FRACTURE MANAGEMENT
In a systematic literature review to identify best practices for perioperative management of elderly patients with hip fracture, Beaupre et al14 found the following measures to be among those with the strongest evidence of benefit:
- Use of spinal or local anesthesia rather than general anesthesia
- Use of pressure-relieving mattresses to prevent pressure ulcers
- Perioperative administration of antibiotics
- Deep vein thrombosis prophylaxis.
The review concluded that providing nutritional supplementation also is probably helpful although the evidence is not robust. Additionally, it was unclear whether minimizing the delay between hospital admission and surgery has any impact on mortality.14
Is early surgery better?
Early studies suggested that the sooner a hip fracture patient goes to surgery, the lower the mortality, but this has not been supported in well-controlled trials: no difference in mortality has been found whether the patient’s conditions are first optimized to reduce the risk of surgery or if the operation commences within 24 hours.
Although mortality does not appear to be affected, avoiding delay of hip fracture repair yields improvement in other outcomes. In a well-designed prospective cohort study, Orosz et al found that medically stable patients with hip fracture (mean age, 82 years) who underwent surgery within 24 hours had fewer days of pain and less intense pain postoperatively than those whose surgery was delayed beyond 24 hours.15 The early-surgery group also had a 1.94-day reduction in average length of stay compared with the late-surgery group.
A role for clinical pathways
To determine how the application of evidence-based perioperative practices affects actual outcomes in elderly hip fracture patients, Beaupre et al used a pre/post study design to evaluate the impact of an evidence-based clinical pathway at their institution.16 Though there were no differences in in-hospital mortality or the overall costs of inpatient care in elderly hip surgery patients before and after pathway implementation, the patients undergoing surgery after pathway implementation were significantly less likely to have postoperative delirium, heart failure, pressure ulcers, and urinary tract infections compared with those undergoing surgery before implementation. The outcomes benefits of this type of multimodal intervention are likely to extend to abdominal surgical procedures as well.
CASE CONTINUED: POSTOP DAY 2―PATIENT IS CONFUSED AND CRYING IN PAIN
On the second postoperative day, our patient appears weak and slightly confused. She is not eating and is crying in pain. Her neurological exam is normal.
Question: Which is the most appropriate next step?
A. Increase physical therapy
B. Begin an antidepressant
C. Insert a nasoenteric feeding tube
D. Increase doses of analgesics
The best answer is D. With no prior history of depression, an antidepressant would probably not be useful. It is premature to recommend nasoenteric feeding. Because pain hampers physical therapy, an increase in physical therapy would likewise be premature. Because we know the patient is in pain, the correct answer perhaps seems obvious. But keep in mind that relieving pain also has many other positive ramifications: intense pain can be a cause of delirium or at least worsen its symptoms, and pain relief is a prerequisite for the physical therapy that this patient needs.
Strategies for pain control
In general, the treatment of choice for postoperative pain is low-dose morphine sulfate (eg, 1–4 mg every 2 hours, titrated as needed). Acetaminophen can be given safely to virtually all patients. Patient-controlled analgesia is reasonable for select patients but not for older patients with cognitive impairment. Nonsteroidal anti-inflammatory drugs might be helpful in younger patients and even in robust elderly patients, but they must be used very cautiously in the older population because of the risk of gastric ulcers and bleeding, acute kidney injury, fluid retention, and exacerbation of congestive heart failure.
POSTOP DAY 3: PATIENT REPORTS LONG-STANDING FATIGUE
On postoperative day 3, the patient is weak and complains of fatigue. She says that before the fracture, she was experiencing mild weight loss, fatigue, and reduced activity.
Question: What is the most likely reason for her symptoms before the fracture?
A. Frailty
B. Occult heart failure
C. Adverse drug reaction to her beta-blocker
D. Clinical depression
The best answer is A. Occult heart failure is a reasonable second choice, as it is very common in older patients and the diagnosis is easy to miss unless florid pulmonary edema or associated symptoms (eg, chest pain) are present. But this patient had no history of heart disease and was only on medications for hypertension. An adverse drug reaction, such as to the beta-blocker, is unlikely and would probably not cause weight loss. The patient had no history of depression, so clinical depression is unlikely. That said, all the choices are reasonable to consider in elderly patients reporting fatigue and weakness.
Frailty is important to recognize
It is important to identify frailty and to aggressively manage frail patients postoperatively. Although frailty is not clearly defined, Fried et al17 identified five clinical features that correlate with its underlying pathophysiology:
- Minimal physical activity (ie, “doing less”)
- Generalized (not focal) muscle weakness
- Slowed performance (eg, walking short distances takes longer)
- Fatigue or poor endurance
- Unintentional weight loss.
The presence of three or more of these features meets the criteria for frailty and is associated with increased risk for mortality over the next 3 years with or without surgery,17 although surgery probably increases the risk.
Frailty is believed to be a failure over time of the homeostatic mechanisms that keep our organ systems functioning in the face of a stress. Decline in the ability of organ systems to maintain normal function is probably caused by inflammation, chronic disease, and normal aging, and has been termed homeostenosis. As a person ages and physiologic reserves are reduced, adding a stress such as surgery or severe infection can result in organ failure—usually multiple-system organ failure. In any intensive care unit, one is likely to see elderly patients who were admitted with one medical or surgical problem and soon end up having renal, liver, or brain dysfunction as well.
Physical therapy immediately after hip fracture surgery is associated with significantly better locomotion 2 months later.18 A number of exercises are effective: range-of-motion exercises, low-impact aerobic activities, and exercises starting with low-intensity resistance (using bands, tubes, and weights) and progressing as tolerated to high-intensity resistance (with machines and pulleys) for an extended period of time.
Nutrition supplementation
Malnourishment can contribute to frailty, yet evidence for the benefits of supplementing nutrition is not strong, as noted above. However, meta-analyses of studies of nutritional interventions with meal supplementation (usually canned supplements) show that meal supplementation can improve mortality risk and reduce morbidity such as pressure ulcers in hospitalized elderly patients.19,20 The patients most likely to benefit are those who are undernourished at baseline and aged 75 years or older.
CASE CONTINUED: WHAT HAPPENS POST-DISCHARGE?
Following surgery, our patient wonders, “Where will I go next? What will my lifestyle be like?”
These are important questions to consider when first evaluating whether an elderly patient should undergo surgery. In the case of hip fracture, standard thinking is that without surgery, the patient will never recover the ability to independently walk and perform activities of daily living. But we also must recognize the considerable risks of surgery in the elderly population, particularly those aged 75 years or older.
Comprehensive discharge planning
Early and intensive discharge management enhances quality of life and may help reduce hospital costs. A good model of care involves collaboration of orthopedic surgeons, hospitalists, general internists, geriatricians, and dietitians to address procedures, diet and nutrition, mobility and activities of daily living, and pain medications.21 A case manager such as a social worker should start addressing care transition the day after surgery—planning ahead is imperative.
Following hip surgery, patients are routinely sent to skilled nursing facilities as soon as possible so they can start intensive physical therapy. Patients with significant functional impairment or who had delirium are more likely to require a prolonged hospital stay.
Naylor et al examined the effectiveness of comprehensive discharge planning in a study that randomized hospitalized patients (including surgical patients) 65 years or older to either usual discharge planning or intensive discharge planning with advanced practice nurses beginning early in hospitalization.22 The intervention group was followed by home care nurses for up to 4 weeks and had continuous telephone access to the nurses. Patients who received the intervention had a significantly lower risk of hospital readmission, and those who were readmitted had significantly shorter hospital stays. The total cost of care was also significantly lower in the intervention group.
Family conferences aid decision making
Family conferences can be very useful for working through the many questions and challenges that surgery in an elderly person can pose, including whether the patient should undergo the operation, postoperative management, and postdischarge placement.23 For patients with an uncertain prognosis because of unclear or multiple concurrent diseases, a family conference can help clarify the goals of therapy, inform the family about likely outcomes, and help determine the patient’s wishes and values. Such issues should be revisited as the postoperative course proceeds.
Family conferences also provide a good opportunity to review advanced directives, the need for life support, and possible transfers to intensive care. Family conferences can also help resolve conflicts in care management, as family members may not agree with the need for surgery, how aggressive treatment should be, or where to send the patient for rehabilitation. Differences among family members on these questions are especially common with elderly patients. Working out such issues will improve patient care, especially when done early in the hospitalization.
DISCUSSION
Question from the audience: In our preoperative clinic, we are trying to intervene to reduce delirium and postoperative cognitive dysfunction. How can we quickly screen for the most important predictors and act to reduce the risk?
Dr. Palmer: The most important risk factor for delirium is age, which obviously can’t be changed. Ask patients about alcohol use and depression. Check on nutritional status and begin supplementation if indicated. Discontinue high-risk medications. Check on electrolytes and their state of hydration; ideally, an electrolyte imbalance can be corrected preoperatively. In addition, other than in patients with end-stage renal disease, try to keep the hemoglobin above 7.5 g/dL, which appears to be associated with better outcomes and less risk of delirium.
It’s also important to remind the family to bring in the patient’s visual aids, hearing devices, and cane or walker so that they’re available right after the operation.
Intraoperative factors that are important for preventing delirium include maintaining good blood pressure levels, giving supplemental oxygen, minimizing the time under general anesthesia, and using local anesthesia if possible.
Question from the audience: How strong is the evidence for using spinal anesthesia as opposed to general anesthesia in preventing postoperative cognitive dysfunction and delirium, especially in the setting of hip fracture repair?
Dr. Palmer: The evidence is fairly soft. For patients undergoing either hip or knee arthroplasty who were randomized to receive either spinal (or local) or general anesthesia, the risk of delirium was similar, but complications such as prolonged bed rest, pressure ulcers, and catheter-related urinary tract infections were somewhat reduced in the spinal/local group.14 The relative risk of developing postoperative cognitive dysfunction is unclear—no randomized controlled trials have been conducted to answer that question.
Question from the audience: How do you use antipsychotic drugs, especially with the concerns from epidemiologic studies about an increased risk of death?
Dr. Palmer: No antipsychotic agents, including haloperidol, have a specific Food and Drug Administration–approved indication for treating agitation, dementia, or delirium. In general, they should not be used without a clear indication. That said, the usual off-label use is for patients who are severely agitated and are at risk of harming themselves or others. In an ICU setting, where patients have multiple lines, the use of these agents can be considered for a very agitated patient. Alternatives exist, but antipsychotics like haloperidol have the advantage that they can be given in small increments very rapidly and achieve good control of severe agitation.
Antipsychotic agents should only be used with great caution and for the shortest duration needed. As delirium resolves, they should be tapered fairly rapidly over a few days and ideally should be discontinued by the time of hospital discharge.
None of the antipsychotic agents—including those in the first generation and the newer atypical agents—is free of this risk of increased mortality. The mechanism is not understood; it may be torsades de pointes or hypotension leading to stroke or sudden cardiac death.
Question from the audience: What is the most efficient way to assess cognitive and physical functioning preoperatively?
Dr. Palmer: There may be a documented history of dementia, or family members may tell you if there has been memory loss or some decline in the patient’s self-care abilities. For patients without dementia, you can ask them directly if they can perform basic activities of daily living, such as getting out of bed or dressing. To assess higher-level function, ask if they can manage their own medications, pay bills, or handle their finances. If not, they might have cognitive impairment and are at higher risk for postoperative delirium. These are rather sensitive measures. There are instruments to assess this more precisely, but few clinicians have time to use them.
Quick bedside tests can help assess for delirium postoperatively. We see if patients are “alert and oriented times three” (“Do you know who you are, where you are, and the date?”). We test for attention by asking them to repeat a random string of numbers spoken 1 second apart in monotone; people who are delirious and many patients with severe dementia can’t repeat more than three numbers. A patient who is alert and oriented, has a good attention span (more than three numbers in correct order), and has no history of dementia probably doesn’t have delirium or dementia.
For physical function, ask if they can walk, get out of bed to a chair, and ambulate. If they don’t give clear answers, observe them get out of bed or a chair, walk 10 feet, and return to bed. If they can do that with good balance, especially within 10 to 15 seconds, they probably have reasonably normal mobility and are at lower risk for postoperative complications such as falls with injury.
- DeFrances CJ, Cullen KA, Kozak LJ. National Hospital Discharge Survey: 2005 annual summary with detailed diagnosis and procedure data. Vital Health Stat (Series 13) 2007; Dec(165):1–209.
- Welcome to HCUPnet: a tool for identifying, tracking, and analyzing national hospital statistics. Agency for Healthcare Research and Quality Web site. http://hcupnet.ahrq.gov. Accessed February 23, 2009.
- Kozak LJ, Owings MF, Hall MJ. National Hospital Discharge Survey: 2002 annual summary with detailed diagnosis and procedure data. Vital Health Stat (Series 13) 2005; Mar(158):1–199.
- Inouye SK, Viscoli CM, Horwitz RI, Hurst LD, Tinetti ME. A predictive model for delirium in hospitalized elderly medical patients based on admission characteristics. Ann Intern Med 1993; 119:474–481.
- Kalisvaart KJ, de Jonghe JF, Bogaards MJ, et al. Haloperidol prophylaxis for elderly hip-surgery patients at risk for delirium: a randomized placebo-controlled study. J Am Geriatr Soc 2005; 53:1658–1666.
- Silverstein JH, Timberger M, Reich DL, Uysal S. Central nervous system dysfunction after noncardiac surgery and anesthesia in the elderly. Anesthesiology 2007; 106:622–628.
- Monk TG, Weldon BC, Garvan CW, et al. Predictors of cognitive dysfunction after major noncardiac surgery. Anesthesiology 2008; 108:18–30.
- Inouye SK. Delirium in older persons. N Engl J Med 2006; 354:1157–1165.
- Marcantonio ER, Goldman L, Mangione CM, et al. A clinical prediction rule for delirium after elective noncardiac surgery. JAMA 1994; 271:134–139.
- Inouye SK. Predisposing and precipitating factors for delirium in hospitalized older patients. Dement Geriatr Cogn Disord 1999; 10:393–400.
- Kalisvaart KJ, Vreeswijk R, de Jonghe JF, et al. Risk factors and prediction of postoperative delirium in elderly hip-surgery patients: implementation and validation of a medical risk factor model. J Am Geriatr Soc 2006; 54:817–822.
- Fick DM, Cooper JW, Wade WE, et al. Updating the Beers criteria for potentially inappropriate medication use in older adults: results of a US consensus panel of experts. Arch Intern Med 2003; 163:2716–2724.
- Marcantonio ER, Flacker JM, Wright RJ, et al. Reducing delirium after hip fracture: a randomized trial. J Am Geriatr Soc 2001; 49:516–522.
- Beaupre LA, Jones CA, Saunders LD, Johnston DW, Buckingham J, Majumdar SR. Best practices for elderly hip fracture patients: a systematic overview of the evidence. J Gen Intern Med 2005; 20:1019–1025.
- Orosz GM, Magaziner J, Hannan EL, et al. Association of timing of surgery for hip fracture and patient outcomes. JAMA 2004; 291:1738–1743.
- Beaupre LA, Cinats JG, Senthilselvan A, et al. Reduced morbidity for elderly patients with a hip fracture after implementation of a perioperative evidence-based clinical pathway. Qual Saf Health Care 2006; 15:375–379.
- Fried LP, Tangen CM, Walston J, et al. Frailty in older adults: evidence for a phenotype. J Gerontol A Biol Sci Med Sci 2001; 56:M146–M156.
- Penrod JD, Boockvar KS, Litke A, et al. Physical therapy and mobility 2 and 6 months after hip fracture. J Am Geriatr Soc 2004; 52:1114–1120.
- Milne AC, Avenell A, Potter J. Meta-analysis: protein and energy supplementation in older people. Ann Intern Med 2006; 144:37–48.
- Avenell A, Handoll HH. Nutritional supplementation for hip fracture aftercare in older people. Cochrane Database Syst Rev 2006; (4):CD001880.
- Miura LN, DiPiero AR, Homer LD. Effects of a geriatrician-led hip fracture program: improvements in clinical and economic outcomes. J Am Geriatr Soc 2009; 57:159–167.
- Naylor MD, Brooten D, Campbell R, et al. Comprehensive discharge planning and home follow-up of hospitalized elders: a randomized clinical trial. JAMA 1999; 281:613–620.
- Palmer RM. Acute hospital care of the elderly: making a difference. Hospitalist 2004; (suppl):4–7.
Acute hospital care is fast becoming acute geriatric care: people aged 65 years or older are only 13% of the population but account for 44% of days of care in nonfederal hospitals and 38% of discharges.1 In general, the elderly have longer hospital stays, incur greater costs, and have a higher risk of adverse outcomes than do their younger counterparts.2
Among the most common surgical procedures for patients older than 65 are percutaneous coronary intervention with stenting, coronary artery bypass graft surgery, and open reduction internal fixation for hip fracture; the latter is the most common operation in patients aged 85 years or older.3
Elderly patients frequently pose many challenges perioperatively that are not often seen in younger patients. Dementia, frailty, impaired ability to care for oneself, and malnourishment may be present at baseline and are likely to worsen postoperatively. The elderly are at increased risk of acute delirium and cognitive impairment postoperatively, which often complicates recovery and discharge placement.
This article uses a case study to review perioperative problems commonly encountered in elderly surgical patients, particularly those undergoing hip surgery. As the case is presented, I will review strategies to assess risks and prevent and mitigate postoperative cognitive dysfunction and other barriers to recovery.
CASE: AN 82-YEAR-OLD WOMAN WITH HIP FRACTURE
An 82-year-old woman is admitted to undergo open reduction internal fixation for hip fracture. She has a history of osteoarthritis, systolic hypertension, and visual impairment (20/70). Her medications include a beta-blocker, a thiazide diuretic, analgesics as needed, and a multivitamin. She was independent in all activities of daily living before the fracture. She is a social drinker and does not smoke. She has no known cardiovascular, lung, or renal disease.
Her laboratory test results are as follows:
- Blood urea nitrogen (BUN), 24 mg/dL
- Creatinine, 1.0 mg/dL
- Hemoglobin, 12.8 g/dL
- Albumin, 3.8 gm/dL
- Normal levels of thyroid-stimulating hormone and vitamin B12.
Thus, her lab results are normal except for the BUN:creatinine ratio being a bit high, at 24:1 (normal is 10:1, with ratios greater than 18:1 being associated with an increased risk of delirium4).
ASSESSING COGNITIVE RISK: POSTOPERATIVE COGNITIVE DYSFUNCTION VS DELIRIUM
Question: Which of these statements about this patient is most correct?
A. She is at high risk (> 40%) of postoperative cognitive dysfunction
B. Her risk of postoperative delirium is 5% to 10%
C. Postoperative delirium cannot be prevented
D. Preoperative haloperidol (1.5 mg/day for 3 days) will reduce the risk of delirium by 25%
The best answer is A. Postoperative cognitive dysfunction is different from delirium, though it is part of a spectrum of cognitive impairment that may occur after surgery and even persist for a prolonged period. The patient’s risk of postoperative delirium is actually a bit higher than 10% (see “Estimating the risk of delirium” below). Some evidence shows that postoperative delirium can be prevented, at least in hip fracture patients. Kalisvaart et al found that preoperative treatment with low-dose haloperidol reduced the duration and severity of delirium in elderly patients following hip surgery but did not reduce its incidence.5
Cognitive dysfunction often follows surgery
Postoperative cognitive dysfunction has long been recognized and was first described in patients after cardiac surgery, especially following coronary artery bypass graft procedures. In the last several years, we have recognized that it also occurs in patients who undergo noncardiac surgery. Postoperative cognitive dysfunction, which may persist for weeks to months, may not be obvious but can be detected by standard neuropsychological testing.6
Postoperative cognitive dysfunction is different from the “emergence delirium” that may immediately follow surgery and that is often associated with the wearing off of anesthesia. It is also distinct from “incident delirium,” which sometimes occurs over the first few postoperative days (discussed below).
Postoperative dysfunction is especially persistent in the elderly
A recent study found cognitive dysfunction to be common at hospital discharge after major noncardiac surgery in adults of all ages: rates at discharge were 36.6% in patients aged 18 to 39 years, 30.4% in those aged 40 to 59, and 41.4% in those 60 or older.7 Notably, however, the oldest group was most likely to have persistent symptoms. Three months after surgery, 12.7% of patients aged 60 or older continued to have postoperative cognitive dysfunction, which was more than double the rates in the young and middle-aged patient groups (5.7% and 5.6%, respectively).7
Although the cause of postoperative cognitive dysfunction is not well understood, predisposing factors in addition to advanced age include metabolic problems, lower educational level, and previous cerebral vascular accident.7 When elective surgery is considered by elderly patients, the decision should take into account their risk of postoperative cognitive dysfunction and the impact it may have on their quality of life.
PREDICTING AND PREVENTING DELIRIUM
Delirium is easily recognized
Delirium is a common complication of surgery. Unlike postoperative cognitive dysfunction, delirium is easy to detect clinically. It is a disorder of attention and cognition and classically presents as an acute change in mental status accompanied by the following8:
- Fluctuation in awareness
- Memory impairment
- Inattention (inability to stay on task, distractibility)
- Disorganized or illogical thinking
- Altered level of consciousness—ie, hyperalertness (agitation, pulling out intravenous lines, etc) or hypoalertness (“quiet delirium”).
Estimating the risk of delirium
Marcantonio and colleagues developed a model to predict the likelihood that delirium will develop in patients undergoing elective surgery.9 The model assigns points to various risk factors as follows:
- Age ≥ 70 years (1 point)
- History of alcohol abuse (1 point)
- Baseline cognitive impairment (1 point)
- Severe physical impairment (reduced ability to walk or perform daily activities) (1 point)
- Abnormal preoperative blood levels of electrolytes or glucose (1 point)
- Noncardiac thoracic surgery (1 point)
- Abdominal aortic aneurysm surgery (2 points).
The study to validate this model found that a score of 0 points is associated with only a 2% risk of developing postoperative delirium. A score of 3 or more points is associated with a 50% risk of postoperative delirium. A score of 1 or 2 points (as for the patient in our case study) is associated with an 11% risk, according to this Marcantonio model.9
Additionally, well-designed cohort studies of medical patients10 have identified four major independent predictors of incident delirium:
- Severe illness (eg, high fever, complicated infections)
- Baseline dementia
- Dehydration (high BUN:creatinine ratio)
- Sensory impairments (particularly visual).
Kalisvaart et al conducted a prospective cohort study to determine whether these risk factors in medical patients are applicable to elderly patients undergoing hip surgery.11 They found that the incidence of delirium was low (4%) in hip surgery patients with none of these factors, increased to 11% in patients with one or two of these factors, and increased to 37% in patients with three or four factors. These findings suggest that hip surgery patients (like our case patient) may be at greater risk of postoperative delirium than is reflected in the Marcantonio model discussed above,9 which was validated in a study of patients undergoing elective (not emergent) surgery.
Several drug classes raise dementia risk
Anticholinergic medications and other drugs with anticholinergic properties, ie, benzodiazepines and the opioid agent meperidine, also raise the risk for delirium. In general, the older an elderly patient is, the less appropriate these agents are. Many drugs that are not typically recognized as anticholinergics may have potent anticholinergic activity, including tricyclic antidepressants, first-generation antihistamines (eg, diphenhydramine), and high-dose H2-receptor blockers (particularly cimetidine); these agents too should be avoided in elderly patients.12
Strategies to reduce postoperative delirium risk
How can we lower the risk of postoperative delirium in elderly hip fracture patients? Marcantonio et al13 randomized 126 patients undergoing hip fracture repair to receive usual care alone or supplemented with the following additional measures:
- Supplemental oxygen during surgery
- Optimization of electrolytes and blood glucose preoperatively
- Discontinuation of high-risk medications
- Adequate nutritional intake (by parenteral route if necessary)
- Encouragement to get out of bed on the first postoperative day
- Treatment of severe pain.
The incidence of delirium was reduced from 50% in the usual-care group to 32% in the intervention group, and the incidence of severe delirium was reduced even more, from 29% to 12%, respectively.13
OTHER BEST PRACTICES IN PERIOPERATIVE HIP FRACTURE MANAGEMENT
In a systematic literature review to identify best practices for perioperative management of elderly patients with hip fracture, Beaupre et al14 found the following measures to be among those with the strongest evidence of benefit:
- Use of spinal or local anesthesia rather than general anesthesia
- Use of pressure-relieving mattresses to prevent pressure ulcers
- Perioperative administration of antibiotics
- Deep vein thrombosis prophylaxis.
The review concluded that providing nutritional supplementation also is probably helpful although the evidence is not robust. Additionally, it was unclear whether minimizing the delay between hospital admission and surgery has any impact on mortality.14
Is early surgery better?
Early studies suggested that the sooner a hip fracture patient goes to surgery, the lower the mortality, but this has not been supported in well-controlled trials: no difference in mortality has been found whether the patient’s conditions are first optimized to reduce the risk of surgery or if the operation commences within 24 hours.
Although mortality does not appear to be affected, avoiding delay of hip fracture repair yields improvement in other outcomes. In a well-designed prospective cohort study, Orosz et al found that medically stable patients with hip fracture (mean age, 82 years) who underwent surgery within 24 hours had fewer days of pain and less intense pain postoperatively than those whose surgery was delayed beyond 24 hours.15 The early-surgery group also had a 1.94-day reduction in average length of stay compared with the late-surgery group.
A role for clinical pathways
To determine how the application of evidence-based perioperative practices affects actual outcomes in elderly hip fracture patients, Beaupre et al used a pre/post study design to evaluate the impact of an evidence-based clinical pathway at their institution.16 Though there were no differences in in-hospital mortality or the overall costs of inpatient care in elderly hip surgery patients before and after pathway implementation, the patients undergoing surgery after pathway implementation were significantly less likely to have postoperative delirium, heart failure, pressure ulcers, and urinary tract infections compared with those undergoing surgery before implementation. The outcomes benefits of this type of multimodal intervention are likely to extend to abdominal surgical procedures as well.
CASE CONTINUED: POSTOP DAY 2―PATIENT IS CONFUSED AND CRYING IN PAIN
On the second postoperative day, our patient appears weak and slightly confused. She is not eating and is crying in pain. Her neurological exam is normal.
Question: Which is the most appropriate next step?
A. Increase physical therapy
B. Begin an antidepressant
C. Insert a nasoenteric feeding tube
D. Increase doses of analgesics
The best answer is D. With no prior history of depression, an antidepressant would probably not be useful. It is premature to recommend nasoenteric feeding. Because pain hampers physical therapy, an increase in physical therapy would likewise be premature. Because we know the patient is in pain, the correct answer perhaps seems obvious. But keep in mind that relieving pain also has many other positive ramifications: intense pain can be a cause of delirium or at least worsen its symptoms, and pain relief is a prerequisite for the physical therapy that this patient needs.
Strategies for pain control
In general, the treatment of choice for postoperative pain is low-dose morphine sulfate (eg, 1–4 mg every 2 hours, titrated as needed). Acetaminophen can be given safely to virtually all patients. Patient-controlled analgesia is reasonable for select patients but not for older patients with cognitive impairment. Nonsteroidal anti-inflammatory drugs might be helpful in younger patients and even in robust elderly patients, but they must be used very cautiously in the older population because of the risk of gastric ulcers and bleeding, acute kidney injury, fluid retention, and exacerbation of congestive heart failure.
POSTOP DAY 3: PATIENT REPORTS LONG-STANDING FATIGUE
On postoperative day 3, the patient is weak and complains of fatigue. She says that before the fracture, she was experiencing mild weight loss, fatigue, and reduced activity.
Question: What is the most likely reason for her symptoms before the fracture?
A. Frailty
B. Occult heart failure
C. Adverse drug reaction to her beta-blocker
D. Clinical depression
The best answer is A. Occult heart failure is a reasonable second choice, as it is very common in older patients and the diagnosis is easy to miss unless florid pulmonary edema or associated symptoms (eg, chest pain) are present. But this patient had no history of heart disease and was only on medications for hypertension. An adverse drug reaction, such as to the beta-blocker, is unlikely and would probably not cause weight loss. The patient had no history of depression, so clinical depression is unlikely. That said, all the choices are reasonable to consider in elderly patients reporting fatigue and weakness.
Frailty is important to recognize
It is important to identify frailty and to aggressively manage frail patients postoperatively. Although frailty is not clearly defined, Fried et al17 identified five clinical features that correlate with its underlying pathophysiology:
- Minimal physical activity (ie, “doing less”)
- Generalized (not focal) muscle weakness
- Slowed performance (eg, walking short distances takes longer)
- Fatigue or poor endurance
- Unintentional weight loss.
The presence of three or more of these features meets the criteria for frailty and is associated with increased risk for mortality over the next 3 years with or without surgery,17 although surgery probably increases the risk.
Frailty is believed to be a failure over time of the homeostatic mechanisms that keep our organ systems functioning in the face of a stress. Decline in the ability of organ systems to maintain normal function is probably caused by inflammation, chronic disease, and normal aging, and has been termed homeostenosis. As a person ages and physiologic reserves are reduced, adding a stress such as surgery or severe infection can result in organ failure—usually multiple-system organ failure. In any intensive care unit, one is likely to see elderly patients who were admitted with one medical or surgical problem and soon end up having renal, liver, or brain dysfunction as well.
Physical therapy immediately after hip fracture surgery is associated with significantly better locomotion 2 months later.18 A number of exercises are effective: range-of-motion exercises, low-impact aerobic activities, and exercises starting with low-intensity resistance (using bands, tubes, and weights) and progressing as tolerated to high-intensity resistance (with machines and pulleys) for an extended period of time.
Nutrition supplementation
Malnourishment can contribute to frailty, yet evidence for the benefits of supplementing nutrition is not strong, as noted above. However, meta-analyses of studies of nutritional interventions with meal supplementation (usually canned supplements) show that meal supplementation can improve mortality risk and reduce morbidity such as pressure ulcers in hospitalized elderly patients.19,20 The patients most likely to benefit are those who are undernourished at baseline and aged 75 years or older.
CASE CONTINUED: WHAT HAPPENS POST-DISCHARGE?
Following surgery, our patient wonders, “Where will I go next? What will my lifestyle be like?”
These are important questions to consider when first evaluating whether an elderly patient should undergo surgery. In the case of hip fracture, standard thinking is that without surgery, the patient will never recover the ability to independently walk and perform activities of daily living. But we also must recognize the considerable risks of surgery in the elderly population, particularly those aged 75 years or older.
Comprehensive discharge planning
Early and intensive discharge management enhances quality of life and may help reduce hospital costs. A good model of care involves collaboration of orthopedic surgeons, hospitalists, general internists, geriatricians, and dietitians to address procedures, diet and nutrition, mobility and activities of daily living, and pain medications.21 A case manager such as a social worker should start addressing care transition the day after surgery—planning ahead is imperative.
Following hip surgery, patients are routinely sent to skilled nursing facilities as soon as possible so they can start intensive physical therapy. Patients with significant functional impairment or who had delirium are more likely to require a prolonged hospital stay.
Naylor et al examined the effectiveness of comprehensive discharge planning in a study that randomized hospitalized patients (including surgical patients) 65 years or older to either usual discharge planning or intensive discharge planning with advanced practice nurses beginning early in hospitalization.22 The intervention group was followed by home care nurses for up to 4 weeks and had continuous telephone access to the nurses. Patients who received the intervention had a significantly lower risk of hospital readmission, and those who were readmitted had significantly shorter hospital stays. The total cost of care was also significantly lower in the intervention group.
Family conferences aid decision making
Family conferences can be very useful for working through the many questions and challenges that surgery in an elderly person can pose, including whether the patient should undergo the operation, postoperative management, and postdischarge placement.23 For patients with an uncertain prognosis because of unclear or multiple concurrent diseases, a family conference can help clarify the goals of therapy, inform the family about likely outcomes, and help determine the patient’s wishes and values. Such issues should be revisited as the postoperative course proceeds.
Family conferences also provide a good opportunity to review advanced directives, the need for life support, and possible transfers to intensive care. Family conferences can also help resolve conflicts in care management, as family members may not agree with the need for surgery, how aggressive treatment should be, or where to send the patient for rehabilitation. Differences among family members on these questions are especially common with elderly patients. Working out such issues will improve patient care, especially when done early in the hospitalization.
DISCUSSION
Question from the audience: In our preoperative clinic, we are trying to intervene to reduce delirium and postoperative cognitive dysfunction. How can we quickly screen for the most important predictors and act to reduce the risk?
Dr. Palmer: The most important risk factor for delirium is age, which obviously can’t be changed. Ask patients about alcohol use and depression. Check on nutritional status and begin supplementation if indicated. Discontinue high-risk medications. Check on electrolytes and their state of hydration; ideally, an electrolyte imbalance can be corrected preoperatively. In addition, other than in patients with end-stage renal disease, try to keep the hemoglobin above 7.5 g/dL, which appears to be associated with better outcomes and less risk of delirium.
It’s also important to remind the family to bring in the patient’s visual aids, hearing devices, and cane or walker so that they’re available right after the operation.
Intraoperative factors that are important for preventing delirium include maintaining good blood pressure levels, giving supplemental oxygen, minimizing the time under general anesthesia, and using local anesthesia if possible.
Question from the audience: How strong is the evidence for using spinal anesthesia as opposed to general anesthesia in preventing postoperative cognitive dysfunction and delirium, especially in the setting of hip fracture repair?
Dr. Palmer: The evidence is fairly soft. For patients undergoing either hip or knee arthroplasty who were randomized to receive either spinal (or local) or general anesthesia, the risk of delirium was similar, but complications such as prolonged bed rest, pressure ulcers, and catheter-related urinary tract infections were somewhat reduced in the spinal/local group.14 The relative risk of developing postoperative cognitive dysfunction is unclear—no randomized controlled trials have been conducted to answer that question.
Question from the audience: How do you use antipsychotic drugs, especially with the concerns from epidemiologic studies about an increased risk of death?
Dr. Palmer: No antipsychotic agents, including haloperidol, have a specific Food and Drug Administration–approved indication for treating agitation, dementia, or delirium. In general, they should not be used without a clear indication. That said, the usual off-label use is for patients who are severely agitated and are at risk of harming themselves or others. In an ICU setting, where patients have multiple lines, the use of these agents can be considered for a very agitated patient. Alternatives exist, but antipsychotics like haloperidol have the advantage that they can be given in small increments very rapidly and achieve good control of severe agitation.
Antipsychotic agents should only be used with great caution and for the shortest duration needed. As delirium resolves, they should be tapered fairly rapidly over a few days and ideally should be discontinued by the time of hospital discharge.
None of the antipsychotic agents—including those in the first generation and the newer atypical agents—is free of this risk of increased mortality. The mechanism is not understood; it may be torsades de pointes or hypotension leading to stroke or sudden cardiac death.
Question from the audience: What is the most efficient way to assess cognitive and physical functioning preoperatively?
Dr. Palmer: There may be a documented history of dementia, or family members may tell you if there has been memory loss or some decline in the patient’s self-care abilities. For patients without dementia, you can ask them directly if they can perform basic activities of daily living, such as getting out of bed or dressing. To assess higher-level function, ask if they can manage their own medications, pay bills, or handle their finances. If not, they might have cognitive impairment and are at higher risk for postoperative delirium. These are rather sensitive measures. There are instruments to assess this more precisely, but few clinicians have time to use them.
Quick bedside tests can help assess for delirium postoperatively. We see if patients are “alert and oriented times three” (“Do you know who you are, where you are, and the date?”). We test for attention by asking them to repeat a random string of numbers spoken 1 second apart in monotone; people who are delirious and many patients with severe dementia can’t repeat more than three numbers. A patient who is alert and oriented, has a good attention span (more than three numbers in correct order), and has no history of dementia probably doesn’t have delirium or dementia.
For physical function, ask if they can walk, get out of bed to a chair, and ambulate. If they don’t give clear answers, observe them get out of bed or a chair, walk 10 feet, and return to bed. If they can do that with good balance, especially within 10 to 15 seconds, they probably have reasonably normal mobility and are at lower risk for postoperative complications such as falls with injury.
Acute hospital care is fast becoming acute geriatric care: people aged 65 years or older are only 13% of the population but account for 44% of days of care in nonfederal hospitals and 38% of discharges.1 In general, the elderly have longer hospital stays, incur greater costs, and have a higher risk of adverse outcomes than do their younger counterparts.2
Among the most common surgical procedures for patients older than 65 are percutaneous coronary intervention with stenting, coronary artery bypass graft surgery, and open reduction internal fixation for hip fracture; the latter is the most common operation in patients aged 85 years or older.3
Elderly patients frequently pose many challenges perioperatively that are not often seen in younger patients. Dementia, frailty, impaired ability to care for oneself, and malnourishment may be present at baseline and are likely to worsen postoperatively. The elderly are at increased risk of acute delirium and cognitive impairment postoperatively, which often complicates recovery and discharge placement.
This article uses a case study to review perioperative problems commonly encountered in elderly surgical patients, particularly those undergoing hip surgery. As the case is presented, I will review strategies to assess risks and prevent and mitigate postoperative cognitive dysfunction and other barriers to recovery.
CASE: AN 82-YEAR-OLD WOMAN WITH HIP FRACTURE
An 82-year-old woman is admitted to undergo open reduction internal fixation for hip fracture. She has a history of osteoarthritis, systolic hypertension, and visual impairment (20/70). Her medications include a beta-blocker, a thiazide diuretic, analgesics as needed, and a multivitamin. She was independent in all activities of daily living before the fracture. She is a social drinker and does not smoke. She has no known cardiovascular, lung, or renal disease.
Her laboratory test results are as follows:
- Blood urea nitrogen (BUN), 24 mg/dL
- Creatinine, 1.0 mg/dL
- Hemoglobin, 12.8 g/dL
- Albumin, 3.8 gm/dL
- Normal levels of thyroid-stimulating hormone and vitamin B12.
Thus, her lab results are normal except for the BUN:creatinine ratio being a bit high, at 24:1 (normal is 10:1, with ratios greater than 18:1 being associated with an increased risk of delirium4).
ASSESSING COGNITIVE RISK: POSTOPERATIVE COGNITIVE DYSFUNCTION VS DELIRIUM
Question: Which of these statements about this patient is most correct?
A. She is at high risk (> 40%) of postoperative cognitive dysfunction
B. Her risk of postoperative delirium is 5% to 10%
C. Postoperative delirium cannot be prevented
D. Preoperative haloperidol (1.5 mg/day for 3 days) will reduce the risk of delirium by 25%
The best answer is A. Postoperative cognitive dysfunction is different from delirium, though it is part of a spectrum of cognitive impairment that may occur after surgery and even persist for a prolonged period. The patient’s risk of postoperative delirium is actually a bit higher than 10% (see “Estimating the risk of delirium” below). Some evidence shows that postoperative delirium can be prevented, at least in hip fracture patients. Kalisvaart et al found that preoperative treatment with low-dose haloperidol reduced the duration and severity of delirium in elderly patients following hip surgery but did not reduce its incidence.5
Cognitive dysfunction often follows surgery
Postoperative cognitive dysfunction has long been recognized and was first described in patients after cardiac surgery, especially following coronary artery bypass graft procedures. In the last several years, we have recognized that it also occurs in patients who undergo noncardiac surgery. Postoperative cognitive dysfunction, which may persist for weeks to months, may not be obvious but can be detected by standard neuropsychological testing.6
Postoperative cognitive dysfunction is different from the “emergence delirium” that may immediately follow surgery and that is often associated with the wearing off of anesthesia. It is also distinct from “incident delirium,” which sometimes occurs over the first few postoperative days (discussed below).
Postoperative dysfunction is especially persistent in the elderly
A recent study found cognitive dysfunction to be common at hospital discharge after major noncardiac surgery in adults of all ages: rates at discharge were 36.6% in patients aged 18 to 39 years, 30.4% in those aged 40 to 59, and 41.4% in those 60 or older.7 Notably, however, the oldest group was most likely to have persistent symptoms. Three months after surgery, 12.7% of patients aged 60 or older continued to have postoperative cognitive dysfunction, which was more than double the rates in the young and middle-aged patient groups (5.7% and 5.6%, respectively).7
Although the cause of postoperative cognitive dysfunction is not well understood, predisposing factors in addition to advanced age include metabolic problems, lower educational level, and previous cerebral vascular accident.7 When elective surgery is considered by elderly patients, the decision should take into account their risk of postoperative cognitive dysfunction and the impact it may have on their quality of life.
PREDICTING AND PREVENTING DELIRIUM
Delirium is easily recognized
Delirium is a common complication of surgery. Unlike postoperative cognitive dysfunction, delirium is easy to detect clinically. It is a disorder of attention and cognition and classically presents as an acute change in mental status accompanied by the following8:
- Fluctuation in awareness
- Memory impairment
- Inattention (inability to stay on task, distractibility)
- Disorganized or illogical thinking
- Altered level of consciousness—ie, hyperalertness (agitation, pulling out intravenous lines, etc) or hypoalertness (“quiet delirium”).
Estimating the risk of delirium
Marcantonio and colleagues developed a model to predict the likelihood that delirium will develop in patients undergoing elective surgery.9 The model assigns points to various risk factors as follows:
- Age ≥ 70 years (1 point)
- History of alcohol abuse (1 point)
- Baseline cognitive impairment (1 point)
- Severe physical impairment (reduced ability to walk or perform daily activities) (1 point)
- Abnormal preoperative blood levels of electrolytes or glucose (1 point)
- Noncardiac thoracic surgery (1 point)
- Abdominal aortic aneurysm surgery (2 points).
The study to validate this model found that a score of 0 points is associated with only a 2% risk of developing postoperative delirium. A score of 3 or more points is associated with a 50% risk of postoperative delirium. A score of 1 or 2 points (as for the patient in our case study) is associated with an 11% risk, according to this Marcantonio model.9
Additionally, well-designed cohort studies of medical patients10 have identified four major independent predictors of incident delirium:
- Severe illness (eg, high fever, complicated infections)
- Baseline dementia
- Dehydration (high BUN:creatinine ratio)
- Sensory impairments (particularly visual).
Kalisvaart et al conducted a prospective cohort study to determine whether these risk factors in medical patients are applicable to elderly patients undergoing hip surgery.11 They found that the incidence of delirium was low (4%) in hip surgery patients with none of these factors, increased to 11% in patients with one or two of these factors, and increased to 37% in patients with three or four factors. These findings suggest that hip surgery patients (like our case patient) may be at greater risk of postoperative delirium than is reflected in the Marcantonio model discussed above,9 which was validated in a study of patients undergoing elective (not emergent) surgery.
Several drug classes raise dementia risk
Anticholinergic medications and other drugs with anticholinergic properties, ie, benzodiazepines and the opioid agent meperidine, also raise the risk for delirium. In general, the older an elderly patient is, the less appropriate these agents are. Many drugs that are not typically recognized as anticholinergics may have potent anticholinergic activity, including tricyclic antidepressants, first-generation antihistamines (eg, diphenhydramine), and high-dose H2-receptor blockers (particularly cimetidine); these agents too should be avoided in elderly patients.12
Strategies to reduce postoperative delirium risk
How can we lower the risk of postoperative delirium in elderly hip fracture patients? Marcantonio et al13 randomized 126 patients undergoing hip fracture repair to receive usual care alone or supplemented with the following additional measures:
- Supplemental oxygen during surgery
- Optimization of electrolytes and blood glucose preoperatively
- Discontinuation of high-risk medications
- Adequate nutritional intake (by parenteral route if necessary)
- Encouragement to get out of bed on the first postoperative day
- Treatment of severe pain.
The incidence of delirium was reduced from 50% in the usual-care group to 32% in the intervention group, and the incidence of severe delirium was reduced even more, from 29% to 12%, respectively.13
OTHER BEST PRACTICES IN PERIOPERATIVE HIP FRACTURE MANAGEMENT
In a systematic literature review to identify best practices for perioperative management of elderly patients with hip fracture, Beaupre et al14 found the following measures to be among those with the strongest evidence of benefit:
- Use of spinal or local anesthesia rather than general anesthesia
- Use of pressure-relieving mattresses to prevent pressure ulcers
- Perioperative administration of antibiotics
- Deep vein thrombosis prophylaxis.
The review concluded that providing nutritional supplementation also is probably helpful although the evidence is not robust. Additionally, it was unclear whether minimizing the delay between hospital admission and surgery has any impact on mortality.14
Is early surgery better?
Early studies suggested that the sooner a hip fracture patient goes to surgery, the lower the mortality, but this has not been supported in well-controlled trials: no difference in mortality has been found whether the patient’s conditions are first optimized to reduce the risk of surgery or if the operation commences within 24 hours.
Although mortality does not appear to be affected, avoiding delay of hip fracture repair yields improvement in other outcomes. In a well-designed prospective cohort study, Orosz et al found that medically stable patients with hip fracture (mean age, 82 years) who underwent surgery within 24 hours had fewer days of pain and less intense pain postoperatively than those whose surgery was delayed beyond 24 hours.15 The early-surgery group also had a 1.94-day reduction in average length of stay compared with the late-surgery group.
A role for clinical pathways
To determine how the application of evidence-based perioperative practices affects actual outcomes in elderly hip fracture patients, Beaupre et al used a pre/post study design to evaluate the impact of an evidence-based clinical pathway at their institution.16 Though there were no differences in in-hospital mortality or the overall costs of inpatient care in elderly hip surgery patients before and after pathway implementation, the patients undergoing surgery after pathway implementation were significantly less likely to have postoperative delirium, heart failure, pressure ulcers, and urinary tract infections compared with those undergoing surgery before implementation. The outcomes benefits of this type of multimodal intervention are likely to extend to abdominal surgical procedures as well.
CASE CONTINUED: POSTOP DAY 2―PATIENT IS CONFUSED AND CRYING IN PAIN
On the second postoperative day, our patient appears weak and slightly confused. She is not eating and is crying in pain. Her neurological exam is normal.
Question: Which is the most appropriate next step?
A. Increase physical therapy
B. Begin an antidepressant
C. Insert a nasoenteric feeding tube
D. Increase doses of analgesics
The best answer is D. With no prior history of depression, an antidepressant would probably not be useful. It is premature to recommend nasoenteric feeding. Because pain hampers physical therapy, an increase in physical therapy would likewise be premature. Because we know the patient is in pain, the correct answer perhaps seems obvious. But keep in mind that relieving pain also has many other positive ramifications: intense pain can be a cause of delirium or at least worsen its symptoms, and pain relief is a prerequisite for the physical therapy that this patient needs.
Strategies for pain control
In general, the treatment of choice for postoperative pain is low-dose morphine sulfate (eg, 1–4 mg every 2 hours, titrated as needed). Acetaminophen can be given safely to virtually all patients. Patient-controlled analgesia is reasonable for select patients but not for older patients with cognitive impairment. Nonsteroidal anti-inflammatory drugs might be helpful in younger patients and even in robust elderly patients, but they must be used very cautiously in the older population because of the risk of gastric ulcers and bleeding, acute kidney injury, fluid retention, and exacerbation of congestive heart failure.
POSTOP DAY 3: PATIENT REPORTS LONG-STANDING FATIGUE
On postoperative day 3, the patient is weak and complains of fatigue. She says that before the fracture, she was experiencing mild weight loss, fatigue, and reduced activity.
Question: What is the most likely reason for her symptoms before the fracture?
A. Frailty
B. Occult heart failure
C. Adverse drug reaction to her beta-blocker
D. Clinical depression
The best answer is A. Occult heart failure is a reasonable second choice, as it is very common in older patients and the diagnosis is easy to miss unless florid pulmonary edema or associated symptoms (eg, chest pain) are present. But this patient had no history of heart disease and was only on medications for hypertension. An adverse drug reaction, such as to the beta-blocker, is unlikely and would probably not cause weight loss. The patient had no history of depression, so clinical depression is unlikely. That said, all the choices are reasonable to consider in elderly patients reporting fatigue and weakness.
Frailty is important to recognize
It is important to identify frailty and to aggressively manage frail patients postoperatively. Although frailty is not clearly defined, Fried et al17 identified five clinical features that correlate with its underlying pathophysiology:
- Minimal physical activity (ie, “doing less”)
- Generalized (not focal) muscle weakness
- Slowed performance (eg, walking short distances takes longer)
- Fatigue or poor endurance
- Unintentional weight loss.
The presence of three or more of these features meets the criteria for frailty and is associated with increased risk for mortality over the next 3 years with or without surgery,17 although surgery probably increases the risk.
Frailty is believed to be a failure over time of the homeostatic mechanisms that keep our organ systems functioning in the face of a stress. Decline in the ability of organ systems to maintain normal function is probably caused by inflammation, chronic disease, and normal aging, and has been termed homeostenosis. As a person ages and physiologic reserves are reduced, adding a stress such as surgery or severe infection can result in organ failure—usually multiple-system organ failure. In any intensive care unit, one is likely to see elderly patients who were admitted with one medical or surgical problem and soon end up having renal, liver, or brain dysfunction as well.
Physical therapy immediately after hip fracture surgery is associated with significantly better locomotion 2 months later.18 A number of exercises are effective: range-of-motion exercises, low-impact aerobic activities, and exercises starting with low-intensity resistance (using bands, tubes, and weights) and progressing as tolerated to high-intensity resistance (with machines and pulleys) for an extended period of time.
Nutrition supplementation
Malnourishment can contribute to frailty, yet evidence for the benefits of supplementing nutrition is not strong, as noted above. However, meta-analyses of studies of nutritional interventions with meal supplementation (usually canned supplements) show that meal supplementation can improve mortality risk and reduce morbidity such as pressure ulcers in hospitalized elderly patients.19,20 The patients most likely to benefit are those who are undernourished at baseline and aged 75 years or older.
CASE CONTINUED: WHAT HAPPENS POST-DISCHARGE?
Following surgery, our patient wonders, “Where will I go next? What will my lifestyle be like?”
These are important questions to consider when first evaluating whether an elderly patient should undergo surgery. In the case of hip fracture, standard thinking is that without surgery, the patient will never recover the ability to independently walk and perform activities of daily living. But we also must recognize the considerable risks of surgery in the elderly population, particularly those aged 75 years or older.
Comprehensive discharge planning
Early and intensive discharge management enhances quality of life and may help reduce hospital costs. A good model of care involves collaboration of orthopedic surgeons, hospitalists, general internists, geriatricians, and dietitians to address procedures, diet and nutrition, mobility and activities of daily living, and pain medications.21 A case manager such as a social worker should start addressing care transition the day after surgery—planning ahead is imperative.
Following hip surgery, patients are routinely sent to skilled nursing facilities as soon as possible so they can start intensive physical therapy. Patients with significant functional impairment or who had delirium are more likely to require a prolonged hospital stay.
Naylor et al examined the effectiveness of comprehensive discharge planning in a study that randomized hospitalized patients (including surgical patients) 65 years or older to either usual discharge planning or intensive discharge planning with advanced practice nurses beginning early in hospitalization.22 The intervention group was followed by home care nurses for up to 4 weeks and had continuous telephone access to the nurses. Patients who received the intervention had a significantly lower risk of hospital readmission, and those who were readmitted had significantly shorter hospital stays. The total cost of care was also significantly lower in the intervention group.
Family conferences aid decision making
Family conferences can be very useful for working through the many questions and challenges that surgery in an elderly person can pose, including whether the patient should undergo the operation, postoperative management, and postdischarge placement.23 For patients with an uncertain prognosis because of unclear or multiple concurrent diseases, a family conference can help clarify the goals of therapy, inform the family about likely outcomes, and help determine the patient’s wishes and values. Such issues should be revisited as the postoperative course proceeds.
Family conferences also provide a good opportunity to review advanced directives, the need for life support, and possible transfers to intensive care. Family conferences can also help resolve conflicts in care management, as family members may not agree with the need for surgery, how aggressive treatment should be, or where to send the patient for rehabilitation. Differences among family members on these questions are especially common with elderly patients. Working out such issues will improve patient care, especially when done early in the hospitalization.
DISCUSSION
Question from the audience: In our preoperative clinic, we are trying to intervene to reduce delirium and postoperative cognitive dysfunction. How can we quickly screen for the most important predictors and act to reduce the risk?
Dr. Palmer: The most important risk factor for delirium is age, which obviously can’t be changed. Ask patients about alcohol use and depression. Check on nutritional status and begin supplementation if indicated. Discontinue high-risk medications. Check on electrolytes and their state of hydration; ideally, an electrolyte imbalance can be corrected preoperatively. In addition, other than in patients with end-stage renal disease, try to keep the hemoglobin above 7.5 g/dL, which appears to be associated with better outcomes and less risk of delirium.
It’s also important to remind the family to bring in the patient’s visual aids, hearing devices, and cane or walker so that they’re available right after the operation.
Intraoperative factors that are important for preventing delirium include maintaining good blood pressure levels, giving supplemental oxygen, minimizing the time under general anesthesia, and using local anesthesia if possible.
Question from the audience: How strong is the evidence for using spinal anesthesia as opposed to general anesthesia in preventing postoperative cognitive dysfunction and delirium, especially in the setting of hip fracture repair?
Dr. Palmer: The evidence is fairly soft. For patients undergoing either hip or knee arthroplasty who were randomized to receive either spinal (or local) or general anesthesia, the risk of delirium was similar, but complications such as prolonged bed rest, pressure ulcers, and catheter-related urinary tract infections were somewhat reduced in the spinal/local group.14 The relative risk of developing postoperative cognitive dysfunction is unclear—no randomized controlled trials have been conducted to answer that question.
Question from the audience: How do you use antipsychotic drugs, especially with the concerns from epidemiologic studies about an increased risk of death?
Dr. Palmer: No antipsychotic agents, including haloperidol, have a specific Food and Drug Administration–approved indication for treating agitation, dementia, or delirium. In general, they should not be used without a clear indication. That said, the usual off-label use is for patients who are severely agitated and are at risk of harming themselves or others. In an ICU setting, where patients have multiple lines, the use of these agents can be considered for a very agitated patient. Alternatives exist, but antipsychotics like haloperidol have the advantage that they can be given in small increments very rapidly and achieve good control of severe agitation.
Antipsychotic agents should only be used with great caution and for the shortest duration needed. As delirium resolves, they should be tapered fairly rapidly over a few days and ideally should be discontinued by the time of hospital discharge.
None of the antipsychotic agents—including those in the first generation and the newer atypical agents—is free of this risk of increased mortality. The mechanism is not understood; it may be torsades de pointes or hypotension leading to stroke or sudden cardiac death.
Question from the audience: What is the most efficient way to assess cognitive and physical functioning preoperatively?
Dr. Palmer: There may be a documented history of dementia, or family members may tell you if there has been memory loss or some decline in the patient’s self-care abilities. For patients without dementia, you can ask them directly if they can perform basic activities of daily living, such as getting out of bed or dressing. To assess higher-level function, ask if they can manage their own medications, pay bills, or handle their finances. If not, they might have cognitive impairment and are at higher risk for postoperative delirium. These are rather sensitive measures. There are instruments to assess this more precisely, but few clinicians have time to use them.
Quick bedside tests can help assess for delirium postoperatively. We see if patients are “alert and oriented times three” (“Do you know who you are, where you are, and the date?”). We test for attention by asking them to repeat a random string of numbers spoken 1 second apart in monotone; people who are delirious and many patients with severe dementia can’t repeat more than three numbers. A patient who is alert and oriented, has a good attention span (more than three numbers in correct order), and has no history of dementia probably doesn’t have delirium or dementia.
For physical function, ask if they can walk, get out of bed to a chair, and ambulate. If they don’t give clear answers, observe them get out of bed or a chair, walk 10 feet, and return to bed. If they can do that with good balance, especially within 10 to 15 seconds, they probably have reasonably normal mobility and are at lower risk for postoperative complications such as falls with injury.
- DeFrances CJ, Cullen KA, Kozak LJ. National Hospital Discharge Survey: 2005 annual summary with detailed diagnosis and procedure data. Vital Health Stat (Series 13) 2007; Dec(165):1–209.
- Welcome to HCUPnet: a tool for identifying, tracking, and analyzing national hospital statistics. Agency for Healthcare Research and Quality Web site. http://hcupnet.ahrq.gov. Accessed February 23, 2009.
- Kozak LJ, Owings MF, Hall MJ. National Hospital Discharge Survey: 2002 annual summary with detailed diagnosis and procedure data. Vital Health Stat (Series 13) 2005; Mar(158):1–199.
- Inouye SK, Viscoli CM, Horwitz RI, Hurst LD, Tinetti ME. A predictive model for delirium in hospitalized elderly medical patients based on admission characteristics. Ann Intern Med 1993; 119:474–481.
- Kalisvaart KJ, de Jonghe JF, Bogaards MJ, et al. Haloperidol prophylaxis for elderly hip-surgery patients at risk for delirium: a randomized placebo-controlled study. J Am Geriatr Soc 2005; 53:1658–1666.
- Silverstein JH, Timberger M, Reich DL, Uysal S. Central nervous system dysfunction after noncardiac surgery and anesthesia in the elderly. Anesthesiology 2007; 106:622–628.
- Monk TG, Weldon BC, Garvan CW, et al. Predictors of cognitive dysfunction after major noncardiac surgery. Anesthesiology 2008; 108:18–30.
- Inouye SK. Delirium in older persons. N Engl J Med 2006; 354:1157–1165.
- Marcantonio ER, Goldman L, Mangione CM, et al. A clinical prediction rule for delirium after elective noncardiac surgery. JAMA 1994; 271:134–139.
- Inouye SK. Predisposing and precipitating factors for delirium in hospitalized older patients. Dement Geriatr Cogn Disord 1999; 10:393–400.
- Kalisvaart KJ, Vreeswijk R, de Jonghe JF, et al. Risk factors and prediction of postoperative delirium in elderly hip-surgery patients: implementation and validation of a medical risk factor model. J Am Geriatr Soc 2006; 54:817–822.
- Fick DM, Cooper JW, Wade WE, et al. Updating the Beers criteria for potentially inappropriate medication use in older adults: results of a US consensus panel of experts. Arch Intern Med 2003; 163:2716–2724.
- Marcantonio ER, Flacker JM, Wright RJ, et al. Reducing delirium after hip fracture: a randomized trial. J Am Geriatr Soc 2001; 49:516–522.
- Beaupre LA, Jones CA, Saunders LD, Johnston DW, Buckingham J, Majumdar SR. Best practices for elderly hip fracture patients: a systematic overview of the evidence. J Gen Intern Med 2005; 20:1019–1025.
- Orosz GM, Magaziner J, Hannan EL, et al. Association of timing of surgery for hip fracture and patient outcomes. JAMA 2004; 291:1738–1743.
- Beaupre LA, Cinats JG, Senthilselvan A, et al. Reduced morbidity for elderly patients with a hip fracture after implementation of a perioperative evidence-based clinical pathway. Qual Saf Health Care 2006; 15:375–379.
- Fried LP, Tangen CM, Walston J, et al. Frailty in older adults: evidence for a phenotype. J Gerontol A Biol Sci Med Sci 2001; 56:M146–M156.
- Penrod JD, Boockvar KS, Litke A, et al. Physical therapy and mobility 2 and 6 months after hip fracture. J Am Geriatr Soc 2004; 52:1114–1120.
- Milne AC, Avenell A, Potter J. Meta-analysis: protein and energy supplementation in older people. Ann Intern Med 2006; 144:37–48.
- Avenell A, Handoll HH. Nutritional supplementation for hip fracture aftercare in older people. Cochrane Database Syst Rev 2006; (4):CD001880.
- Miura LN, DiPiero AR, Homer LD. Effects of a geriatrician-led hip fracture program: improvements in clinical and economic outcomes. J Am Geriatr Soc 2009; 57:159–167.
- Naylor MD, Brooten D, Campbell R, et al. Comprehensive discharge planning and home follow-up of hospitalized elders: a randomized clinical trial. JAMA 1999; 281:613–620.
- Palmer RM. Acute hospital care of the elderly: making a difference. Hospitalist 2004; (suppl):4–7.
- DeFrances CJ, Cullen KA, Kozak LJ. National Hospital Discharge Survey: 2005 annual summary with detailed diagnosis and procedure data. Vital Health Stat (Series 13) 2007; Dec(165):1–209.
- Welcome to HCUPnet: a tool for identifying, tracking, and analyzing national hospital statistics. Agency for Healthcare Research and Quality Web site. http://hcupnet.ahrq.gov. Accessed February 23, 2009.
- Kozak LJ, Owings MF, Hall MJ. National Hospital Discharge Survey: 2002 annual summary with detailed diagnosis and procedure data. Vital Health Stat (Series 13) 2005; Mar(158):1–199.
- Inouye SK, Viscoli CM, Horwitz RI, Hurst LD, Tinetti ME. A predictive model for delirium in hospitalized elderly medical patients based on admission characteristics. Ann Intern Med 1993; 119:474–481.
- Kalisvaart KJ, de Jonghe JF, Bogaards MJ, et al. Haloperidol prophylaxis for elderly hip-surgery patients at risk for delirium: a randomized placebo-controlled study. J Am Geriatr Soc 2005; 53:1658–1666.
- Silverstein JH, Timberger M, Reich DL, Uysal S. Central nervous system dysfunction after noncardiac surgery and anesthesia in the elderly. Anesthesiology 2007; 106:622–628.
- Monk TG, Weldon BC, Garvan CW, et al. Predictors of cognitive dysfunction after major noncardiac surgery. Anesthesiology 2008; 108:18–30.
- Inouye SK. Delirium in older persons. N Engl J Med 2006; 354:1157–1165.
- Marcantonio ER, Goldman L, Mangione CM, et al. A clinical prediction rule for delirium after elective noncardiac surgery. JAMA 1994; 271:134–139.
- Inouye SK. Predisposing and precipitating factors for delirium in hospitalized older patients. Dement Geriatr Cogn Disord 1999; 10:393–400.
- Kalisvaart KJ, Vreeswijk R, de Jonghe JF, et al. Risk factors and prediction of postoperative delirium in elderly hip-surgery patients: implementation and validation of a medical risk factor model. J Am Geriatr Soc 2006; 54:817–822.
- Fick DM, Cooper JW, Wade WE, et al. Updating the Beers criteria for potentially inappropriate medication use in older adults: results of a US consensus panel of experts. Arch Intern Med 2003; 163:2716–2724.
- Marcantonio ER, Flacker JM, Wright RJ, et al. Reducing delirium after hip fracture: a randomized trial. J Am Geriatr Soc 2001; 49:516–522.
- Beaupre LA, Jones CA, Saunders LD, Johnston DW, Buckingham J, Majumdar SR. Best practices for elderly hip fracture patients: a systematic overview of the evidence. J Gen Intern Med 2005; 20:1019–1025.
- Orosz GM, Magaziner J, Hannan EL, et al. Association of timing of surgery for hip fracture and patient outcomes. JAMA 2004; 291:1738–1743.
- Beaupre LA, Cinats JG, Senthilselvan A, et al. Reduced morbidity for elderly patients with a hip fracture after implementation of a perioperative evidence-based clinical pathway. Qual Saf Health Care 2006; 15:375–379.
- Fried LP, Tangen CM, Walston J, et al. Frailty in older adults: evidence for a phenotype. J Gerontol A Biol Sci Med Sci 2001; 56:M146–M156.
- Penrod JD, Boockvar KS, Litke A, et al. Physical therapy and mobility 2 and 6 months after hip fracture. J Am Geriatr Soc 2004; 52:1114–1120.
- Milne AC, Avenell A, Potter J. Meta-analysis: protein and energy supplementation in older people. Ann Intern Med 2006; 144:37–48.
- Avenell A, Handoll HH. Nutritional supplementation for hip fracture aftercare in older people. Cochrane Database Syst Rev 2006; (4):CD001880.
- Miura LN, DiPiero AR, Homer LD. Effects of a geriatrician-led hip fracture program: improvements in clinical and economic outcomes. J Am Geriatr Soc 2009; 57:159–167.
- Naylor MD, Brooten D, Campbell R, et al. Comprehensive discharge planning and home follow-up of hospitalized elders: a randomized clinical trial. JAMA 1999; 281:613–620.
- Palmer RM. Acute hospital care of the elderly: making a difference. Hospitalist 2004; (suppl):4–7.
KEY POINTS
- Postoperative cognitive dysfunction and delirium are distinct conditions, though both are common in the elderly. Postoperative cognitive dysfunction may persist for weeks to months and may not be obvious, whereas delirium, a disorder of attention and cognition, is easier to detect clinically.
- Major predictors of postoperative delirium are severe illness, baseline dementia, dehydration, and sensory impairment.
- Drugs that raise dementia risk include anticholinergics, benzodiazepines, meperidine, tricyclic antidepressants, first-generation antihistamines, and high-dose H2-receptor blockers.
- Early performance of hip fracture surgery in the elderly (ie, within 24 hours of admission) has not been shown to lower mortality but appears to improve other outcomes.
- Identifying and managing frail elderly patients is important. Signs of frailty are minimal activity, generalized muscle weakness, slowed performance, fatigue, and weight loss.
The role of testing in the preoperative evaluation
Routine presurgical assessment of patients with a standard battery of tests not only is wasteful but can lead to more unnecessary expense, delay, and even risk to the patient and physician. Any abnormal tests results, even if likely to be clinically unimportant to the upcoming surgery, will need to be followed up to rule out a significant abnormality that may have later implications. This review will outline strategies for making decisions about which tests are clinically useful for preoperative assessment of a given patient and also discuss the value of preoperative evaluation centers in promoting appropriate preoperative testing.
PREOPERATIVE EVALUATION SHOULD BE CLEARLY DIRECTED
Most patients scheduled for surgery at Brigham and Women’s Hospital are assessed by the staff at our preoperative evaluation center. We take a medical history and conduct a physical examination, review the medical records, order laboratory tests or other studies as indicated, and determine which patients need further work-up or consultations. The goals are to evaluate patient readiness for anesthesia and surgery, optimize patient health before surgery, enhance the quality of perioperative care, reduce the morbidity of surgery and length of stay, and return the patient to normal functioning.1,2
The above goals are generally achieved by directed laboratory testing, managing the patient’s medications, stabilizing disorders when possible, and creating plans for postoperative care and pain management. Communication among the surgeon, the anesthesiology team, and the preoperative medical consultant (if there is one) is critical.1,2
In contrast, “clearing the patient for surgery” is not a legitimate goal of consultation. The real issues to be taken up in a consult are:
- What is the patient’s risk of complications (cardiac and noncardiac)?
- Would further risk stratification alter patient management?
- Can anything be done to reduce the patient’s risk?
If indicated, a consult should cover the entire perioperative period, offering opinions on operative risk and suggesting treatments that affect long-term patient outcomes. Rarely is preoperative intervention necessary just to lower the risk of surgery. Most interventions that are needed should be done regardless of the surgery.
Everyone on the medical team should have the goal of efficient resource utilization, including avoidance of unnecessary visits, laboratory testing, and consultations.
PREOPERATIVE TESTING: WHAT IS NEEDED?
Preoperative testing is extremely expensive: even more than 20 years ago, preoperative medical testing for all types of surgery accounted for approximately $30 billion in US health care costs annually.3 The likelihood of abnormal test results increases with age, and the more tests performed, the more likely a false positive will occur, further driving up costs.
Preoperative testing should generally be directed by a targeted history and physical examination, and the relevance of any tests should be considered in light of the type of procedure that is planned, particularly the hemodynamic changes and blood loss involved. Before ordering a test, physicians should be sure that there is a good reason for the test, that it is consistent with established guidelines, and that the results will be useful (ie, have the potential to change management).
Case study: Inguinal surgery in a healthy elderly man
A 72-year-old man is being evaluated prior to a right inguinal herniorrhaphy. He has osteoarthritis but is otherwise healthy and jogs 3 to 5 miles several times a week. He takes no medications and has no known drug allergies.
Question: Which of the following tests is necessary prior to surgery?
A. Complete blood cell count (CBC)
B. Prothrombin time and partial thromboplastin time
C. Electrocardiogram (ECG)
D. All of the above
E. None of the above
The correct answer is E (none of the above), for the reasons laid out in the following section.
Unnecessary testing may cause more harm than good
Untargeted testing should be avoided. An unexpected result will probably not be clinically significant for the surgery and will only lead to more needless testing, unnecessary anxiety for the patient, and delays in proceeding to the operating room.4 The more tests that are ordered, the higher the likelihood of having an abnormal result by chance: for a test with 95% specificity, results for 1 out of 13 ordered tests will likely be abnormal without there being a true underlying physiologic abnormality.
Researchers at Johns Hopkins University assessed the value of routine preoperative medical testing in a randomized study of nearly 20,000 patients undergoing elective cataract surgery whose preoperative history and physical examination was either preceded or not preceded by a standard battery of tests, including ECG, CBC, electrolytes, urea nitrogen, creatinine, and glucose.5 This was an ideal study population, given the relatively noninvasive nature of the procedure (with minimal hemodynamic changes) and cataract patients’ relatively advanced age and resulting likelihood of comorbidities. Notably, there were no differences between the two groups in the overall rate of complications (approximately 3%), which led the researchers to conclude that routine preoperative medical testing does not increase the safety of cataract procedures. These results could be applied to other low-risk cases.
Unnecessary testing is also expensive. Researchers at Stanford University Hospital retrospectively compared preoperative test orders during 6-month periods before and 1 year after development of an anesthesia preoperative evaluation clinic.6 They found a 55% reduction in the number of preoperative tests ordered from the period before the clinic was established, when tests were ordered by surgeons and primary care physicians, to the period after the clinic was established, when test ordering was transferred to anesthesiologists at the clinic. This reduction in the number of tests ordered resulted in a 59% reduction in the hospital’s expenditures for preoperative tests, yielding $112 in cost savings per patient. No operating room cancellations, delays, or adverse patient events were reported as a result of the change.
Similar results were reported more recently by researchers at a Canadian hospital, who found that selective preoperative test ordering by staff anesthesiologists reduced the number and cost of preoperative studies compared with usual practice without a resulting increase in complications.7
What are the real legal risks?
Many surgeons express the fear that they will be sued if they do not routinely order preoperative tests. My view is that from a medicolegal standpoint, it is usually better not to order an unnecessary test if the next step to take in the event of an abnormal result would be unclear. The legal risk is greater for not following an abnormal test result than for not ordering a test that was not indicated. One may uncover an abnormal laboratory test finding that is not likely to be clinically significant but that could result in legal action if it were not evaluated further. A complication that may not be related to the abnormal result may develop at some point in the future and be blamed on the lack of follow-up. At our center, we insist that when a physician orders a test, he or she is responsible for the results and for following up on abnormalities.
Should testing be based on age?
Using age as a criterion for preoperative testing is controversial. There is no doubt that the older a patient is, the more likely he or she is to have abnormal test results: patients aged 70 years or older have about a 10% chance of having abnormal levels of serum creatinine, hemoglobin, or glucose8 and a 75% chance of having at least one abnormality on their ECG (and a 50% chance of having a major ECG abnormality).9 However, these factors were found not to be predictive of postoperative complications. In contrast, predictive factors for this age group are an American Society of Anesthesiologists (ASA) physical status classification of at least 3 (indicating severe systemic disease), the risk of the surgical procedure, and a history of congestive heart failure.8,9
Guidelines for testing—and for not testing
About 10 years ago, the ASA attempted to develop a practice guideline for routine preoperative testing. The available data were so inconsistent, however, that the ASA could not reach a consensus and instead issued a practice advisory.10
Even so, there are a number of general principles for avoiding unnecessary preoperative testing:
- Routine laboratory tests are not good screening devices and should not be used to screen for disease
- Repetition should be avoided: there is no need to repeat a recent test
- Healthy patients may not need testing
- Patients undergoing minimally invasive procedures may not need testing
- A test should be ordered only if its results will influence management.
A CLOSER LOOK AT A FEW SPECIFIC TESTS
Question: Which of the following tests is most likely to provide useful information to aid clinical decision-making during a preoperative evaluation for laparoscopic cholecystectomy?
A. A chest radiograph in a 43-year-old woman with asthma
B. An ECG in a 71-year-old man with hypertension
C. A pregnancy test in an 18-year-old woman with amenorrhea
D. A prothrombin time in a 51-year-old man with anemia
E. A urinalysis in a 67-year-old woman with diabetes
The best answer is C (pregnancy test); an ECG in the 71-year-old man would be less useful (see below). The remaining choices—chest radiograph, prothrombin time, and urinalysis—are even less appropriate. A chest radiograph in an asthmatic patient is not likely to yield more information than what is obtained from the history and physical exam. Patients with anemia are not likely to have abnormal coagulation, and the role of urinalysis in detecting glucose and protein in asymptomatic diabetic patients is limited.
Routine pregnancy testing is justifiable
There are a number of reasons to justify a low threshold for preoperative pregnancy testing10:
- Patients, especially adolescents, are often unreliable in suspecting that they might be pregnant (in several studies of routine preoperative pregnancy screening, 0.3% to 2.2% of tests were positive)
- History and physical examination are often insufficient to determine early pregnancy
- Management usually changes if it is discovered that a patient is pregnant.
Using the four criteria from Table 1, pregnancy testing rates high as a useful test: it would identify “abnormality,” it would determine a diagnosis, and it would likely change management.
Routine ECG has limited utility
In contrast, routine preoperative ECG is not well supported. A recent study from the Netherlands assessed the added value of a preoperative ECG in predicting myocardial infarction and death following noncardiac surgery among 2,422 patients older than age 50 years.12 It showed that ECG findings were no more predictive of complications than findings from the history and physical examination and the patient’s activity level.
From our own data at Brigham and Women’s Hospital,13 we found that the presence of any of the following six risk factors predicted all but 0.44% of ECG abnormalities in patients aged 50 years or older presenting for preoperative evaluation:
- Age greater than 65 years
- Congestive heart failure
- High cholesterol
- Angina
- Myocardial infarction
- Severe valvular disease.
The 2007 guidelines on perioperative risk assessment from the American College of Cardiology and American Heart Association (ACC/AHA) do not consider ECG to be indicated in asymptomatic patients undergoing low-risk noncardiac procedures regardless of patient age,14 like the 71-year-old man with hypertension in the above case question. These guidelines also state that evidence for routine ECG orders is not well established in patients with at least one clinical risk factor undergoing intermediate-risk procedures.
The aforementioned ASA practice advisory acknowledges that the likelihood of ECG abnormalities rises with increasing patient age, but the ASA was unable to reach consensus on a minimum age for routinely ordering an ECG in surgical candidates.10 The advisory recommends taking into account other factors, such as cardiac risk factors, the presence of cardiocirculatory or respiratory disease, and the type and invasiveness of the surgical procedure.10
In recommendations not specific to the perioperative setting, the US Preventive Services Task Force advises against routine screening for coronary heart disease with ECG or exercise treadmill testing.15 It gives routine screening a “D” recommendation, indicating that risk is greater than benefit because of the potential for unnecessary invasive procedures, overtreatment, and mislabeling of patients.
Our group at Brigham and Women’s Hospital recently surveyed anesthesiology program directors at US teaching hospitals to determine their preoperative test-ordering practices.16 Among the 75 respondents (58% response rate), 95% said their institutions have no requirements for ordering ECGs unless indicated based on age, history, or surgery type; 71% said their institutions have age-based requirements for ordering ECGs (usually > 50 years). Most respondents reported that their institutions are ordering fewer ECGs since the publication of the 2007 ACC/AHA guidelines on perioperative evaluation.
Whether or not age should be used as a criterion for ECG testing is controversial, and editorials on this subject abound.17–19 They point out that clinicians must be careful before abandoning routine ECGs in elderly patients, for several reasons:
- An abnormal ECG (or abnormal lab test results) may modify a patient’s ASA classification (which is predictive of complications)
- At least one-quarter of myocardial infarctions in elderly persons are “silent” or clinically unrecognized
- A preoperative ECG provides a useful baseline if the patient should develop ECG changes, chest pain, or cardiac complications during the perioperative period.
Most institutions use age as a criterion for ordering tests, especially for ECGs. If such a policy is used, a threshold of 60 years or older is probably most appropriate. However, a patient with good functional capacity who is undergoing a low-risk procedure does not need cardiac testing.14,20
An additional consideration is cost. Although the cost of a single ECG is modest, the cumulative cost of preoperative ECGs for all older surgical patients is significant over the course of a year. Because the Centers for Medicare and Medicaid Services (CMS) no longer cover routine preoperative ECGs, routine testing can be very costly to an institution over time.
COST AND REGULATORY BENEFITS OF PREOPERATIVE CENTERS
Preoperative evaluation centers tend to be cost-effective, as they keep consultations and redundant provider interviews to a minimum, encourage more appropriate targeting of tests, and help to avoid last-minute operating room delays and cancellations.21,22 They also provide an efficient means of compiling the chart for the operating room.
The merits of standardization
Preoperative evaluation centers likewise encourage more standardized preoperative assessment, which can facilitate compliance with surgical quality measures such as those from the National Surgical Quality Improvement Program and the Leapfrog Group. Standardization also fosters more efficient and consistent regulatory documentation, making it easier to follow requirements from CMS (often linked to reimbursement) and the Joint Commission. It also tends to improve reimbursement by encouraging more appropriate coding under CMS’ diagnosis-related group (DRG) system to indicate that whatever testing is ordered is related to the surgical diagnosis or to relevant comorbidities.
No excessive dictates from Joint Commission or CMS
Contrary to what many believe, the Joint Commission does not require excessive preoperative testing. The Joint Commission has no mandate for routine diagnostic tests but requires only what is necessary for determining a patient’s health care needs.23
CMS provides no guidance as to what to do or not do in a preoperative assessment, but it does not reimburse for routine screening tests or for age-based testing.24 Reimbursement for a preoperative ECG, for example, requires documentation of the patient’s signs or symptoms; for an ECG that is indicated, reimbursement includes review and interpretation by the physician.25
A new partner for proper preoperative assessment
Appropriate preoperative evaluation and testing is one of the goals promoted by the recently formed Society for Perioperative Assessment and Quality Improvement (SPAQI). The mission of this international nonprofit organization is to optimize surgical outcomes by sharing best practices and promoting research and communication among health professionals across multiple disciplines. More information is available at www.spaqi.org.
DISCUSSION
Question from the audience: At my hospital, we teach residents about limiting the preoperative tests they order, but surgeons routinely expect many of these tests, including chest x-rays in patients with pulmonary conditions. Are any surgical societies involved in efforts to reduce preoperative testing? Or are surgical societies’ recommendations actually driving some of the unnecessary testing?
Dr. Hepner: I’m not aware of recommendations from surgical societies regarding preoperative testing. Many surgeons believe that the more testing that’s done, the likelier they are to uncover an occult disease. They also often want baseline information, which may actually be warranted in some cases.
Question from the audience: If you’re already ordering a “type and screen” or “type and hold” for a patient, isn’t it worthwhile to just add on a CBC? The patient is already getting the phlebotomy, so isn’t there a cost benefit to getting other routine tests done at the same time rather than calling the patient back for more tests if another indication arises?
Dr. Hepner: Charges are generally assessed for each individual test, not for drawing blood, so I would only get the tests that are indicated.
Question from the audience: In institutions without a preoperative clinic, sometimes the surgeons do the work-up without discussing the case with the primary doctor, and the surgeons want an ECG so that the case isn’t cancelled at the last minute. Can you give straightforward criteria in such cases, such as an age threshold, or would you not order an ECG for anyone?
Dr. Hepner: Based on our most recent data, 60 years seems to be a reasonable cutoff if you are going to use age as a criterion.
Question from the audience: What criteria do you use for preoperative screening with pregnancy tests?
Dr. Hepner: If you have an unreliable patient population, general screening should be done. We don’t have such a requirement, but we have a very low threshold. If a patient appears very reliable, knowing the exact date of her last menstrual period, we’ll go by that. If a patient is unsure, we’ll do a pregnancy test.
Question from the audience: My hospital doesn’t have a preoperative clinic, and until recently, the anesthesiology department has helped surgeons with ordering of preoperative tests. We followed a guideline protocol for about 20 years. Now the newer surgeons say they don’t want to be responsible for abnormal test results. Yet we anesthesiologists aren’t seeing the patients, so we can’t use clinical judgment; we can only go by the guidelines. The surgeons are the only physicians on the case who actually do the history and physical exam. So who should sign the test orders and be responsible for abnormal results?
Dr. Hepner: In our preoperative test center, we tell the surgical team that if they are uncertain about which tests to order, we will handle it. And if we order a test, we follow up on the results. You must ensure that orders are signed and not rubber-stamped; that way, the person who orders a test will get called with any abnormal results. If you order it, you own it.
Question from the audience: I agree that no testing is needed for the 72-year-old man you presented who was undergoing surgery for inguinal hernia, but it always worries me not to do an ECG since part of the standard of care for anesthesia is intraoperative ECG monitoring. If we see some sort of unusual arrhythmia when we take the patient in, we might cancel the case if we don’t know whether it was present at baseline. Surgeons will ask me, “Why didn’t you order a baseline if you’re going to monitor the ECG in the operating room? If you’re not going to order a baseline, then why monitor the ECG?” These are medicolegal issues that I haven’t seen addressed.
Dr. Hepner: A case like you describe will be addressed in the upcoming medicolegal session (see page S119). You make a good point that many times just having a baseline is helpful, but I would argue that it is more helpful for intermediate- or high-risk cases.
- Klafta JM, Roizen MF. Current understanding of patients’ attitudes toward and preparation for anesthesia: a review. Anesth Analg 1996; 83:1314–1321.
- Halaszynski TM, Juda R, Silverman DG. Optimizing postoperative outcomes with efficient preoperative assessment and management. Crit Care Med 2004; 32(suppl 4):S76–S86.
- Roizen MF, Kaplan EB, Schreider BD, Lichtor LJ, Orkin FK. The relative roles of the history and physical examination, and laboratory testing in preoperative evaluation for outpatient surgery: the “Starling” curve of preoperative laboratory testing. Anesthesiol Clin North Am 1987; 5:15–34.
- Roizen MF. More preoperative assessment by physicians and less by laboratory tests. N Engl J Med 2000; 342:204–205.
- Schein OD, Katz J, Bass EB, et al. The value of routine preoperative medical testing before cataract surgery. Study of Medical Testing for Cataract Surgery. N Engl J Med 2000; 342:168–175.
- Fischer SP. Development and effectiveness of an anesthesia preoperative evaluation clinic in a teaching hospital. Anesthesiology 1996; 85:196–206.
- Finegan BA, Rashiq S, McAlister FA, O’Connor P. Selective ordering of preoperative investigations by anesthesiologists reduces the number and cost of tests. Can J Anaesth 2005; 52:575–580.
- Dzankic S, Pastor D, Gonzalez C, Leung JM. The prevalence and predictive value of abnormal preoperative laboratory tests in elderly surgical patients. Anesth Analg 2001; 93:301–308.
- Liu LL, Dzankic S, Leung JM. Preoperative electrocardiogram abnormalities do not predict postoperative cardiac complications in geriatric surgical patients. J Am Geriatr Soc 2002; 50:1186–1191.
- American Society of Anesthesiologists Task Force on Preanesthesia Evaluation. Practice advisory for preanesthesia evaluation: a report by the American Society of Anesthesiologists Task Force on Preanesthesia Evaluation. Anesthesiology 2002; 96:485–496.
- Silverstein MD, Boland BJ. Conceptual framework for evaluating laboratory tests: case-finding in ambulatory patients. Clin Chem 1994; 40:1621–1627.
- van Klei WA, Bryson GL, Yang H, et al. The value of routine preoperative electrocardiography in predicting myocardial infarction after noncardiac surgery. Ann Surg 2007; 246:165–170.
- Correll DJ, Hepner DL, Chang C, Tsen L, Hevelone ND, Bader AM. Preoperative electrocardiograms: patient factors predictive of abnormalities. Anesthesiology 2009; 110:1217–1222.
- Fleisher LA, Beckman JA, Brown KA, et al. ACC/AHA 2007 guidelines on perioperative cardiovascular evaluation and care for noncardiac surgery: executive summary. J Am Coll Cardiol 2007; 50:1707–1732.
- U.S. Preventive Services Task Force. Screening for coronary heart disease: recommendation statement. Agency for Healthcare Research and Quality Web site. http://www.ahrq.gov/clinic/3rduspstf/chd/chdrs.htm. Accessed March 20, 2009.
- Hepner DL, Bader AR, Correll D, Tsen LC, Segal BS, Bader AM. An analysis of preoperative testing protocols in academic anesthesiology programs. Poster presented at: 4th Annual Perioperative Medicine Summit; February 5–7, 2009; Miami Beach, FL. http://www.ccjm.org/content/76/Electronic_Suppl_1/eS18.full.pdf+html. Accessed May 28, 2009.
- De Hert SG. Preoperative electrocardiograms: obsolete or still useful? Anesthesiology 2009; 110:1205–1206.
- Fleisher LA. The preoperative electrocardiogram: what is the role in 2007? Ann Surg 2007; 246:171–172.
- Rich MW. The preoperative electrocardiogram: have we reached the end of an era? J Am Geriatr Soc 2002; 50:1301–1303.
- Noordzij PG, Boersma E, Bax JJ, et al. Prognostic value of routine preoperative electrocardiography in patients undergoing noncardiac surgery. Am J Cardiol 2006; 97:1103–1106.
- Ferschl MB, Tung A, Sweitzer B, Huo D, Glick DB. Preoperative clinic visits reduce operating room cancellations and delays. Anesthesiology 2005; 103:855–859.
- Correll DJ, Bader AM, Hull MW, Tsen LC, Hepner DL. The value of preoperative clinic visits in identifying issues with potential impact on operating room efficiency. Anesthesiology 2006; 105:1254–1259.
- The Joint Commission. 2009 Comprehensive Accreditation Manual for Hospitals: The Official Handbook. Oak Brook, IL: Joint Commission Resources; 2008.
- Medicare coverage center. Centers for Medicare & Medicaid Services Web site. http://www.cms.hhs.gov/Center/coverage.asp. Accessed March 20, 2009.
- CMS Manual System: Pub. 100-03 Medicare National Coverage Determinations. Dept of Health & Human Services (DHHS), Centers for Medicare & Medicaid Services (CMS). Transmittal 26. December 10, 2004.
Routine presurgical assessment of patients with a standard battery of tests not only is wasteful but can lead to more unnecessary expense, delay, and even risk to the patient and physician. Any abnormal tests results, even if likely to be clinically unimportant to the upcoming surgery, will need to be followed up to rule out a significant abnormality that may have later implications. This review will outline strategies for making decisions about which tests are clinically useful for preoperative assessment of a given patient and also discuss the value of preoperative evaluation centers in promoting appropriate preoperative testing.
PREOPERATIVE EVALUATION SHOULD BE CLEARLY DIRECTED
Most patients scheduled for surgery at Brigham and Women’s Hospital are assessed by the staff at our preoperative evaluation center. We take a medical history and conduct a physical examination, review the medical records, order laboratory tests or other studies as indicated, and determine which patients need further work-up or consultations. The goals are to evaluate patient readiness for anesthesia and surgery, optimize patient health before surgery, enhance the quality of perioperative care, reduce the morbidity of surgery and length of stay, and return the patient to normal functioning.1,2
The above goals are generally achieved by directed laboratory testing, managing the patient’s medications, stabilizing disorders when possible, and creating plans for postoperative care and pain management. Communication among the surgeon, the anesthesiology team, and the preoperative medical consultant (if there is one) is critical.1,2
In contrast, “clearing the patient for surgery” is not a legitimate goal of consultation. The real issues to be taken up in a consult are:
- What is the patient’s risk of complications (cardiac and noncardiac)?
- Would further risk stratification alter patient management?
- Can anything be done to reduce the patient’s risk?
If indicated, a consult should cover the entire perioperative period, offering opinions on operative risk and suggesting treatments that affect long-term patient outcomes. Rarely is preoperative intervention necessary just to lower the risk of surgery. Most interventions that are needed should be done regardless of the surgery.
Everyone on the medical team should have the goal of efficient resource utilization, including avoidance of unnecessary visits, laboratory testing, and consultations.
PREOPERATIVE TESTING: WHAT IS NEEDED?
Preoperative testing is extremely expensive: even more than 20 years ago, preoperative medical testing for all types of surgery accounted for approximately $30 billion in US health care costs annually.3 The likelihood of abnormal test results increases with age, and the more tests performed, the more likely a false positive will occur, further driving up costs.
Preoperative testing should generally be directed by a targeted history and physical examination, and the relevance of any tests should be considered in light of the type of procedure that is planned, particularly the hemodynamic changes and blood loss involved. Before ordering a test, physicians should be sure that there is a good reason for the test, that it is consistent with established guidelines, and that the results will be useful (ie, have the potential to change management).
Case study: Inguinal surgery in a healthy elderly man
A 72-year-old man is being evaluated prior to a right inguinal herniorrhaphy. He has osteoarthritis but is otherwise healthy and jogs 3 to 5 miles several times a week. He takes no medications and has no known drug allergies.
Question: Which of the following tests is necessary prior to surgery?
A. Complete blood cell count (CBC)
B. Prothrombin time and partial thromboplastin time
C. Electrocardiogram (ECG)
D. All of the above
E. None of the above
The correct answer is E (none of the above), for the reasons laid out in the following section.
Unnecessary testing may cause more harm than good
Untargeted testing should be avoided. An unexpected result will probably not be clinically significant for the surgery and will only lead to more needless testing, unnecessary anxiety for the patient, and delays in proceeding to the operating room.4 The more tests that are ordered, the higher the likelihood of having an abnormal result by chance: for a test with 95% specificity, results for 1 out of 13 ordered tests will likely be abnormal without there being a true underlying physiologic abnormality.
Researchers at Johns Hopkins University assessed the value of routine preoperative medical testing in a randomized study of nearly 20,000 patients undergoing elective cataract surgery whose preoperative history and physical examination was either preceded or not preceded by a standard battery of tests, including ECG, CBC, electrolytes, urea nitrogen, creatinine, and glucose.5 This was an ideal study population, given the relatively noninvasive nature of the procedure (with minimal hemodynamic changes) and cataract patients’ relatively advanced age and resulting likelihood of comorbidities. Notably, there were no differences between the two groups in the overall rate of complications (approximately 3%), which led the researchers to conclude that routine preoperative medical testing does not increase the safety of cataract procedures. These results could be applied to other low-risk cases.
Unnecessary testing is also expensive. Researchers at Stanford University Hospital retrospectively compared preoperative test orders during 6-month periods before and 1 year after development of an anesthesia preoperative evaluation clinic.6 They found a 55% reduction in the number of preoperative tests ordered from the period before the clinic was established, when tests were ordered by surgeons and primary care physicians, to the period after the clinic was established, when test ordering was transferred to anesthesiologists at the clinic. This reduction in the number of tests ordered resulted in a 59% reduction in the hospital’s expenditures for preoperative tests, yielding $112 in cost savings per patient. No operating room cancellations, delays, or adverse patient events were reported as a result of the change.
Similar results were reported more recently by researchers at a Canadian hospital, who found that selective preoperative test ordering by staff anesthesiologists reduced the number and cost of preoperative studies compared with usual practice without a resulting increase in complications.7
What are the real legal risks?
Many surgeons express the fear that they will be sued if they do not routinely order preoperative tests. My view is that from a medicolegal standpoint, it is usually better not to order an unnecessary test if the next step to take in the event of an abnormal result would be unclear. The legal risk is greater for not following an abnormal test result than for not ordering a test that was not indicated. One may uncover an abnormal laboratory test finding that is not likely to be clinically significant but that could result in legal action if it were not evaluated further. A complication that may not be related to the abnormal result may develop at some point in the future and be blamed on the lack of follow-up. At our center, we insist that when a physician orders a test, he or she is responsible for the results and for following up on abnormalities.
Should testing be based on age?
Using age as a criterion for preoperative testing is controversial. There is no doubt that the older a patient is, the more likely he or she is to have abnormal test results: patients aged 70 years or older have about a 10% chance of having abnormal levels of serum creatinine, hemoglobin, or glucose8 and a 75% chance of having at least one abnormality on their ECG (and a 50% chance of having a major ECG abnormality).9 However, these factors were found not to be predictive of postoperative complications. In contrast, predictive factors for this age group are an American Society of Anesthesiologists (ASA) physical status classification of at least 3 (indicating severe systemic disease), the risk of the surgical procedure, and a history of congestive heart failure.8,9
Guidelines for testing—and for not testing
About 10 years ago, the ASA attempted to develop a practice guideline for routine preoperative testing. The available data were so inconsistent, however, that the ASA could not reach a consensus and instead issued a practice advisory.10
Even so, there are a number of general principles for avoiding unnecessary preoperative testing:
- Routine laboratory tests are not good screening devices and should not be used to screen for disease
- Repetition should be avoided: there is no need to repeat a recent test
- Healthy patients may not need testing
- Patients undergoing minimally invasive procedures may not need testing
- A test should be ordered only if its results will influence management.
A CLOSER LOOK AT A FEW SPECIFIC TESTS
Question: Which of the following tests is most likely to provide useful information to aid clinical decision-making during a preoperative evaluation for laparoscopic cholecystectomy?
A. A chest radiograph in a 43-year-old woman with asthma
B. An ECG in a 71-year-old man with hypertension
C. A pregnancy test in an 18-year-old woman with amenorrhea
D. A prothrombin time in a 51-year-old man with anemia
E. A urinalysis in a 67-year-old woman with diabetes
The best answer is C (pregnancy test); an ECG in the 71-year-old man would be less useful (see below). The remaining choices—chest radiograph, prothrombin time, and urinalysis—are even less appropriate. A chest radiograph in an asthmatic patient is not likely to yield more information than what is obtained from the history and physical exam. Patients with anemia are not likely to have abnormal coagulation, and the role of urinalysis in detecting glucose and protein in asymptomatic diabetic patients is limited.
Routine pregnancy testing is justifiable
There are a number of reasons to justify a low threshold for preoperative pregnancy testing10:
- Patients, especially adolescents, are often unreliable in suspecting that they might be pregnant (in several studies of routine preoperative pregnancy screening, 0.3% to 2.2% of tests were positive)
- History and physical examination are often insufficient to determine early pregnancy
- Management usually changes if it is discovered that a patient is pregnant.
Using the four criteria from Table 1, pregnancy testing rates high as a useful test: it would identify “abnormality,” it would determine a diagnosis, and it would likely change management.
Routine ECG has limited utility
In contrast, routine preoperative ECG is not well supported. A recent study from the Netherlands assessed the added value of a preoperative ECG in predicting myocardial infarction and death following noncardiac surgery among 2,422 patients older than age 50 years.12 It showed that ECG findings were no more predictive of complications than findings from the history and physical examination and the patient’s activity level.
From our own data at Brigham and Women’s Hospital,13 we found that the presence of any of the following six risk factors predicted all but 0.44% of ECG abnormalities in patients aged 50 years or older presenting for preoperative evaluation:
- Age greater than 65 years
- Congestive heart failure
- High cholesterol
- Angina
- Myocardial infarction
- Severe valvular disease.
The 2007 guidelines on perioperative risk assessment from the American College of Cardiology and American Heart Association (ACC/AHA) do not consider ECG to be indicated in asymptomatic patients undergoing low-risk noncardiac procedures regardless of patient age,14 like the 71-year-old man with hypertension in the above case question. These guidelines also state that evidence for routine ECG orders is not well established in patients with at least one clinical risk factor undergoing intermediate-risk procedures.
The aforementioned ASA practice advisory acknowledges that the likelihood of ECG abnormalities rises with increasing patient age, but the ASA was unable to reach consensus on a minimum age for routinely ordering an ECG in surgical candidates.10 The advisory recommends taking into account other factors, such as cardiac risk factors, the presence of cardiocirculatory or respiratory disease, and the type and invasiveness of the surgical procedure.10
In recommendations not specific to the perioperative setting, the US Preventive Services Task Force advises against routine screening for coronary heart disease with ECG or exercise treadmill testing.15 It gives routine screening a “D” recommendation, indicating that risk is greater than benefit because of the potential for unnecessary invasive procedures, overtreatment, and mislabeling of patients.
Our group at Brigham and Women’s Hospital recently surveyed anesthesiology program directors at US teaching hospitals to determine their preoperative test-ordering practices.16 Among the 75 respondents (58% response rate), 95% said their institutions have no requirements for ordering ECGs unless indicated based on age, history, or surgery type; 71% said their institutions have age-based requirements for ordering ECGs (usually > 50 years). Most respondents reported that their institutions are ordering fewer ECGs since the publication of the 2007 ACC/AHA guidelines on perioperative evaluation.
Whether or not age should be used as a criterion for ECG testing is controversial, and editorials on this subject abound.17–19 They point out that clinicians must be careful before abandoning routine ECGs in elderly patients, for several reasons:
- An abnormal ECG (or abnormal lab test results) may modify a patient’s ASA classification (which is predictive of complications)
- At least one-quarter of myocardial infarctions in elderly persons are “silent” or clinically unrecognized
- A preoperative ECG provides a useful baseline if the patient should develop ECG changes, chest pain, or cardiac complications during the perioperative period.
Most institutions use age as a criterion for ordering tests, especially for ECGs. If such a policy is used, a threshold of 60 years or older is probably most appropriate. However, a patient with good functional capacity who is undergoing a low-risk procedure does not need cardiac testing.14,20
An additional consideration is cost. Although the cost of a single ECG is modest, the cumulative cost of preoperative ECGs for all older surgical patients is significant over the course of a year. Because the Centers for Medicare and Medicaid Services (CMS) no longer cover routine preoperative ECGs, routine testing can be very costly to an institution over time.
COST AND REGULATORY BENEFITS OF PREOPERATIVE CENTERS
Preoperative evaluation centers tend to be cost-effective, as they keep consultations and redundant provider interviews to a minimum, encourage more appropriate targeting of tests, and help to avoid last-minute operating room delays and cancellations.21,22 They also provide an efficient means of compiling the chart for the operating room.
The merits of standardization
Preoperative evaluation centers likewise encourage more standardized preoperative assessment, which can facilitate compliance with surgical quality measures such as those from the National Surgical Quality Improvement Program and the Leapfrog Group. Standardization also fosters more efficient and consistent regulatory documentation, making it easier to follow requirements from CMS (often linked to reimbursement) and the Joint Commission. It also tends to improve reimbursement by encouraging more appropriate coding under CMS’ diagnosis-related group (DRG) system to indicate that whatever testing is ordered is related to the surgical diagnosis or to relevant comorbidities.
No excessive dictates from Joint Commission or CMS
Contrary to what many believe, the Joint Commission does not require excessive preoperative testing. The Joint Commission has no mandate for routine diagnostic tests but requires only what is necessary for determining a patient’s health care needs.23
CMS provides no guidance as to what to do or not do in a preoperative assessment, but it does not reimburse for routine screening tests or for age-based testing.24 Reimbursement for a preoperative ECG, for example, requires documentation of the patient’s signs or symptoms; for an ECG that is indicated, reimbursement includes review and interpretation by the physician.25
A new partner for proper preoperative assessment
Appropriate preoperative evaluation and testing is one of the goals promoted by the recently formed Society for Perioperative Assessment and Quality Improvement (SPAQI). The mission of this international nonprofit organization is to optimize surgical outcomes by sharing best practices and promoting research and communication among health professionals across multiple disciplines. More information is available at www.spaqi.org.
DISCUSSION
Question from the audience: At my hospital, we teach residents about limiting the preoperative tests they order, but surgeons routinely expect many of these tests, including chest x-rays in patients with pulmonary conditions. Are any surgical societies involved in efforts to reduce preoperative testing? Or are surgical societies’ recommendations actually driving some of the unnecessary testing?
Dr. Hepner: I’m not aware of recommendations from surgical societies regarding preoperative testing. Many surgeons believe that the more testing that’s done, the likelier they are to uncover an occult disease. They also often want baseline information, which may actually be warranted in some cases.
Question from the audience: If you’re already ordering a “type and screen” or “type and hold” for a patient, isn’t it worthwhile to just add on a CBC? The patient is already getting the phlebotomy, so isn’t there a cost benefit to getting other routine tests done at the same time rather than calling the patient back for more tests if another indication arises?
Dr. Hepner: Charges are generally assessed for each individual test, not for drawing blood, so I would only get the tests that are indicated.
Question from the audience: In institutions without a preoperative clinic, sometimes the surgeons do the work-up without discussing the case with the primary doctor, and the surgeons want an ECG so that the case isn’t cancelled at the last minute. Can you give straightforward criteria in such cases, such as an age threshold, or would you not order an ECG for anyone?
Dr. Hepner: Based on our most recent data, 60 years seems to be a reasonable cutoff if you are going to use age as a criterion.
Question from the audience: What criteria do you use for preoperative screening with pregnancy tests?
Dr. Hepner: If you have an unreliable patient population, general screening should be done. We don’t have such a requirement, but we have a very low threshold. If a patient appears very reliable, knowing the exact date of her last menstrual period, we’ll go by that. If a patient is unsure, we’ll do a pregnancy test.
Question from the audience: My hospital doesn’t have a preoperative clinic, and until recently, the anesthesiology department has helped surgeons with ordering of preoperative tests. We followed a guideline protocol for about 20 years. Now the newer surgeons say they don’t want to be responsible for abnormal test results. Yet we anesthesiologists aren’t seeing the patients, so we can’t use clinical judgment; we can only go by the guidelines. The surgeons are the only physicians on the case who actually do the history and physical exam. So who should sign the test orders and be responsible for abnormal results?
Dr. Hepner: In our preoperative test center, we tell the surgical team that if they are uncertain about which tests to order, we will handle it. And if we order a test, we follow up on the results. You must ensure that orders are signed and not rubber-stamped; that way, the person who orders a test will get called with any abnormal results. If you order it, you own it.
Question from the audience: I agree that no testing is needed for the 72-year-old man you presented who was undergoing surgery for inguinal hernia, but it always worries me not to do an ECG since part of the standard of care for anesthesia is intraoperative ECG monitoring. If we see some sort of unusual arrhythmia when we take the patient in, we might cancel the case if we don’t know whether it was present at baseline. Surgeons will ask me, “Why didn’t you order a baseline if you’re going to monitor the ECG in the operating room? If you’re not going to order a baseline, then why monitor the ECG?” These are medicolegal issues that I haven’t seen addressed.
Dr. Hepner: A case like you describe will be addressed in the upcoming medicolegal session (see page S119). You make a good point that many times just having a baseline is helpful, but I would argue that it is more helpful for intermediate- or high-risk cases.
Routine presurgical assessment of patients with a standard battery of tests not only is wasteful but can lead to more unnecessary expense, delay, and even risk to the patient and physician. Any abnormal tests results, even if likely to be clinically unimportant to the upcoming surgery, will need to be followed up to rule out a significant abnormality that may have later implications. This review will outline strategies for making decisions about which tests are clinically useful for preoperative assessment of a given patient and also discuss the value of preoperative evaluation centers in promoting appropriate preoperative testing.
PREOPERATIVE EVALUATION SHOULD BE CLEARLY DIRECTED
Most patients scheduled for surgery at Brigham and Women’s Hospital are assessed by the staff at our preoperative evaluation center. We take a medical history and conduct a physical examination, review the medical records, order laboratory tests or other studies as indicated, and determine which patients need further work-up or consultations. The goals are to evaluate patient readiness for anesthesia and surgery, optimize patient health before surgery, enhance the quality of perioperative care, reduce the morbidity of surgery and length of stay, and return the patient to normal functioning.1,2
The above goals are generally achieved by directed laboratory testing, managing the patient’s medications, stabilizing disorders when possible, and creating plans for postoperative care and pain management. Communication among the surgeon, the anesthesiology team, and the preoperative medical consultant (if there is one) is critical.1,2
In contrast, “clearing the patient for surgery” is not a legitimate goal of consultation. The real issues to be taken up in a consult are:
- What is the patient’s risk of complications (cardiac and noncardiac)?
- Would further risk stratification alter patient management?
- Can anything be done to reduce the patient’s risk?
If indicated, a consult should cover the entire perioperative period, offering opinions on operative risk and suggesting treatments that affect long-term patient outcomes. Rarely is preoperative intervention necessary just to lower the risk of surgery. Most interventions that are needed should be done regardless of the surgery.
Everyone on the medical team should have the goal of efficient resource utilization, including avoidance of unnecessary visits, laboratory testing, and consultations.
PREOPERATIVE TESTING: WHAT IS NEEDED?
Preoperative testing is extremely expensive: even more than 20 years ago, preoperative medical testing for all types of surgery accounted for approximately $30 billion in US health care costs annually.3 The likelihood of abnormal test results increases with age, and the more tests performed, the more likely a false positive will occur, further driving up costs.
Preoperative testing should generally be directed by a targeted history and physical examination, and the relevance of any tests should be considered in light of the type of procedure that is planned, particularly the hemodynamic changes and blood loss involved. Before ordering a test, physicians should be sure that there is a good reason for the test, that it is consistent with established guidelines, and that the results will be useful (ie, have the potential to change management).
Case study: Inguinal surgery in a healthy elderly man
A 72-year-old man is being evaluated prior to a right inguinal herniorrhaphy. He has osteoarthritis but is otherwise healthy and jogs 3 to 5 miles several times a week. He takes no medications and has no known drug allergies.
Question: Which of the following tests is necessary prior to surgery?
A. Complete blood cell count (CBC)
B. Prothrombin time and partial thromboplastin time
C. Electrocardiogram (ECG)
D. All of the above
E. None of the above
The correct answer is E (none of the above), for the reasons laid out in the following section.
Unnecessary testing may cause more harm than good
Untargeted testing should be avoided. An unexpected result will probably not be clinically significant for the surgery and will only lead to more needless testing, unnecessary anxiety for the patient, and delays in proceeding to the operating room.4 The more tests that are ordered, the higher the likelihood of having an abnormal result by chance: for a test with 95% specificity, results for 1 out of 13 ordered tests will likely be abnormal without there being a true underlying physiologic abnormality.
Researchers at Johns Hopkins University assessed the value of routine preoperative medical testing in a randomized study of nearly 20,000 patients undergoing elective cataract surgery whose preoperative history and physical examination was either preceded or not preceded by a standard battery of tests, including ECG, CBC, electrolytes, urea nitrogen, creatinine, and glucose.5 This was an ideal study population, given the relatively noninvasive nature of the procedure (with minimal hemodynamic changes) and cataract patients’ relatively advanced age and resulting likelihood of comorbidities. Notably, there were no differences between the two groups in the overall rate of complications (approximately 3%), which led the researchers to conclude that routine preoperative medical testing does not increase the safety of cataract procedures. These results could be applied to other low-risk cases.
Unnecessary testing is also expensive. Researchers at Stanford University Hospital retrospectively compared preoperative test orders during 6-month periods before and 1 year after development of an anesthesia preoperative evaluation clinic.6 They found a 55% reduction in the number of preoperative tests ordered from the period before the clinic was established, when tests were ordered by surgeons and primary care physicians, to the period after the clinic was established, when test ordering was transferred to anesthesiologists at the clinic. This reduction in the number of tests ordered resulted in a 59% reduction in the hospital’s expenditures for preoperative tests, yielding $112 in cost savings per patient. No operating room cancellations, delays, or adverse patient events were reported as a result of the change.
Similar results were reported more recently by researchers at a Canadian hospital, who found that selective preoperative test ordering by staff anesthesiologists reduced the number and cost of preoperative studies compared with usual practice without a resulting increase in complications.7
What are the real legal risks?
Many surgeons express the fear that they will be sued if they do not routinely order preoperative tests. My view is that from a medicolegal standpoint, it is usually better not to order an unnecessary test if the next step to take in the event of an abnormal result would be unclear. The legal risk is greater for not following an abnormal test result than for not ordering a test that was not indicated. One may uncover an abnormal laboratory test finding that is not likely to be clinically significant but that could result in legal action if it were not evaluated further. A complication that may not be related to the abnormal result may develop at some point in the future and be blamed on the lack of follow-up. At our center, we insist that when a physician orders a test, he or she is responsible for the results and for following up on abnormalities.
Should testing be based on age?
Using age as a criterion for preoperative testing is controversial. There is no doubt that the older a patient is, the more likely he or she is to have abnormal test results: patients aged 70 years or older have about a 10% chance of having abnormal levels of serum creatinine, hemoglobin, or glucose8 and a 75% chance of having at least one abnormality on their ECG (and a 50% chance of having a major ECG abnormality).9 However, these factors were found not to be predictive of postoperative complications. In contrast, predictive factors for this age group are an American Society of Anesthesiologists (ASA) physical status classification of at least 3 (indicating severe systemic disease), the risk of the surgical procedure, and a history of congestive heart failure.8,9
Guidelines for testing—and for not testing
About 10 years ago, the ASA attempted to develop a practice guideline for routine preoperative testing. The available data were so inconsistent, however, that the ASA could not reach a consensus and instead issued a practice advisory.10
Even so, there are a number of general principles for avoiding unnecessary preoperative testing:
- Routine laboratory tests are not good screening devices and should not be used to screen for disease
- Repetition should be avoided: there is no need to repeat a recent test
- Healthy patients may not need testing
- Patients undergoing minimally invasive procedures may not need testing
- A test should be ordered only if its results will influence management.
A CLOSER LOOK AT A FEW SPECIFIC TESTS
Question: Which of the following tests is most likely to provide useful information to aid clinical decision-making during a preoperative evaluation for laparoscopic cholecystectomy?
A. A chest radiograph in a 43-year-old woman with asthma
B. An ECG in a 71-year-old man with hypertension
C. A pregnancy test in an 18-year-old woman with amenorrhea
D. A prothrombin time in a 51-year-old man with anemia
E. A urinalysis in a 67-year-old woman with diabetes
The best answer is C (pregnancy test); an ECG in the 71-year-old man would be less useful (see below). The remaining choices—chest radiograph, prothrombin time, and urinalysis—are even less appropriate. A chest radiograph in an asthmatic patient is not likely to yield more information than what is obtained from the history and physical exam. Patients with anemia are not likely to have abnormal coagulation, and the role of urinalysis in detecting glucose and protein in asymptomatic diabetic patients is limited.
Routine pregnancy testing is justifiable
There are a number of reasons to justify a low threshold for preoperative pregnancy testing10:
- Patients, especially adolescents, are often unreliable in suspecting that they might be pregnant (in several studies of routine preoperative pregnancy screening, 0.3% to 2.2% of tests were positive)
- History and physical examination are often insufficient to determine early pregnancy
- Management usually changes if it is discovered that a patient is pregnant.
Using the four criteria from Table 1, pregnancy testing rates high as a useful test: it would identify “abnormality,” it would determine a diagnosis, and it would likely change management.
Routine ECG has limited utility
In contrast, routine preoperative ECG is not well supported. A recent study from the Netherlands assessed the added value of a preoperative ECG in predicting myocardial infarction and death following noncardiac surgery among 2,422 patients older than age 50 years.12 It showed that ECG findings were no more predictive of complications than findings from the history and physical examination and the patient’s activity level.
From our own data at Brigham and Women’s Hospital,13 we found that the presence of any of the following six risk factors predicted all but 0.44% of ECG abnormalities in patients aged 50 years or older presenting for preoperative evaluation:
- Age greater than 65 years
- Congestive heart failure
- High cholesterol
- Angina
- Myocardial infarction
- Severe valvular disease.
The 2007 guidelines on perioperative risk assessment from the American College of Cardiology and American Heart Association (ACC/AHA) do not consider ECG to be indicated in asymptomatic patients undergoing low-risk noncardiac procedures regardless of patient age,14 like the 71-year-old man with hypertension in the above case question. These guidelines also state that evidence for routine ECG orders is not well established in patients with at least one clinical risk factor undergoing intermediate-risk procedures.
The aforementioned ASA practice advisory acknowledges that the likelihood of ECG abnormalities rises with increasing patient age, but the ASA was unable to reach consensus on a minimum age for routinely ordering an ECG in surgical candidates.10 The advisory recommends taking into account other factors, such as cardiac risk factors, the presence of cardiocirculatory or respiratory disease, and the type and invasiveness of the surgical procedure.10
In recommendations not specific to the perioperative setting, the US Preventive Services Task Force advises against routine screening for coronary heart disease with ECG or exercise treadmill testing.15 It gives routine screening a “D” recommendation, indicating that risk is greater than benefit because of the potential for unnecessary invasive procedures, overtreatment, and mislabeling of patients.
Our group at Brigham and Women’s Hospital recently surveyed anesthesiology program directors at US teaching hospitals to determine their preoperative test-ordering practices.16 Among the 75 respondents (58% response rate), 95% said their institutions have no requirements for ordering ECGs unless indicated based on age, history, or surgery type; 71% said their institutions have age-based requirements for ordering ECGs (usually > 50 years). Most respondents reported that their institutions are ordering fewer ECGs since the publication of the 2007 ACC/AHA guidelines on perioperative evaluation.
Whether or not age should be used as a criterion for ECG testing is controversial, and editorials on this subject abound.17–19 They point out that clinicians must be careful before abandoning routine ECGs in elderly patients, for several reasons:
- An abnormal ECG (or abnormal lab test results) may modify a patient’s ASA classification (which is predictive of complications)
- At least one-quarter of myocardial infarctions in elderly persons are “silent” or clinically unrecognized
- A preoperative ECG provides a useful baseline if the patient should develop ECG changes, chest pain, or cardiac complications during the perioperative period.
Most institutions use age as a criterion for ordering tests, especially for ECGs. If such a policy is used, a threshold of 60 years or older is probably most appropriate. However, a patient with good functional capacity who is undergoing a low-risk procedure does not need cardiac testing.14,20
An additional consideration is cost. Although the cost of a single ECG is modest, the cumulative cost of preoperative ECGs for all older surgical patients is significant over the course of a year. Because the Centers for Medicare and Medicaid Services (CMS) no longer cover routine preoperative ECGs, routine testing can be very costly to an institution over time.
COST AND REGULATORY BENEFITS OF PREOPERATIVE CENTERS
Preoperative evaluation centers tend to be cost-effective, as they keep consultations and redundant provider interviews to a minimum, encourage more appropriate targeting of tests, and help to avoid last-minute operating room delays and cancellations.21,22 They also provide an efficient means of compiling the chart for the operating room.
The merits of standardization
Preoperative evaluation centers likewise encourage more standardized preoperative assessment, which can facilitate compliance with surgical quality measures such as those from the National Surgical Quality Improvement Program and the Leapfrog Group. Standardization also fosters more efficient and consistent regulatory documentation, making it easier to follow requirements from CMS (often linked to reimbursement) and the Joint Commission. It also tends to improve reimbursement by encouraging more appropriate coding under CMS’ diagnosis-related group (DRG) system to indicate that whatever testing is ordered is related to the surgical diagnosis or to relevant comorbidities.
No excessive dictates from Joint Commission or CMS
Contrary to what many believe, the Joint Commission does not require excessive preoperative testing. The Joint Commission has no mandate for routine diagnostic tests but requires only what is necessary for determining a patient’s health care needs.23
CMS provides no guidance as to what to do or not do in a preoperative assessment, but it does not reimburse for routine screening tests or for age-based testing.24 Reimbursement for a preoperative ECG, for example, requires documentation of the patient’s signs or symptoms; for an ECG that is indicated, reimbursement includes review and interpretation by the physician.25
A new partner for proper preoperative assessment
Appropriate preoperative evaluation and testing is one of the goals promoted by the recently formed Society for Perioperative Assessment and Quality Improvement (SPAQI). The mission of this international nonprofit organization is to optimize surgical outcomes by sharing best practices and promoting research and communication among health professionals across multiple disciplines. More information is available at www.spaqi.org.
DISCUSSION
Question from the audience: At my hospital, we teach residents about limiting the preoperative tests they order, but surgeons routinely expect many of these tests, including chest x-rays in patients with pulmonary conditions. Are any surgical societies involved in efforts to reduce preoperative testing? Or are surgical societies’ recommendations actually driving some of the unnecessary testing?
Dr. Hepner: I’m not aware of recommendations from surgical societies regarding preoperative testing. Many surgeons believe that the more testing that’s done, the likelier they are to uncover an occult disease. They also often want baseline information, which may actually be warranted in some cases.
Question from the audience: If you’re already ordering a “type and screen” or “type and hold” for a patient, isn’t it worthwhile to just add on a CBC? The patient is already getting the phlebotomy, so isn’t there a cost benefit to getting other routine tests done at the same time rather than calling the patient back for more tests if another indication arises?
Dr. Hepner: Charges are generally assessed for each individual test, not for drawing blood, so I would only get the tests that are indicated.
Question from the audience: In institutions without a preoperative clinic, sometimes the surgeons do the work-up without discussing the case with the primary doctor, and the surgeons want an ECG so that the case isn’t cancelled at the last minute. Can you give straightforward criteria in such cases, such as an age threshold, or would you not order an ECG for anyone?
Dr. Hepner: Based on our most recent data, 60 years seems to be a reasonable cutoff if you are going to use age as a criterion.
Question from the audience: What criteria do you use for preoperative screening with pregnancy tests?
Dr. Hepner: If you have an unreliable patient population, general screening should be done. We don’t have such a requirement, but we have a very low threshold. If a patient appears very reliable, knowing the exact date of her last menstrual period, we’ll go by that. If a patient is unsure, we’ll do a pregnancy test.
Question from the audience: My hospital doesn’t have a preoperative clinic, and until recently, the anesthesiology department has helped surgeons with ordering of preoperative tests. We followed a guideline protocol for about 20 years. Now the newer surgeons say they don’t want to be responsible for abnormal test results. Yet we anesthesiologists aren’t seeing the patients, so we can’t use clinical judgment; we can only go by the guidelines. The surgeons are the only physicians on the case who actually do the history and physical exam. So who should sign the test orders and be responsible for abnormal results?
Dr. Hepner: In our preoperative test center, we tell the surgical team that if they are uncertain about which tests to order, we will handle it. And if we order a test, we follow up on the results. You must ensure that orders are signed and not rubber-stamped; that way, the person who orders a test will get called with any abnormal results. If you order it, you own it.
Question from the audience: I agree that no testing is needed for the 72-year-old man you presented who was undergoing surgery for inguinal hernia, but it always worries me not to do an ECG since part of the standard of care for anesthesia is intraoperative ECG monitoring. If we see some sort of unusual arrhythmia when we take the patient in, we might cancel the case if we don’t know whether it was present at baseline. Surgeons will ask me, “Why didn’t you order a baseline if you’re going to monitor the ECG in the operating room? If you’re not going to order a baseline, then why monitor the ECG?” These are medicolegal issues that I haven’t seen addressed.
Dr. Hepner: A case like you describe will be addressed in the upcoming medicolegal session (see page S119). You make a good point that many times just having a baseline is helpful, but I would argue that it is more helpful for intermediate- or high-risk cases.
- Klafta JM, Roizen MF. Current understanding of patients’ attitudes toward and preparation for anesthesia: a review. Anesth Analg 1996; 83:1314–1321.
- Halaszynski TM, Juda R, Silverman DG. Optimizing postoperative outcomes with efficient preoperative assessment and management. Crit Care Med 2004; 32(suppl 4):S76–S86.
- Roizen MF, Kaplan EB, Schreider BD, Lichtor LJ, Orkin FK. The relative roles of the history and physical examination, and laboratory testing in preoperative evaluation for outpatient surgery: the “Starling” curve of preoperative laboratory testing. Anesthesiol Clin North Am 1987; 5:15–34.
- Roizen MF. More preoperative assessment by physicians and less by laboratory tests. N Engl J Med 2000; 342:204–205.
- Schein OD, Katz J, Bass EB, et al. The value of routine preoperative medical testing before cataract surgery. Study of Medical Testing for Cataract Surgery. N Engl J Med 2000; 342:168–175.
- Fischer SP. Development and effectiveness of an anesthesia preoperative evaluation clinic in a teaching hospital. Anesthesiology 1996; 85:196–206.
- Finegan BA, Rashiq S, McAlister FA, O’Connor P. Selective ordering of preoperative investigations by anesthesiologists reduces the number and cost of tests. Can J Anaesth 2005; 52:575–580.
- Dzankic S, Pastor D, Gonzalez C, Leung JM. The prevalence and predictive value of abnormal preoperative laboratory tests in elderly surgical patients. Anesth Analg 2001; 93:301–308.
- Liu LL, Dzankic S, Leung JM. Preoperative electrocardiogram abnormalities do not predict postoperative cardiac complications in geriatric surgical patients. J Am Geriatr Soc 2002; 50:1186–1191.
- American Society of Anesthesiologists Task Force on Preanesthesia Evaluation. Practice advisory for preanesthesia evaluation: a report by the American Society of Anesthesiologists Task Force on Preanesthesia Evaluation. Anesthesiology 2002; 96:485–496.
- Silverstein MD, Boland BJ. Conceptual framework for evaluating laboratory tests: case-finding in ambulatory patients. Clin Chem 1994; 40:1621–1627.
- van Klei WA, Bryson GL, Yang H, et al. The value of routine preoperative electrocardiography in predicting myocardial infarction after noncardiac surgery. Ann Surg 2007; 246:165–170.
- Correll DJ, Hepner DL, Chang C, Tsen L, Hevelone ND, Bader AM. Preoperative electrocardiograms: patient factors predictive of abnormalities. Anesthesiology 2009; 110:1217–1222.
- Fleisher LA, Beckman JA, Brown KA, et al. ACC/AHA 2007 guidelines on perioperative cardiovascular evaluation and care for noncardiac surgery: executive summary. J Am Coll Cardiol 2007; 50:1707–1732.
- U.S. Preventive Services Task Force. Screening for coronary heart disease: recommendation statement. Agency for Healthcare Research and Quality Web site. http://www.ahrq.gov/clinic/3rduspstf/chd/chdrs.htm. Accessed March 20, 2009.
- Hepner DL, Bader AR, Correll D, Tsen LC, Segal BS, Bader AM. An analysis of preoperative testing protocols in academic anesthesiology programs. Poster presented at: 4th Annual Perioperative Medicine Summit; February 5–7, 2009; Miami Beach, FL. http://www.ccjm.org/content/76/Electronic_Suppl_1/eS18.full.pdf+html. Accessed May 28, 2009.
- De Hert SG. Preoperative electrocardiograms: obsolete or still useful? Anesthesiology 2009; 110:1205–1206.
- Fleisher LA. The preoperative electrocardiogram: what is the role in 2007? Ann Surg 2007; 246:171–172.
- Rich MW. The preoperative electrocardiogram: have we reached the end of an era? J Am Geriatr Soc 2002; 50:1301–1303.
- Noordzij PG, Boersma E, Bax JJ, et al. Prognostic value of routine preoperative electrocardiography in patients undergoing noncardiac surgery. Am J Cardiol 2006; 97:1103–1106.
- Ferschl MB, Tung A, Sweitzer B, Huo D, Glick DB. Preoperative clinic visits reduce operating room cancellations and delays. Anesthesiology 2005; 103:855–859.
- Correll DJ, Bader AM, Hull MW, Tsen LC, Hepner DL. The value of preoperative clinic visits in identifying issues with potential impact on operating room efficiency. Anesthesiology 2006; 105:1254–1259.
- The Joint Commission. 2009 Comprehensive Accreditation Manual for Hospitals: The Official Handbook. Oak Brook, IL: Joint Commission Resources; 2008.
- Medicare coverage center. Centers for Medicare & Medicaid Services Web site. http://www.cms.hhs.gov/Center/coverage.asp. Accessed March 20, 2009.
- CMS Manual System: Pub. 100-03 Medicare National Coverage Determinations. Dept of Health & Human Services (DHHS), Centers for Medicare & Medicaid Services (CMS). Transmittal 26. December 10, 2004.
- Klafta JM, Roizen MF. Current understanding of patients’ attitudes toward and preparation for anesthesia: a review. Anesth Analg 1996; 83:1314–1321.
- Halaszynski TM, Juda R, Silverman DG. Optimizing postoperative outcomes with efficient preoperative assessment and management. Crit Care Med 2004; 32(suppl 4):S76–S86.
- Roizen MF, Kaplan EB, Schreider BD, Lichtor LJ, Orkin FK. The relative roles of the history and physical examination, and laboratory testing in preoperative evaluation for outpatient surgery: the “Starling” curve of preoperative laboratory testing. Anesthesiol Clin North Am 1987; 5:15–34.
- Roizen MF. More preoperative assessment by physicians and less by laboratory tests. N Engl J Med 2000; 342:204–205.
- Schein OD, Katz J, Bass EB, et al. The value of routine preoperative medical testing before cataract surgery. Study of Medical Testing for Cataract Surgery. N Engl J Med 2000; 342:168–175.
- Fischer SP. Development and effectiveness of an anesthesia preoperative evaluation clinic in a teaching hospital. Anesthesiology 1996; 85:196–206.
- Finegan BA, Rashiq S, McAlister FA, O’Connor P. Selective ordering of preoperative investigations by anesthesiologists reduces the number and cost of tests. Can J Anaesth 2005; 52:575–580.
- Dzankic S, Pastor D, Gonzalez C, Leung JM. The prevalence and predictive value of abnormal preoperative laboratory tests in elderly surgical patients. Anesth Analg 2001; 93:301–308.
- Liu LL, Dzankic S, Leung JM. Preoperative electrocardiogram abnormalities do not predict postoperative cardiac complications in geriatric surgical patients. J Am Geriatr Soc 2002; 50:1186–1191.
- American Society of Anesthesiologists Task Force on Preanesthesia Evaluation. Practice advisory for preanesthesia evaluation: a report by the American Society of Anesthesiologists Task Force on Preanesthesia Evaluation. Anesthesiology 2002; 96:485–496.
- Silverstein MD, Boland BJ. Conceptual framework for evaluating laboratory tests: case-finding in ambulatory patients. Clin Chem 1994; 40:1621–1627.
- van Klei WA, Bryson GL, Yang H, et al. The value of routine preoperative electrocardiography in predicting myocardial infarction after noncardiac surgery. Ann Surg 2007; 246:165–170.
- Correll DJ, Hepner DL, Chang C, Tsen L, Hevelone ND, Bader AM. Preoperative electrocardiograms: patient factors predictive of abnormalities. Anesthesiology 2009; 110:1217–1222.
- Fleisher LA, Beckman JA, Brown KA, et al. ACC/AHA 2007 guidelines on perioperative cardiovascular evaluation and care for noncardiac surgery: executive summary. J Am Coll Cardiol 2007; 50:1707–1732.
- U.S. Preventive Services Task Force. Screening for coronary heart disease: recommendation statement. Agency for Healthcare Research and Quality Web site. http://www.ahrq.gov/clinic/3rduspstf/chd/chdrs.htm. Accessed March 20, 2009.
- Hepner DL, Bader AR, Correll D, Tsen LC, Segal BS, Bader AM. An analysis of preoperative testing protocols in academic anesthesiology programs. Poster presented at: 4th Annual Perioperative Medicine Summit; February 5–7, 2009; Miami Beach, FL. http://www.ccjm.org/content/76/Electronic_Suppl_1/eS18.full.pdf+html. Accessed May 28, 2009.
- De Hert SG. Preoperative electrocardiograms: obsolete or still useful? Anesthesiology 2009; 110:1205–1206.
- Fleisher LA. The preoperative electrocardiogram: what is the role in 2007? Ann Surg 2007; 246:171–172.
- Rich MW. The preoperative electrocardiogram: have we reached the end of an era? J Am Geriatr Soc 2002; 50:1301–1303.
- Noordzij PG, Boersma E, Bax JJ, et al. Prognostic value of routine preoperative electrocardiography in patients undergoing noncardiac surgery. Am J Cardiol 2006; 97:1103–1106.
- Ferschl MB, Tung A, Sweitzer B, Huo D, Glick DB. Preoperative clinic visits reduce operating room cancellations and delays. Anesthesiology 2005; 103:855–859.
- Correll DJ, Bader AM, Hull MW, Tsen LC, Hepner DL. The value of preoperative clinic visits in identifying issues with potential impact on operating room efficiency. Anesthesiology 2006; 105:1254–1259.
- The Joint Commission. 2009 Comprehensive Accreditation Manual for Hospitals: The Official Handbook. Oak Brook, IL: Joint Commission Resources; 2008.
- Medicare coverage center. Centers for Medicare & Medicaid Services Web site. http://www.cms.hhs.gov/Center/coverage.asp. Accessed March 20, 2009.
- CMS Manual System: Pub. 100-03 Medicare National Coverage Determinations. Dept of Health & Human Services (DHHS), Centers for Medicare & Medicaid Services (CMS). Transmittal 26. December 10, 2004.
KEY POINTS
- Age-based criteria for preoperative testing are controversial because test abnormalities are common in older people but are not as predictive of complications as information gained from the history and physical exam.
- Pregnancy testing is an example of an appropriate preoperative test because pregnancy is often not detectable by the history and physical exam and a positive result would affect case management.
- Routine ordering of preoperative electrocardiograms is not recommended because they are unlikely to offer predictive value beyond the history and physical exam and are costly to an institution over time.
- Routine and aged-based preoperative tests are no longer reimbursed by the Centers for Medicare and Medicaid Services.
Perioperative fluid management: Progress despite lingering controversies
Perioperative fluid management remains controversial. Until recently, fluid management was guided by targets such as urine output, static pressures, blood pressure, and other physiologic variables. Such physiologic signs, however, are inadequate for detecting subclinical hypovolemia. This has prompted the emergence of an approach to fluid administration based on stroke volume and cardiac output—a “flow-guided” approach—designed to overcome the inadequacies of conventional physiologic signs and improve outcomes. Recent technological advances are permitting noninvasive guidance of intravenous fluid therapy to optimize intravascular volume status.
This article reviews the rationale for perioperative fluid management, strategies for perioperative fluid therapy and their associated outcomes, the types of volume expanders used, and considerations for improving perioperative fluid administration.
WHY FLUID MANAGEMENT MATTERS
Postoperative complications predict survival
In 2005, Khuri et al published a study of survival after major surgery that starkly illustrated the prognostic importance of postoperative complications.1 In an effort to identify predictors of long-term survival, they analyzed a National Surgical Quality Improvement Program database of 105,951 patients who underwent eight common operations at Veterans Administration facilities. They found that the most important determinant of reduced postoperative survival over 8 years of follow-up was the occurrence of a complication within 30 days after surgery. The presence of a postoperative complication was a stronger predictor of death than any intraoperative or preoperative risk factor.
Fluid management is key to preventing complications
Optimizing perioperative fluid management is essential to reducing the risk of postoperative complications and mortality. Surgical patients are more likely to have serious complications and die if they have limited physiologic reserve. Adequate fluid administration may reduce the stress response to surgical trauma and support recovery.
Building on early work showing that survivors of major surgery have consistently higher postoperative cardiac output and oxygen delivery (DO2) than do nonsurvivors,2,3 a seminal study by Shoemaker et al showed that these types of blood flow–related parameters are predictive of both survival and complication-free survival.4 Specifically, Shoemaker and his team showed that a protocol designed to achieve DO2 of at least 600 mL/min/m2 was associated with reductions in both postoperative complications and death.4
PROBLEMS WITH PERIOPERATIVE FLUID THERAPY―AND EFFORTS TO OVERCOME THEM
Despite the utility of fluid management in reducing postoperative complications, perioperative fluid therapy is fraught with several fundamental problems:
- Blood volume cannot be evaluated accurately.
- Fluid overload cannot be identified accurately, apart from tissue edema as a result of gross fluid overload.
- Hypovolemia cannot be identified accurately. Commonly measured variables (heart rate, blood pressure, base excess, lactate) are late markers, and the patient’s status upon admission to the operating room is often unknown.
- Tissue perfusion cannot be evaluated accurately. Although lactate and venous oxygen saturation are surrogate markers, genuinely accurate markers for tissue perfusion are lacking.
For these reasons, fluids are commonly administered without the guidance of direct markers of fluid status.
Assessing flow-guided fluid therapy
These shortcomings prompted me and several other researchers to assess the evidence regarding a flow-guided approach to fluid administration, which aims to achieve maximal cardiac output and stroke volume while avoiding excess fluid administration. We conducted a systematic literature search for randomized controlled trials evaluating the postsurgical effects of perioperative fluid therapy to increase global blood flow to explicitly defined goals, after which we performed a meta-analysis of the 22 qualifying studies.5 The trials collectively included 4,546 patients undergoing relatively high-risk elective or emergency surgery, consisting of general, vascular, cardiac, orthopedic, and urologic procedures. Overall mortality in these trials was 10.6% (481 deaths). The primary outcome assessed was mortality; secondary outcomes included morbidity and length of stay in the hospital and in the intensive care unit. Outcomes were assessed according to the timing of the intervention, the fluid type, and explicit measured goals. Fluids were given to all patients, usually as a dynamic bolus, using a flow-guided approach above and beyond that of the control group.
Our analysis found that a flow-guided protocol was associated with a significant reduction in mortality compared with control protocols (odds ratio = 0.82 [95% CI, 0.67–0.99]; P = .04).5 However, sensitivity analysis showed that the largest and best-designed studies tended to yield no significant differences in mortality between the groups, which highlights the remaining need for larger studies to more definitively clarify the effect on mortality.
Timing of administration (ie, whether fluid was given pre-, intra-, or postoperatively) influenced the primary outcome: compared with control, flow-guided fluid therapy was associated with a significant reduction in mortality only when administered intraoperatively, but not when given preoperatively or postoperatively.5
Length of hospital stay was reduced by approximately 1.6 days with flow-guided therapy compared with control (P < .00001), but there was no significant difference between approaches in terms of intensive care unit stay.5
Postoperative complication rates are difficult to compare, given the lack of a uniform definition of a complication and the relative importance of different complications. Nevertheless, when grouped as a whole, the rate of complications was 48% lower (P < .00001) with flow-guided therapy compared with control. Of all outcomes assessed, the effect on complications was the most consistent among all the studies in the analysis. To provide an example using one easily defined complication, the incidence of renal failure was reduced by 35% with flow-guided therapy compared with control (P = .002).5
COLLOID OR CRYSTALLOID?
Two pharmacologically distinct classes
Intravenous fluids can be broadly classified into colloid and crystalloid solutions, and the relative merits of these two fluid classes are at the center of an enduring debate that predates the advent of flow-based fluid administration. Despite fundamental differences in their pharmacokinetics and other characteristics, colloids and crystalloids are often not sufficiently distinguished from one another in discussions of perioperative fluid therapy.
The effect of a colloid depends on its molecular weight. Ninety minutes following administration, a significant proportion of a colloid with a high molecular weight (eg, hydroxyethyl starch) will be retained in the circulation. In contrast, crystalloid solutions (eg, 0.9% saline) readily disappear from the circulation, owing to the ease with which they travel across the cell membrane.6
No evidence of outcome differences
A systematic literature review by Choi et al reflects the current state of knowledge on the relative effects of colloids and crystalloids for fluid resuscitation.7 It concluded that there are no apparent differences between these fluid classes in their effects on pulmonary edema, mortality, or length of stay. The authors noted that methodologic limitations of the available comparative studies prevent meaningful conclusions and that larger randomized controlled trials are needed to detect any differences in outcomes between the two classes.
Although using a crystalloid for fluid resuscitation probably results in a greater volume of fluid given, a study known as SAFE (Saline versus Albumin Fluid Evaluation),8 published after the Choi analysis, showed no differences in 28-day all-cause mortality or other significant outcomes between patients randomized to the colloid (4% albumin) and those assigned to the crystalloid (0.9% saline). Patients receiving the colloid had a higher central venous pressure at all time points, a lower heart rate at the end of the first day, and less overall volume on days 1 and 2 compared with patients receiving the crystalloid. While SAFE was conducted in critically ill patients, these physiologic advantages of the colloid may have implications for results in the perioperative arena, although this remains speculative.
INTRAOPERATIVE MONITORING TO OPTIMIZE FLUID THERAPY
Another important issue is the emergence of minimally invasive technologies for monitoring hemodynamic measures intraoperatively. The aim is to enable more precise tailoring of fluid therapy to meet patient needs on a case-by-case basis.
One of the simplest of these techniques is esophageal Doppler monitoring to measure descending aortic blood flow using Doppler ultrasonography. The technique is used to titrate repeated boluses of fluid based on continuous estimations of stroke volume and surrogate markers of preload indices. Typical protocols for esophageal Doppler monitoring call for administration of colloid to maintain a descending thoracic corrected flow time of no more than 0.35 seconds and stroke volume increments of 10%.
Phan et al recently published a meta-analysis to assess the effect of intraoperative esophageal Doppler monitoring in guiding fluid therapy to optimize intravascular volume status.9 The analysis, which included nine randomized controlled trials in a total of 920 patients, found statistically significant reductions in the rate of complications and in length of hospital stay with the use of esophageal Doppler monitoring; there was no difference in mortality. Use of Doppler monitoring was associated with an increase (+671 mL) in the volume of colloid administered and a decrease (–156 mL) in the volume of crystalloid.
Timing of fluid administration can be critical
One of the trials in the above meta-analysis illustrated that the timing of fluid administration might be more critical than the volume of fluid given. Noblett et al placed an esophageal Doppler probe in each of a series of 108 patients undergoing colorectal resection;10 the control group received perioperative fluid at the anesthesiologist’s discretion, whereas the intervention group received additional colloid boluses based on Doppler assessment. While the overall volume of colloid given was comparable between the two groups, the intervention group received nearly 100% of the total volume during the first quarter of surgery. The intervention group had significantly fewer postoperative complications than the control group as well as a 2-day reduction in average length of stay. Circulating levels of interleukin-6 and cytokines also were significantly lower in the intervention group, which suggests that the intervention blunted the inflammatory response to surgery.
Fluid management must be individualized
Intraoperative fluid needs are highly variable and patient-specific. Parker et al tested an approach in which they universally administered 500 mL of a gelatin colloid solution prior to hip fracture surgery and compared it with a conventional intravenous saline crystalloid solution; neither approach used invasive intraoperative monitoring.11 They found no significant difference in length of stay, 30-day mortality, or postoperative complications between the two study arms. They concluded that more invasive investigation of patients before or during surgery may have been able to identify a subgroup in whom the colloid therapy or more precise fluid control would have been beneficial.
THE ROAD AHEAD
Fluid management remains suboptimal
Despite being a fundamental component of surgical and perioperative care, fluid management remains suboptimal in clinical practice. I can speak most directly to the practice of fluid management in the United Kingdom (UK), but the same types of shortcomings apply broadly to the United States as well.
In 1999, the UK’s National Confidential Enquiry into Patient Outcome and Death examined perioperative death in the UK, concluding that patients were dying as a result of too much or too little perioperative fluid administration.12 Their report cited staff inexperience as an important contributor to the problem, as junior physicians order and deliver the majority of postoperative fluid regimens.
This cautionary report from 10 years ago appears not to have produced substantial improvements in practice, at least according to a recent study by Walsh et al.13 These researchers prospectively audited postoperative fluid management practices in 106 consecutive patients undergoing laparotomy in a UK general surgical unit over a 6-month period in 2003. They found no correlation between available fluid balance data and the quantities of fluids prescribed, suggesting that physicians routinely ignore such data when prescribing. Fifty-four percent of the patients developed at least one fluid-related complication. Patients routinely received significantly greater amounts of fluid and sodium than were physiologically needed, and multivariate analysis showed that mean daily fluid load predicted development of fluid-related complications.
Guidance from a new British consensus document
Where can clinicians turn for a good synthesis of current evidence to guide better perioperative fluid management? I would recommend the newly released British Consensus Guidelines on Intravenous Fluid Therapy for Adult Surgical Patients,14 which are available on the Evidence Based Peri-Operative Medicine Web site (http://www.ebpom.org). These guidelines were developed by a multidisciplinary team of clinicians to improve perioperative fluid prescribing. They cover principles of preoperative, intraoperative, and postoperative fluid management, as well as fluid therapy in acute kidney injury. They present 28 recommendations in all, at least 12 of which are based on high-level (grade 1a or 1b) evidence.
DISCUSSION
Question from the audience: What is the relationship between perioperative fluid management, gut edema from perioperative fluid use, and postoperative ileus?
Dr. Hamilton: There’s no easy answer. Excessive administration of sodium and fluid does predispose to gut and tissue fluid edema. Many of the enhanced surgery recovery programs require no preoperative fasting. There’s no bowel prep. The enteral route is used primarily as quickly as possible. In the UK, we no longer use nasogastric tubes for many of those programs. But there’s no doubt that tissue edema still occurs with excess fluid therapy.
The premise for individualizing fluid therapy is that less is not more but that more is not the right approach either. The stroke volume approaches or the corrected flow time approaches have been related to return of gastrointestinal function and return of flatus, which is a function of gastrointestinal recovery.
Question from the audience: Can you comment on the perioperative use of the Swan-Ganz catheter for fluid management?
Dr. Hamilton: I don’t use it intraoperatively, and not many hospitals in the UK use it apart from liver resection surgery. Having said that, Swan-Ganz catheters were the predominant monitor for 30% to 40% of the original studies of hemodynamic optimization. I cannot give you intraoperative data to support the use of Swan-Ganz catheters for monitoring, but if you lift evidence from the other methods of monitoring hemodynamics, if you’re optimizing flow in a bolus and dynamic fashion, then you should see the kinds of improvements in outcomes that are associated with the other modalities.
The drawback with the Swan-Ganz catheter, obviously, is the morbidity associated with its insertion and its interpretation. But if you’re confident in doing those things, I think it’s a perfectly good monitor.
- Khuri SF, Henderson WG, DePalma RG, et al; Participants in the VA National Surgical Quality Improvement Program. Determinants of long-term survival after major surgery and the adverse effect of postoperative complications. Ann Surg 2005; 242:326–343.
- Boyd AD, Tremblay RE, Spencer FC, Bahnson HT. Estimation of cardiac output soon after intracardiac surgery with cardiopulmonary bypass. Ann Surg 1959; 150:613–626.
- Clowes GH Jr, Del Guercio LR. Circulatory response to trauma of surgical operations. Metabolism 1960; 9:67–81.
- Shoemaker WC, Appel PL, Kram HB, Waxman K, Lee TS. Prospective trial of supranormal values of survivors as therapeutic goals in high-risk surgical patients. Chest 1988; 94:1176–1186.
- Grocott MPW, Hamilton MA, Bennett ED, Harrison D, Rowan K. Perioperative increase in global blood flow to explicit defined goals and outcomes following surgery (Protocol). Cochrane Database Syst Rev 2006; Issue 2. Art. No.: CD004082. doi:10.1002/14651858.CD004082.pub4.
- Lamke LO, Liljedahl SO. Plasma volume changes after infusion of various plasma expanders. Resuscitation 1976; 5:93–102.
- Choi PT, Yip G, Quinonez LG, Cook DJ. Crystalloids vs. colloids in fluid resuscitation: a systematic review. Crit Care Med 1999; 27:200–210.
- The SAFE Study Investigators. A comparison of albumin and saline for fluid resuscitation in the intensive care unit. N Engl J Med 2004; 350:2247–2256.
- Phan TD, Ismail H, Heriot AG, Ho KM. Improving perioperative outcomes: fluid optimization with the esophageal Doppler monitor, a metaanalysis and review. J Am Coll Surg 2008; 207:935–941.
- Noblett SE, Snowden CP, Shenton BK, Horgan AF. Randomized clinical trial assessing the effect of Doppler-optimized fluid management on outcome after elective colorectal resection. Br J Surg 2006; 93:1069–1076.
- Parker MJ, Griffiths R, Boyle A. Preoperative saline versus gelatin for hip fracture patients: a randomized trial of 396 patients. Br J Anaesth 2004; 92:67–70.
- National Confidential Enquiry into Patient Outcome and Death (NCEPOD) Web site. http://www.ncepod.org.uk. Accessed March 25, 2009.
- Walsh SR, Cook EJ, Bentley R, et al. Perioperative fluid management: prospective audit. Int J Clin Pract 2008; 62:492–497.
- Powell-Tuck J, Gosling P, Lobo DN, et al. British consensus guidelines on intravenous fluid therapy for adult surgical patients; 2009. Evidence Based Peri-Operative Medicine (EBPOM) Web site. http://www.ebpom.org. Accessed March 26, 2009.
Perioperative fluid management remains controversial. Until recently, fluid management was guided by targets such as urine output, static pressures, blood pressure, and other physiologic variables. Such physiologic signs, however, are inadequate for detecting subclinical hypovolemia. This has prompted the emergence of an approach to fluid administration based on stroke volume and cardiac output—a “flow-guided” approach—designed to overcome the inadequacies of conventional physiologic signs and improve outcomes. Recent technological advances are permitting noninvasive guidance of intravenous fluid therapy to optimize intravascular volume status.
This article reviews the rationale for perioperative fluid management, strategies for perioperative fluid therapy and their associated outcomes, the types of volume expanders used, and considerations for improving perioperative fluid administration.
WHY FLUID MANAGEMENT MATTERS
Postoperative complications predict survival
In 2005, Khuri et al published a study of survival after major surgery that starkly illustrated the prognostic importance of postoperative complications.1 In an effort to identify predictors of long-term survival, they analyzed a National Surgical Quality Improvement Program database of 105,951 patients who underwent eight common operations at Veterans Administration facilities. They found that the most important determinant of reduced postoperative survival over 8 years of follow-up was the occurrence of a complication within 30 days after surgery. The presence of a postoperative complication was a stronger predictor of death than any intraoperative or preoperative risk factor.
Fluid management is key to preventing complications
Optimizing perioperative fluid management is essential to reducing the risk of postoperative complications and mortality. Surgical patients are more likely to have serious complications and die if they have limited physiologic reserve. Adequate fluid administration may reduce the stress response to surgical trauma and support recovery.
Building on early work showing that survivors of major surgery have consistently higher postoperative cardiac output and oxygen delivery (DO2) than do nonsurvivors,2,3 a seminal study by Shoemaker et al showed that these types of blood flow–related parameters are predictive of both survival and complication-free survival.4 Specifically, Shoemaker and his team showed that a protocol designed to achieve DO2 of at least 600 mL/min/m2 was associated with reductions in both postoperative complications and death.4
PROBLEMS WITH PERIOPERATIVE FLUID THERAPY―AND EFFORTS TO OVERCOME THEM
Despite the utility of fluid management in reducing postoperative complications, perioperative fluid therapy is fraught with several fundamental problems:
- Blood volume cannot be evaluated accurately.
- Fluid overload cannot be identified accurately, apart from tissue edema as a result of gross fluid overload.
- Hypovolemia cannot be identified accurately. Commonly measured variables (heart rate, blood pressure, base excess, lactate) are late markers, and the patient’s status upon admission to the operating room is often unknown.
- Tissue perfusion cannot be evaluated accurately. Although lactate and venous oxygen saturation are surrogate markers, genuinely accurate markers for tissue perfusion are lacking.
For these reasons, fluids are commonly administered without the guidance of direct markers of fluid status.
Assessing flow-guided fluid therapy
These shortcomings prompted me and several other researchers to assess the evidence regarding a flow-guided approach to fluid administration, which aims to achieve maximal cardiac output and stroke volume while avoiding excess fluid administration. We conducted a systematic literature search for randomized controlled trials evaluating the postsurgical effects of perioperative fluid therapy to increase global blood flow to explicitly defined goals, after which we performed a meta-analysis of the 22 qualifying studies.5 The trials collectively included 4,546 patients undergoing relatively high-risk elective or emergency surgery, consisting of general, vascular, cardiac, orthopedic, and urologic procedures. Overall mortality in these trials was 10.6% (481 deaths). The primary outcome assessed was mortality; secondary outcomes included morbidity and length of stay in the hospital and in the intensive care unit. Outcomes were assessed according to the timing of the intervention, the fluid type, and explicit measured goals. Fluids were given to all patients, usually as a dynamic bolus, using a flow-guided approach above and beyond that of the control group.
Our analysis found that a flow-guided protocol was associated with a significant reduction in mortality compared with control protocols (odds ratio = 0.82 [95% CI, 0.67–0.99]; P = .04).5 However, sensitivity analysis showed that the largest and best-designed studies tended to yield no significant differences in mortality between the groups, which highlights the remaining need for larger studies to more definitively clarify the effect on mortality.
Timing of administration (ie, whether fluid was given pre-, intra-, or postoperatively) influenced the primary outcome: compared with control, flow-guided fluid therapy was associated with a significant reduction in mortality only when administered intraoperatively, but not when given preoperatively or postoperatively.5
Length of hospital stay was reduced by approximately 1.6 days with flow-guided therapy compared with control (P < .00001), but there was no significant difference between approaches in terms of intensive care unit stay.5
Postoperative complication rates are difficult to compare, given the lack of a uniform definition of a complication and the relative importance of different complications. Nevertheless, when grouped as a whole, the rate of complications was 48% lower (P < .00001) with flow-guided therapy compared with control. Of all outcomes assessed, the effect on complications was the most consistent among all the studies in the analysis. To provide an example using one easily defined complication, the incidence of renal failure was reduced by 35% with flow-guided therapy compared with control (P = .002).5
COLLOID OR CRYSTALLOID?
Two pharmacologically distinct classes
Intravenous fluids can be broadly classified into colloid and crystalloid solutions, and the relative merits of these two fluid classes are at the center of an enduring debate that predates the advent of flow-based fluid administration. Despite fundamental differences in their pharmacokinetics and other characteristics, colloids and crystalloids are often not sufficiently distinguished from one another in discussions of perioperative fluid therapy.
The effect of a colloid depends on its molecular weight. Ninety minutes following administration, a significant proportion of a colloid with a high molecular weight (eg, hydroxyethyl starch) will be retained in the circulation. In contrast, crystalloid solutions (eg, 0.9% saline) readily disappear from the circulation, owing to the ease with which they travel across the cell membrane.6
No evidence of outcome differences
A systematic literature review by Choi et al reflects the current state of knowledge on the relative effects of colloids and crystalloids for fluid resuscitation.7 It concluded that there are no apparent differences between these fluid classes in their effects on pulmonary edema, mortality, or length of stay. The authors noted that methodologic limitations of the available comparative studies prevent meaningful conclusions and that larger randomized controlled trials are needed to detect any differences in outcomes between the two classes.
Although using a crystalloid for fluid resuscitation probably results in a greater volume of fluid given, a study known as SAFE (Saline versus Albumin Fluid Evaluation),8 published after the Choi analysis, showed no differences in 28-day all-cause mortality or other significant outcomes between patients randomized to the colloid (4% albumin) and those assigned to the crystalloid (0.9% saline). Patients receiving the colloid had a higher central venous pressure at all time points, a lower heart rate at the end of the first day, and less overall volume on days 1 and 2 compared with patients receiving the crystalloid. While SAFE was conducted in critically ill patients, these physiologic advantages of the colloid may have implications for results in the perioperative arena, although this remains speculative.
INTRAOPERATIVE MONITORING TO OPTIMIZE FLUID THERAPY
Another important issue is the emergence of minimally invasive technologies for monitoring hemodynamic measures intraoperatively. The aim is to enable more precise tailoring of fluid therapy to meet patient needs on a case-by-case basis.
One of the simplest of these techniques is esophageal Doppler monitoring to measure descending aortic blood flow using Doppler ultrasonography. The technique is used to titrate repeated boluses of fluid based on continuous estimations of stroke volume and surrogate markers of preload indices. Typical protocols for esophageal Doppler monitoring call for administration of colloid to maintain a descending thoracic corrected flow time of no more than 0.35 seconds and stroke volume increments of 10%.
Phan et al recently published a meta-analysis to assess the effect of intraoperative esophageal Doppler monitoring in guiding fluid therapy to optimize intravascular volume status.9 The analysis, which included nine randomized controlled trials in a total of 920 patients, found statistically significant reductions in the rate of complications and in length of hospital stay with the use of esophageal Doppler monitoring; there was no difference in mortality. Use of Doppler monitoring was associated with an increase (+671 mL) in the volume of colloid administered and a decrease (–156 mL) in the volume of crystalloid.
Timing of fluid administration can be critical
One of the trials in the above meta-analysis illustrated that the timing of fluid administration might be more critical than the volume of fluid given. Noblett et al placed an esophageal Doppler probe in each of a series of 108 patients undergoing colorectal resection;10 the control group received perioperative fluid at the anesthesiologist’s discretion, whereas the intervention group received additional colloid boluses based on Doppler assessment. While the overall volume of colloid given was comparable between the two groups, the intervention group received nearly 100% of the total volume during the first quarter of surgery. The intervention group had significantly fewer postoperative complications than the control group as well as a 2-day reduction in average length of stay. Circulating levels of interleukin-6 and cytokines also were significantly lower in the intervention group, which suggests that the intervention blunted the inflammatory response to surgery.
Fluid management must be individualized
Intraoperative fluid needs are highly variable and patient-specific. Parker et al tested an approach in which they universally administered 500 mL of a gelatin colloid solution prior to hip fracture surgery and compared it with a conventional intravenous saline crystalloid solution; neither approach used invasive intraoperative monitoring.11 They found no significant difference in length of stay, 30-day mortality, or postoperative complications between the two study arms. They concluded that more invasive investigation of patients before or during surgery may have been able to identify a subgroup in whom the colloid therapy or more precise fluid control would have been beneficial.
THE ROAD AHEAD
Fluid management remains suboptimal
Despite being a fundamental component of surgical and perioperative care, fluid management remains suboptimal in clinical practice. I can speak most directly to the practice of fluid management in the United Kingdom (UK), but the same types of shortcomings apply broadly to the United States as well.
In 1999, the UK’s National Confidential Enquiry into Patient Outcome and Death examined perioperative death in the UK, concluding that patients were dying as a result of too much or too little perioperative fluid administration.12 Their report cited staff inexperience as an important contributor to the problem, as junior physicians order and deliver the majority of postoperative fluid regimens.
This cautionary report from 10 years ago appears not to have produced substantial improvements in practice, at least according to a recent study by Walsh et al.13 These researchers prospectively audited postoperative fluid management practices in 106 consecutive patients undergoing laparotomy in a UK general surgical unit over a 6-month period in 2003. They found no correlation between available fluid balance data and the quantities of fluids prescribed, suggesting that physicians routinely ignore such data when prescribing. Fifty-four percent of the patients developed at least one fluid-related complication. Patients routinely received significantly greater amounts of fluid and sodium than were physiologically needed, and multivariate analysis showed that mean daily fluid load predicted development of fluid-related complications.
Guidance from a new British consensus document
Where can clinicians turn for a good synthesis of current evidence to guide better perioperative fluid management? I would recommend the newly released British Consensus Guidelines on Intravenous Fluid Therapy for Adult Surgical Patients,14 which are available on the Evidence Based Peri-Operative Medicine Web site (http://www.ebpom.org). These guidelines were developed by a multidisciplinary team of clinicians to improve perioperative fluid prescribing. They cover principles of preoperative, intraoperative, and postoperative fluid management, as well as fluid therapy in acute kidney injury. They present 28 recommendations in all, at least 12 of which are based on high-level (grade 1a or 1b) evidence.
DISCUSSION
Question from the audience: What is the relationship between perioperative fluid management, gut edema from perioperative fluid use, and postoperative ileus?
Dr. Hamilton: There’s no easy answer. Excessive administration of sodium and fluid does predispose to gut and tissue fluid edema. Many of the enhanced surgery recovery programs require no preoperative fasting. There’s no bowel prep. The enteral route is used primarily as quickly as possible. In the UK, we no longer use nasogastric tubes for many of those programs. But there’s no doubt that tissue edema still occurs with excess fluid therapy.
The premise for individualizing fluid therapy is that less is not more but that more is not the right approach either. The stroke volume approaches or the corrected flow time approaches have been related to return of gastrointestinal function and return of flatus, which is a function of gastrointestinal recovery.
Question from the audience: Can you comment on the perioperative use of the Swan-Ganz catheter for fluid management?
Dr. Hamilton: I don’t use it intraoperatively, and not many hospitals in the UK use it apart from liver resection surgery. Having said that, Swan-Ganz catheters were the predominant monitor for 30% to 40% of the original studies of hemodynamic optimization. I cannot give you intraoperative data to support the use of Swan-Ganz catheters for monitoring, but if you lift evidence from the other methods of monitoring hemodynamics, if you’re optimizing flow in a bolus and dynamic fashion, then you should see the kinds of improvements in outcomes that are associated with the other modalities.
The drawback with the Swan-Ganz catheter, obviously, is the morbidity associated with its insertion and its interpretation. But if you’re confident in doing those things, I think it’s a perfectly good monitor.
Perioperative fluid management remains controversial. Until recently, fluid management was guided by targets such as urine output, static pressures, blood pressure, and other physiologic variables. Such physiologic signs, however, are inadequate for detecting subclinical hypovolemia. This has prompted the emergence of an approach to fluid administration based on stroke volume and cardiac output—a “flow-guided” approach—designed to overcome the inadequacies of conventional physiologic signs and improve outcomes. Recent technological advances are permitting noninvasive guidance of intravenous fluid therapy to optimize intravascular volume status.
This article reviews the rationale for perioperative fluid management, strategies for perioperative fluid therapy and their associated outcomes, the types of volume expanders used, and considerations for improving perioperative fluid administration.
WHY FLUID MANAGEMENT MATTERS
Postoperative complications predict survival
In 2005, Khuri et al published a study of survival after major surgery that starkly illustrated the prognostic importance of postoperative complications.1 In an effort to identify predictors of long-term survival, they analyzed a National Surgical Quality Improvement Program database of 105,951 patients who underwent eight common operations at Veterans Administration facilities. They found that the most important determinant of reduced postoperative survival over 8 years of follow-up was the occurrence of a complication within 30 days after surgery. The presence of a postoperative complication was a stronger predictor of death than any intraoperative or preoperative risk factor.
Fluid management is key to preventing complications
Optimizing perioperative fluid management is essential to reducing the risk of postoperative complications and mortality. Surgical patients are more likely to have serious complications and die if they have limited physiologic reserve. Adequate fluid administration may reduce the stress response to surgical trauma and support recovery.
Building on early work showing that survivors of major surgery have consistently higher postoperative cardiac output and oxygen delivery (DO2) than do nonsurvivors,2,3 a seminal study by Shoemaker et al showed that these types of blood flow–related parameters are predictive of both survival and complication-free survival.4 Specifically, Shoemaker and his team showed that a protocol designed to achieve DO2 of at least 600 mL/min/m2 was associated with reductions in both postoperative complications and death.4
PROBLEMS WITH PERIOPERATIVE FLUID THERAPY―AND EFFORTS TO OVERCOME THEM
Despite the utility of fluid management in reducing postoperative complications, perioperative fluid therapy is fraught with several fundamental problems:
- Blood volume cannot be evaluated accurately.
- Fluid overload cannot be identified accurately, apart from tissue edema as a result of gross fluid overload.
- Hypovolemia cannot be identified accurately. Commonly measured variables (heart rate, blood pressure, base excess, lactate) are late markers, and the patient’s status upon admission to the operating room is often unknown.
- Tissue perfusion cannot be evaluated accurately. Although lactate and venous oxygen saturation are surrogate markers, genuinely accurate markers for tissue perfusion are lacking.
For these reasons, fluids are commonly administered without the guidance of direct markers of fluid status.
Assessing flow-guided fluid therapy
These shortcomings prompted me and several other researchers to assess the evidence regarding a flow-guided approach to fluid administration, which aims to achieve maximal cardiac output and stroke volume while avoiding excess fluid administration. We conducted a systematic literature search for randomized controlled trials evaluating the postsurgical effects of perioperative fluid therapy to increase global blood flow to explicitly defined goals, after which we performed a meta-analysis of the 22 qualifying studies.5 The trials collectively included 4,546 patients undergoing relatively high-risk elective or emergency surgery, consisting of general, vascular, cardiac, orthopedic, and urologic procedures. Overall mortality in these trials was 10.6% (481 deaths). The primary outcome assessed was mortality; secondary outcomes included morbidity and length of stay in the hospital and in the intensive care unit. Outcomes were assessed according to the timing of the intervention, the fluid type, and explicit measured goals. Fluids were given to all patients, usually as a dynamic bolus, using a flow-guided approach above and beyond that of the control group.
Our analysis found that a flow-guided protocol was associated with a significant reduction in mortality compared with control protocols (odds ratio = 0.82 [95% CI, 0.67–0.99]; P = .04).5 However, sensitivity analysis showed that the largest and best-designed studies tended to yield no significant differences in mortality between the groups, which highlights the remaining need for larger studies to more definitively clarify the effect on mortality.
Timing of administration (ie, whether fluid was given pre-, intra-, or postoperatively) influenced the primary outcome: compared with control, flow-guided fluid therapy was associated with a significant reduction in mortality only when administered intraoperatively, but not when given preoperatively or postoperatively.5
Length of hospital stay was reduced by approximately 1.6 days with flow-guided therapy compared with control (P < .00001), but there was no significant difference between approaches in terms of intensive care unit stay.5
Postoperative complication rates are difficult to compare, given the lack of a uniform definition of a complication and the relative importance of different complications. Nevertheless, when grouped as a whole, the rate of complications was 48% lower (P < .00001) with flow-guided therapy compared with control. Of all outcomes assessed, the effect on complications was the most consistent among all the studies in the analysis. To provide an example using one easily defined complication, the incidence of renal failure was reduced by 35% with flow-guided therapy compared with control (P = .002).5
COLLOID OR CRYSTALLOID?
Two pharmacologically distinct classes
Intravenous fluids can be broadly classified into colloid and crystalloid solutions, and the relative merits of these two fluid classes are at the center of an enduring debate that predates the advent of flow-based fluid administration. Despite fundamental differences in their pharmacokinetics and other characteristics, colloids and crystalloids are often not sufficiently distinguished from one another in discussions of perioperative fluid therapy.
The effect of a colloid depends on its molecular weight. Ninety minutes following administration, a significant proportion of a colloid with a high molecular weight (eg, hydroxyethyl starch) will be retained in the circulation. In contrast, crystalloid solutions (eg, 0.9% saline) readily disappear from the circulation, owing to the ease with which they travel across the cell membrane.6
No evidence of outcome differences
A systematic literature review by Choi et al reflects the current state of knowledge on the relative effects of colloids and crystalloids for fluid resuscitation.7 It concluded that there are no apparent differences between these fluid classes in their effects on pulmonary edema, mortality, or length of stay. The authors noted that methodologic limitations of the available comparative studies prevent meaningful conclusions and that larger randomized controlled trials are needed to detect any differences in outcomes between the two classes.
Although using a crystalloid for fluid resuscitation probably results in a greater volume of fluid given, a study known as SAFE (Saline versus Albumin Fluid Evaluation),8 published after the Choi analysis, showed no differences in 28-day all-cause mortality or other significant outcomes between patients randomized to the colloid (4% albumin) and those assigned to the crystalloid (0.9% saline). Patients receiving the colloid had a higher central venous pressure at all time points, a lower heart rate at the end of the first day, and less overall volume on days 1 and 2 compared with patients receiving the crystalloid. While SAFE was conducted in critically ill patients, these physiologic advantages of the colloid may have implications for results in the perioperative arena, although this remains speculative.
INTRAOPERATIVE MONITORING TO OPTIMIZE FLUID THERAPY
Another important issue is the emergence of minimally invasive technologies for monitoring hemodynamic measures intraoperatively. The aim is to enable more precise tailoring of fluid therapy to meet patient needs on a case-by-case basis.
One of the simplest of these techniques is esophageal Doppler monitoring to measure descending aortic blood flow using Doppler ultrasonography. The technique is used to titrate repeated boluses of fluid based on continuous estimations of stroke volume and surrogate markers of preload indices. Typical protocols for esophageal Doppler monitoring call for administration of colloid to maintain a descending thoracic corrected flow time of no more than 0.35 seconds and stroke volume increments of 10%.
Phan et al recently published a meta-analysis to assess the effect of intraoperative esophageal Doppler monitoring in guiding fluid therapy to optimize intravascular volume status.9 The analysis, which included nine randomized controlled trials in a total of 920 patients, found statistically significant reductions in the rate of complications and in length of hospital stay with the use of esophageal Doppler monitoring; there was no difference in mortality. Use of Doppler monitoring was associated with an increase (+671 mL) in the volume of colloid administered and a decrease (–156 mL) in the volume of crystalloid.
Timing of fluid administration can be critical
One of the trials in the above meta-analysis illustrated that the timing of fluid administration might be more critical than the volume of fluid given. Noblett et al placed an esophageal Doppler probe in each of a series of 108 patients undergoing colorectal resection;10 the control group received perioperative fluid at the anesthesiologist’s discretion, whereas the intervention group received additional colloid boluses based on Doppler assessment. While the overall volume of colloid given was comparable between the two groups, the intervention group received nearly 100% of the total volume during the first quarter of surgery. The intervention group had significantly fewer postoperative complications than the control group as well as a 2-day reduction in average length of stay. Circulating levels of interleukin-6 and cytokines also were significantly lower in the intervention group, which suggests that the intervention blunted the inflammatory response to surgery.
Fluid management must be individualized
Intraoperative fluid needs are highly variable and patient-specific. Parker et al tested an approach in which they universally administered 500 mL of a gelatin colloid solution prior to hip fracture surgery and compared it with a conventional intravenous saline crystalloid solution; neither approach used invasive intraoperative monitoring.11 They found no significant difference in length of stay, 30-day mortality, or postoperative complications between the two study arms. They concluded that more invasive investigation of patients before or during surgery may have been able to identify a subgroup in whom the colloid therapy or more precise fluid control would have been beneficial.
THE ROAD AHEAD
Fluid management remains suboptimal
Despite being a fundamental component of surgical and perioperative care, fluid management remains suboptimal in clinical practice. I can speak most directly to the practice of fluid management in the United Kingdom (UK), but the same types of shortcomings apply broadly to the United States as well.
In 1999, the UK’s National Confidential Enquiry into Patient Outcome and Death examined perioperative death in the UK, concluding that patients were dying as a result of too much or too little perioperative fluid administration.12 Their report cited staff inexperience as an important contributor to the problem, as junior physicians order and deliver the majority of postoperative fluid regimens.
This cautionary report from 10 years ago appears not to have produced substantial improvements in practice, at least according to a recent study by Walsh et al.13 These researchers prospectively audited postoperative fluid management practices in 106 consecutive patients undergoing laparotomy in a UK general surgical unit over a 6-month period in 2003. They found no correlation between available fluid balance data and the quantities of fluids prescribed, suggesting that physicians routinely ignore such data when prescribing. Fifty-four percent of the patients developed at least one fluid-related complication. Patients routinely received significantly greater amounts of fluid and sodium than were physiologically needed, and multivariate analysis showed that mean daily fluid load predicted development of fluid-related complications.
Guidance from a new British consensus document
Where can clinicians turn for a good synthesis of current evidence to guide better perioperative fluid management? I would recommend the newly released British Consensus Guidelines on Intravenous Fluid Therapy for Adult Surgical Patients,14 which are available on the Evidence Based Peri-Operative Medicine Web site (http://www.ebpom.org). These guidelines were developed by a multidisciplinary team of clinicians to improve perioperative fluid prescribing. They cover principles of preoperative, intraoperative, and postoperative fluid management, as well as fluid therapy in acute kidney injury. They present 28 recommendations in all, at least 12 of which are based on high-level (grade 1a or 1b) evidence.
DISCUSSION
Question from the audience: What is the relationship between perioperative fluid management, gut edema from perioperative fluid use, and postoperative ileus?
Dr. Hamilton: There’s no easy answer. Excessive administration of sodium and fluid does predispose to gut and tissue fluid edema. Many of the enhanced surgery recovery programs require no preoperative fasting. There’s no bowel prep. The enteral route is used primarily as quickly as possible. In the UK, we no longer use nasogastric tubes for many of those programs. But there’s no doubt that tissue edema still occurs with excess fluid therapy.
The premise for individualizing fluid therapy is that less is not more but that more is not the right approach either. The stroke volume approaches or the corrected flow time approaches have been related to return of gastrointestinal function and return of flatus, which is a function of gastrointestinal recovery.
Question from the audience: Can you comment on the perioperative use of the Swan-Ganz catheter for fluid management?
Dr. Hamilton: I don’t use it intraoperatively, and not many hospitals in the UK use it apart from liver resection surgery. Having said that, Swan-Ganz catheters were the predominant monitor for 30% to 40% of the original studies of hemodynamic optimization. I cannot give you intraoperative data to support the use of Swan-Ganz catheters for monitoring, but if you lift evidence from the other methods of monitoring hemodynamics, if you’re optimizing flow in a bolus and dynamic fashion, then you should see the kinds of improvements in outcomes that are associated with the other modalities.
The drawback with the Swan-Ganz catheter, obviously, is the morbidity associated with its insertion and its interpretation. But if you’re confident in doing those things, I think it’s a perfectly good monitor.
- Khuri SF, Henderson WG, DePalma RG, et al; Participants in the VA National Surgical Quality Improvement Program. Determinants of long-term survival after major surgery and the adverse effect of postoperative complications. Ann Surg 2005; 242:326–343.
- Boyd AD, Tremblay RE, Spencer FC, Bahnson HT. Estimation of cardiac output soon after intracardiac surgery with cardiopulmonary bypass. Ann Surg 1959; 150:613–626.
- Clowes GH Jr, Del Guercio LR. Circulatory response to trauma of surgical operations. Metabolism 1960; 9:67–81.
- Shoemaker WC, Appel PL, Kram HB, Waxman K, Lee TS. Prospective trial of supranormal values of survivors as therapeutic goals in high-risk surgical patients. Chest 1988; 94:1176–1186.
- Grocott MPW, Hamilton MA, Bennett ED, Harrison D, Rowan K. Perioperative increase in global blood flow to explicit defined goals and outcomes following surgery (Protocol). Cochrane Database Syst Rev 2006; Issue 2. Art. No.: CD004082. doi:10.1002/14651858.CD004082.pub4.
- Lamke LO, Liljedahl SO. Plasma volume changes after infusion of various plasma expanders. Resuscitation 1976; 5:93–102.
- Choi PT, Yip G, Quinonez LG, Cook DJ. Crystalloids vs. colloids in fluid resuscitation: a systematic review. Crit Care Med 1999; 27:200–210.
- The SAFE Study Investigators. A comparison of albumin and saline for fluid resuscitation in the intensive care unit. N Engl J Med 2004; 350:2247–2256.
- Phan TD, Ismail H, Heriot AG, Ho KM. Improving perioperative outcomes: fluid optimization with the esophageal Doppler monitor, a metaanalysis and review. J Am Coll Surg 2008; 207:935–941.
- Noblett SE, Snowden CP, Shenton BK, Horgan AF. Randomized clinical trial assessing the effect of Doppler-optimized fluid management on outcome after elective colorectal resection. Br J Surg 2006; 93:1069–1076.
- Parker MJ, Griffiths R, Boyle A. Preoperative saline versus gelatin for hip fracture patients: a randomized trial of 396 patients. Br J Anaesth 2004; 92:67–70.
- National Confidential Enquiry into Patient Outcome and Death (NCEPOD) Web site. http://www.ncepod.org.uk. Accessed March 25, 2009.
- Walsh SR, Cook EJ, Bentley R, et al. Perioperative fluid management: prospective audit. Int J Clin Pract 2008; 62:492–497.
- Powell-Tuck J, Gosling P, Lobo DN, et al. British consensus guidelines on intravenous fluid therapy for adult surgical patients; 2009. Evidence Based Peri-Operative Medicine (EBPOM) Web site. http://www.ebpom.org. Accessed March 26, 2009.
- Khuri SF, Henderson WG, DePalma RG, et al; Participants in the VA National Surgical Quality Improvement Program. Determinants of long-term survival after major surgery and the adverse effect of postoperative complications. Ann Surg 2005; 242:326–343.
- Boyd AD, Tremblay RE, Spencer FC, Bahnson HT. Estimation of cardiac output soon after intracardiac surgery with cardiopulmonary bypass. Ann Surg 1959; 150:613–626.
- Clowes GH Jr, Del Guercio LR. Circulatory response to trauma of surgical operations. Metabolism 1960; 9:67–81.
- Shoemaker WC, Appel PL, Kram HB, Waxman K, Lee TS. Prospective trial of supranormal values of survivors as therapeutic goals in high-risk surgical patients. Chest 1988; 94:1176–1186.
- Grocott MPW, Hamilton MA, Bennett ED, Harrison D, Rowan K. Perioperative increase in global blood flow to explicit defined goals and outcomes following surgery (Protocol). Cochrane Database Syst Rev 2006; Issue 2. Art. No.: CD004082. doi:10.1002/14651858.CD004082.pub4.
- Lamke LO, Liljedahl SO. Plasma volume changes after infusion of various plasma expanders. Resuscitation 1976; 5:93–102.
- Choi PT, Yip G, Quinonez LG, Cook DJ. Crystalloids vs. colloids in fluid resuscitation: a systematic review. Crit Care Med 1999; 27:200–210.
- The SAFE Study Investigators. A comparison of albumin and saline for fluid resuscitation in the intensive care unit. N Engl J Med 2004; 350:2247–2256.
- Phan TD, Ismail H, Heriot AG, Ho KM. Improving perioperative outcomes: fluid optimization with the esophageal Doppler monitor, a metaanalysis and review. J Am Coll Surg 2008; 207:935–941.
- Noblett SE, Snowden CP, Shenton BK, Horgan AF. Randomized clinical trial assessing the effect of Doppler-optimized fluid management on outcome after elective colorectal resection. Br J Surg 2006; 93:1069–1076.
- Parker MJ, Griffiths R, Boyle A. Preoperative saline versus gelatin for hip fracture patients: a randomized trial of 396 patients. Br J Anaesth 2004; 92:67–70.
- National Confidential Enquiry into Patient Outcome and Death (NCEPOD) Web site. http://www.ncepod.org.uk. Accessed March 25, 2009.
- Walsh SR, Cook EJ, Bentley R, et al. Perioperative fluid management: prospective audit. Int J Clin Pract 2008; 62:492–497.
- Powell-Tuck J, Gosling P, Lobo DN, et al. British consensus guidelines on intravenous fluid therapy for adult surgical patients; 2009. Evidence Based Peri-Operative Medicine (EBPOM) Web site. http://www.ebpom.org. Accessed March 26, 2009.
KEY POINTS
- A flow-guided approach to fluid administration is associated with reductions in mortality, postoperative complications, and length of stay compared with fluid management guided by traditional physiologic targets.
- Studies to date have shown no consistent difference between colloids and crystalloids in their effects on clinical outcomes.
- Intraoperative esophageal Doppler monitoring is a simple technique for titrating boluses of fluid based on continuous estimations of stroke volume.
- Administration of sufficient fluids early in the course of surgery may be more important than the total volume of fluid administered in improving patient outcomes.
- Intraoperative fluid needs are highly variable, underscoring the need for individual monitoring and assessment.
Giving anesthesiologists what they want: How to write a useful preoperative consult
The ideal preoperative medical consultation (consult) is useful to the whole surgical team, ensures maximal patient readiness for surgery, and promotes optimal perioperative care of the patient. Too often, however, consults are ignored or, even worse, set the stage for legal problems. This article identifies problems frequently seen in preoperative consults, particularly from the perspective of anesthesiologists, and gives guidance to those who write consults—hospitalists, internists, cardiologists, and other medical consultants—on providing the information that is most needed by those who use them.
A WIDE RANGE OF END USERS
Anesthesiologists are most often the primary users of the information in preoperative consults, but many other members of the surgical and perioperative team benefit from a well-developed consult, including surgeons, intensivists, nurses, and pain management specialists. Most important, patients stand to benefit, as a good consult helps to ensure that the full breadth of relevant patient-specific information is brought to bear to anticipate potential difficulties and promote optimal care.
Purpose of a consult is in the eye of the beholder
The literature on medical consults in the perioperative arena is scant. The only fairly recent assessment of physician attitudes toward the role of consults was reported by Katz et al in 1998.1 These researchers surveyed attitudes about the various perceived purposes of preoperative cardiology consults, and received rather different responses from anesthesiologists, cardiologists, and surgeons.
There was consensus among all three specialties that two particular functions of a consult are important:
- Treating an inadequately managed cardiac condition before surgery
- Providing data to use in anesthetic management.
Additionally, all three specialties deemed the suggestion of intraoperative treatment modalities to be reasonably important when such suggestions were specifically included in the consult request, although anesthesiologists assigned less importance to this function.1
In contrast, anesthesiologists considered suggestions about intraoperative treatment generally unimportant when not specifically requested, and they viewed suggestions of intraoperative monitoring and advice on the safest type of anesthesia as even less important. Anesthesiologists also deemed “clearing the patient for surgery” as an unimportant function of the consult. Cardiologists rated all of these functions as more important than anesthesiologists did and in some cases as considerably more important. To many of the survey questions, surgeons responded that a specific purpose of a consult was “neither important nor unimportant.”1 That may be because the surgeon’s purpose in obtaining the consult is often simply to address the concerns of the anesthesiologist, who might otherwise delay or cancel a needed surgery.
Consult deficiencies: Vagueness, illegibility, dictating anesthetic choice
The survey by Katz et al also assessed each specialty’s perceptions of the most common deficiencies of preoperative cardiology consults. The deficiencies deemed most common were failure to give specific facts, illegible handwriting, and attempts to dictate the type of anesthesia to be used. Anesthesiologists considered each of the deficiencies assessed as occurring more commonly than their cardiologist or surgeon colleagues did.1
The requester–user disconnect
The differing perceptions of preoperative consults by anesthesiologists and surgeons underscore a fundamental problem: the primary requesters of consults (surgeons) are different from the primary users of consults (anesthesiologists).
Ideally, preoperative consults should be requested by anesthesiologists. Unless and until the ordering of consults changes on a wide scale, however, our advice is for consultants to ask the anesthesiologist what he or she needs to know, in addition to any questions directed to the requesting surgeon. Communication between the surgeon and anesthesiologist should be encouraged as much as possible, and consultants should keep both the anesthesiologist and surgeon in mind when writing consult notes.
A final end user: The plaintiff’s attorney
It is wise to keep in mind one more potential user of your consult: a plaintiff’s attorney. A poorly written consult may benefit plaintiffs’ lawyers. Consults should never give absolute instructions; it is better to use such phrases as “Strongly consider…” or “The current literature strongly suggests…” Otherwise, the surgical team is placed in an awkward position if it does not follow your recommendations, even if for good reason. If a certain recommendation absolutely must be followed, then direct oral communication from the consultant to the attending anesthesiologist (or surgeon) is best.
CONSIDER THE PRIMARY USER: WHAT ANESTHESIOLOGISTS ALREADY KNOW
For the purpose of preoperative consults, it is helpful to think of anesthesiologists as experts in acute medical care. Their 4-year training consists of the following:
- 1 year of internship, often in medicine, including 6 months of basic patient care in the ward or clinic (the last time they will manage chronic disease)
- 4 months in the intensive care unit (ICU) and 1 month in the recovery room, which yields solid intensivist training
- 3 months in pain management, covering acute and chronic pain and regional blocks
- ~24 months in the operating room, often devoted to care of complex problems in surgical subspecialties (obstetric, pediatric, neurologic, cardiothoracic, vascular)
- 1 month of preoperative screening and consultations (a recent requirement).
An optional fifth year may be spent in a subspecialty.
Since the large part of anesthesiologists’ training is in acute care, they generally do not need advice about the acute treatment of any ailment. Consults should not advise anesthesiologists on subjects in which they have considerable expertise. They already have well-established ideas about addressing hypertension, myocardial ischemia, heart failure, arrhythmias (unless unusual therapies are needed), bronchospasm, glucose levels, and pain in the operating room, so they are apt to ignore advice on such topics.
There are several additional topics in which anesthesiologists have considerable expertise and do not need guidance in consults:
- Choice of anesthetic type and its impact on outcome
- Choice of invasive or noninvasive monitoring for any comorbidity and operation
- Postoperative patient disposition (ie, whether to send a patient home, to the postanesthesia care unit, to the ICU, or to a step-down unit)
- Impact of optimizing organ function on perioperative outcome
- Cardiovascular and respiratory physiology
- Pharmacology of intravenous agents.
WHAT ANESTHESIOLOGISTS MAY NOT KNOW―AND NEED FROM CONSULTANTS
How to manage chronic diseases
Follow-up care (eg, for poorly controlled diabetes or hypertension) often can wait until after the operation, and a consultant’s opinion about that is appreciated. It is especially helpful to know that the patient will be followed without the surgeon or anesthesiologist having to arrange for it.
New evidence-based guidance from the literature
One case when recommendations on acute medical management should be provided is when they involve new information from the literature—ie, important data or guidelines published within the prior 5 years or so. It can take time for new information and recommendations to reach all clinicians even within a single specialty. Moreover, important information, such as on the perioperative use of beta-blockers and statins, is not necessarily published in the anesthesiology literature. It is critical to relay information such as the recent recommendation not to withdraw statins prior to surgery, as the current editions of most anesthesiology textbooks have incorrect information suggesting discontinuation. Thus, consultants should include pertinent recent data and guideline recommendations, especially if they go against previous dogma.
Rare diseases, blood disorders, other special cases
As outlined in Table 1, advice on perioperative management is appreciated for patients with rare diseases, coagulation disorders or other blood disorders, and brittle diabetes and other endocrine disorders, as most anesthesiologists are not intimately familiar with these conditions. Anesthesiologists also need, but often do not get, basic details on coronary stents and other implanted devices (see Table 1), as well as guidance on the latest anticoagulation recommendations, with which it is difficult to keep up to date.
A sensitivity to audience and context
It is always appropriate to ask the surgeon requesting a consult—and the anesthesiologist assigned to the case, if known—what he or she wants to know from the consult. If guidance on specific cases is impractical, it is appropriate to ask the chair of the anesthesiology department, or several anesthesiologists collectively, for general guidance on what they look for from preoperative consults.
Anesthesiologists, like consultants, comprise a broad range of people, and it is always important to be sensitive to context. Generalists who work mainly on healthy patients or in a community setting may have forgotten some of their training in acute medicine and are more likely to appreciate advice on intraoperative care. On the other hand, an anesthesiologist who trained in a cardiothoracic subspecialty fellowship, who routinely deals with issues such as left ventricular assist devices and heart transplants, would not benefit from such advice.
WHAT A CONSULT SHOULD―AND SHOULD NOT―BE
The above advice can be distilled into a few principles:
- A consult is an opportunity for the medical consultant to provide helpful management suggestions to the operative team.
- A fundamental objective of a consult is to optimize a patient’s underlying disease before it is compounded by the insult of surgery.
- The purpose of a consult is never to “clear” a patient for surgery. Whether or not to proceed to surgery is a question for the anesthesiologist, surgeon, and patient to decide after weighing the risks and benefits once the patient’s comorbidities are optimally managed. The consult is an important contributing factor to this decision, but it should never be the mechanism of the decision. Though the note from the surgeon requesting a consult may routinely be written as, “Clear the patient for surgery,” consultants should recognize this for what it is—the surgeon’s attempt to avoid having the anesthesiologist cancel the operation—and refrain from weighing in on “clearance” one way or the other.
CASE STUDY: CARDIAC CONSULT REQUESTED BEFORE FEM-POP BYPASS SURGERY
Cardiovascular problems are the most common reasons for requesting preoperative consults. The following case illustrates a typical scenario for a cardiac consult request and presents examples of good and bad notes requesting consults and good and bad consults written in response.
The case
A 47-year-old man is scheduled for femoral-popliteal bypass surgery. His medical history is significant for diabetes, a myocardial infarction (MI) 3 years ago followed by placement of a stent, and a limited ability to assess exercise tolerance. Evidence of an anteroseptal MI is present on 12-lead electrocardiography. His medications include metoprolol 25 mg twice daily and an oral hypoglycemic agent. His blood pressure is 152/89 mm Hg, heart rate 81 beats per minute, respiratory rate 14 breaths per minute, and arterial oxygen saturation 96% on room air.
The consult request: Bad and good examples
A bad consult request in this case would be, “Clear the patient for surgery.” Although this type of request is routinely written, it is routinely useless.
For this complex surgery with significant fluid requirements, a much better consult request would include several specific requests and questions and might read as follows:
—Please evaluate patient’s post-MI therapy for his CAD. Is further therapy required to optimize CAD treatment?
—Do his blood pressure or diabetic regimens need modification? If so, can this be done postoperatively?
—Please evaluate patient’s myocardial function in light of a lack of info on exercise tolerance. Is an echo indicated?
—Are other tests, therapies, or interventions warranted pre- or postoperatively?
Example of a bad consult
A poorly written consult in a case like this may:
- Include a brief history repeating facts that are already known and noting that “the patient is at his baseline without obvious ischemia.”
- State that the patient is cleared for “spinal” anesthesia. “Clearing a patient for anesthesia” is useless to begin with, but clearing for a certain type of anesthesia places the anesthesiologist in a terrible medicolegal position if general anesthesia turns out to be needed. Moreover, there are no proven major outcome differences related to the type of anesthetic chosen.
- Recommend that “a pulmonary artery catheter might be indicated to monitor hemodynamics.” Besides the fact that such catheters probably do more harm than good, such a recommendation is unnecessary since the anesthesiologist is already expert in managing perioperative care.
- Recommend that “the anesthesia team should monitor the patient carefully in the perioperative arena for hypoxia and hypotension.” Qualitative advice, such as “avoid hypoxia, hypotension, and tachycardia,” is not useful, but quantitative information, such as “during ischemic stress testing, the patient exhibited ischemia when his heart rate went to 142,” can be very helpful.
- State that the patient be sent to the ICU following surgery. Mandating an ICU stay in advance makes no sense unless the operation itself demands ICU care, which is the call of the surgeon and anesthesiologist anyway.
A consult like this doesn’t tell the perioperative team anything that it didn’t already know. Such a consult not only is unhelpful but also actually creates more work since much of the advice needs to be “undone” lest a lawyer see the chart and it not be clear why the consultant’s recommendations were not followed.
Example of a good consult
In contrast, a good consult for this case would involve:
- A detailed history examining the potential for silent ischemia associated with the diabetes, as well as the relationship of the hypertension and beta-blocker therapy to episodes of ischemia. The level of ischemia should be clearly categorized. If it cannot be determined, this should be noted; if it can be determined only that the ischemia is not New York Heart Association class III or IV, note this as well (the perioperative outcomes literature suggests that no preoperative ischemia testing is needed with class I or II angina).
- Guidance on blood pressure optimization in light of the relative urgency of the procedure. Blood pressure need not be normalized preoperatively in this case, but if the operation were totally elective and the consultant felt it could make a difference, it would be appropriate to suggest that blood pressure be optimized beforehand.
- A recommendation on whether and when to change the beta-blocker dosage. If the dosage needs to be increased, the anesthesiologist will want to know how many doses are needed to reach a new steady state. Joint guidelines from the American College of Cardiology (ACC) and American Heart Association (AHA)2 recommend 7 to 30 days, but this time frame is unrealistic in this setting, so more practical advice would be appreciated. A good consult notes any deviation from established guidelines, however, and explains the rationale for such deviation.
- Evaluation of the myocardium at risk. This is especially important with left main disease, as it influences the decision whether to test or intervene versus proceeding with careful beta-blocker titration.
- Evaluation of myocardial function and, if appropriate, a therapy suggestion for optimization.
- Notation of the heart rate or blood pressure thresholds at which ischemia develops if a stress test is performed.
GOOD GUIDANCE FROM THE ACC/AHA GUIDELINES
Our advice here is broadly consistent with the aforementioned 2007 ACC/AHA guidelines on perioperative cardiovascular evaluation for noncardiac surgery.2 The following observation on cardiac evaluations from these guidelines applies to preoperative consults in general:
The purpose of preoperative evaluation is not to give medical clearance but rather to perform an evaluation of the patient’s current medical status; make recommendations concerning the evaluation, management, and risk of cardiac problems over the entire perioperative period; and provide a clinical risk profile that [can be used] in making treatment decisions that may influence short- and long-term cardiac outcomes.2
These guidelines contain a good description of the ideal preoperative evaluation and consult in a short section (Section II, “General Approach to the Patient”)2 that is worthy of wide dissemination.
DISCUSSION
Question from the audience: Many consults are written more for the surgical team than for the anesthesiologists, hence advice such as managing intraoperative diabetes. Isn’t that appropriate?
Dr. Lubarsky: There are a variety of users of the information in a consult note. I focused on the anesthesiologist, but certainly the surgical staff and house staff would benefit from suggestions about postoperative management. However, they would not benefit from suggestions on intraoperative management; surgeons simply do not need this information and the anesthesiologist will have his or her own regimen. But if there is a specific type of insulin infusion that’s been shown to be best in the specific patient at hand, then detailing that obviously is beneficial.
Question from the audience: We all agree that communication is key, but how does the consultant reach the anesthesiologist to find out what he or she wants to know when the anesthesiologist isn’t usually assigned to the case until a day before surgery?
Dr. Lubarsky: If no anesthesiologist is yet assigned to a case, the consultant can discuss the case with the chief of the anesthesiology department. The discussion should be documented in the note. But it’s important that the system be changed so that anesthesiologists are assigned to cases well in advance. I instituted such a policy at my previous hospital. Many hospitals schedule surgeries 3 months in advance, and many anesthesiology departments have schedules made at least 1 month and often 2 to 3 months in advance. The department could assign a specific anesthesiologist to a future scheduled case with ease.
Question from the audience: How do anesthesiologists educate all the various people we rely on for consults when we can’t get them in one place at one time?
Dr. Lubarsky: It’s a challenge. I try many things, such as going to cardiology rounds, but there are always new people coming through. A good monograph or a set of guidelines with examples would help. If each specialty educates the other and speaks at each other’s conferences more often, that should help. Anesthesiologists would benefit from hearing about the challenges medical consultants face; we may not be doing all we can to optimize perioperative care. There’s room for improvement through communication on both sides. I should also emphasize that we’re all trying to do the right thing. Doctors try to be accommodating, but that doesn’t always make for good decisions. Recently a consultant in my hospital did a preoperative stress test on a patient who didn’t need one. When I asked why, he said, “Because the surgeon asked me to.”
Question from the audience: But don’t you agree that many anesthesiologists would like to see that negative stress test, even if a stress test is not indicated by the guidelines? Cardiologists know that the anesthesiologists are often looking for that on the morning of surgery.
Dr. Lubarsky: The point is that physicians should be responsible for what they have expertise in. When I am asked to intubate a patient, my response as an expert in intubation might be, “Actually, he doesn’t need to be intubated right now.” In the case of this unnecessary stress test, the cardiologist probably should have called the surgeon and said, “It’s really not indicated because the patient had a negative stress test 2 years ago, there’s been no change in symptoms and no angina since then, and he operates well above 4 metabolic equivalents. There’s a clear-cut reason not to do it.” If the surgeon still wanted the test done just to be reassured, that’s simply a poor use of society’s resources. We depend on experts to identify the tests that are indicated to evaluate a patient’s disease and not just do tests for the sake of doing them.
- Katz RI, Barnhart JM, Ho G, et al. A survey on the intended purposes and perceived utility of preoperative cardiology consultations. Anesth Analg 1998; 87:830–836.
- Fleisher LA, Beckman JA, Brown KA, et al. ACC/AHA 2007 guidelines on perioperative cardiovascular evaluation and care for noncardiac surgery: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. Circulation 2007; 116:e418–e499.
The ideal preoperative medical consultation (consult) is useful to the whole surgical team, ensures maximal patient readiness for surgery, and promotes optimal perioperative care of the patient. Too often, however, consults are ignored or, even worse, set the stage for legal problems. This article identifies problems frequently seen in preoperative consults, particularly from the perspective of anesthesiologists, and gives guidance to those who write consults—hospitalists, internists, cardiologists, and other medical consultants—on providing the information that is most needed by those who use them.
A WIDE RANGE OF END USERS
Anesthesiologists are most often the primary users of the information in preoperative consults, but many other members of the surgical and perioperative team benefit from a well-developed consult, including surgeons, intensivists, nurses, and pain management specialists. Most important, patients stand to benefit, as a good consult helps to ensure that the full breadth of relevant patient-specific information is brought to bear to anticipate potential difficulties and promote optimal care.
Purpose of a consult is in the eye of the beholder
The literature on medical consults in the perioperative arena is scant. The only fairly recent assessment of physician attitudes toward the role of consults was reported by Katz et al in 1998.1 These researchers surveyed attitudes about the various perceived purposes of preoperative cardiology consults, and received rather different responses from anesthesiologists, cardiologists, and surgeons.
There was consensus among all three specialties that two particular functions of a consult are important:
- Treating an inadequately managed cardiac condition before surgery
- Providing data to use in anesthetic management.
Additionally, all three specialties deemed the suggestion of intraoperative treatment modalities to be reasonably important when such suggestions were specifically included in the consult request, although anesthesiologists assigned less importance to this function.1
In contrast, anesthesiologists considered suggestions about intraoperative treatment generally unimportant when not specifically requested, and they viewed suggestions of intraoperative monitoring and advice on the safest type of anesthesia as even less important. Anesthesiologists also deemed “clearing the patient for surgery” as an unimportant function of the consult. Cardiologists rated all of these functions as more important than anesthesiologists did and in some cases as considerably more important. To many of the survey questions, surgeons responded that a specific purpose of a consult was “neither important nor unimportant.”1 That may be because the surgeon’s purpose in obtaining the consult is often simply to address the concerns of the anesthesiologist, who might otherwise delay or cancel a needed surgery.
Consult deficiencies: Vagueness, illegibility, dictating anesthetic choice
The survey by Katz et al also assessed each specialty’s perceptions of the most common deficiencies of preoperative cardiology consults. The deficiencies deemed most common were failure to give specific facts, illegible handwriting, and attempts to dictate the type of anesthesia to be used. Anesthesiologists considered each of the deficiencies assessed as occurring more commonly than their cardiologist or surgeon colleagues did.1
The requester–user disconnect
The differing perceptions of preoperative consults by anesthesiologists and surgeons underscore a fundamental problem: the primary requesters of consults (surgeons) are different from the primary users of consults (anesthesiologists).
Ideally, preoperative consults should be requested by anesthesiologists. Unless and until the ordering of consults changes on a wide scale, however, our advice is for consultants to ask the anesthesiologist what he or she needs to know, in addition to any questions directed to the requesting surgeon. Communication between the surgeon and anesthesiologist should be encouraged as much as possible, and consultants should keep both the anesthesiologist and surgeon in mind when writing consult notes.
A final end user: The plaintiff’s attorney
It is wise to keep in mind one more potential user of your consult: a plaintiff’s attorney. A poorly written consult may benefit plaintiffs’ lawyers. Consults should never give absolute instructions; it is better to use such phrases as “Strongly consider…” or “The current literature strongly suggests…” Otherwise, the surgical team is placed in an awkward position if it does not follow your recommendations, even if for good reason. If a certain recommendation absolutely must be followed, then direct oral communication from the consultant to the attending anesthesiologist (or surgeon) is best.
CONSIDER THE PRIMARY USER: WHAT ANESTHESIOLOGISTS ALREADY KNOW
For the purpose of preoperative consults, it is helpful to think of anesthesiologists as experts in acute medical care. Their 4-year training consists of the following:
- 1 year of internship, often in medicine, including 6 months of basic patient care in the ward or clinic (the last time they will manage chronic disease)
- 4 months in the intensive care unit (ICU) and 1 month in the recovery room, which yields solid intensivist training
- 3 months in pain management, covering acute and chronic pain and regional blocks
- ~24 months in the operating room, often devoted to care of complex problems in surgical subspecialties (obstetric, pediatric, neurologic, cardiothoracic, vascular)
- 1 month of preoperative screening and consultations (a recent requirement).
An optional fifth year may be spent in a subspecialty.
Since the large part of anesthesiologists’ training is in acute care, they generally do not need advice about the acute treatment of any ailment. Consults should not advise anesthesiologists on subjects in which they have considerable expertise. They already have well-established ideas about addressing hypertension, myocardial ischemia, heart failure, arrhythmias (unless unusual therapies are needed), bronchospasm, glucose levels, and pain in the operating room, so they are apt to ignore advice on such topics.
There are several additional topics in which anesthesiologists have considerable expertise and do not need guidance in consults:
- Choice of anesthetic type and its impact on outcome
- Choice of invasive or noninvasive monitoring for any comorbidity and operation
- Postoperative patient disposition (ie, whether to send a patient home, to the postanesthesia care unit, to the ICU, or to a step-down unit)
- Impact of optimizing organ function on perioperative outcome
- Cardiovascular and respiratory physiology
- Pharmacology of intravenous agents.
WHAT ANESTHESIOLOGISTS MAY NOT KNOW―AND NEED FROM CONSULTANTS
How to manage chronic diseases
Follow-up care (eg, for poorly controlled diabetes or hypertension) often can wait until after the operation, and a consultant’s opinion about that is appreciated. It is especially helpful to know that the patient will be followed without the surgeon or anesthesiologist having to arrange for it.
New evidence-based guidance from the literature
One case when recommendations on acute medical management should be provided is when they involve new information from the literature—ie, important data or guidelines published within the prior 5 years or so. It can take time for new information and recommendations to reach all clinicians even within a single specialty. Moreover, important information, such as on the perioperative use of beta-blockers and statins, is not necessarily published in the anesthesiology literature. It is critical to relay information such as the recent recommendation not to withdraw statins prior to surgery, as the current editions of most anesthesiology textbooks have incorrect information suggesting discontinuation. Thus, consultants should include pertinent recent data and guideline recommendations, especially if they go against previous dogma.
Rare diseases, blood disorders, other special cases
As outlined in Table 1, advice on perioperative management is appreciated for patients with rare diseases, coagulation disorders or other blood disorders, and brittle diabetes and other endocrine disorders, as most anesthesiologists are not intimately familiar with these conditions. Anesthesiologists also need, but often do not get, basic details on coronary stents and other implanted devices (see Table 1), as well as guidance on the latest anticoagulation recommendations, with which it is difficult to keep up to date.
A sensitivity to audience and context
It is always appropriate to ask the surgeon requesting a consult—and the anesthesiologist assigned to the case, if known—what he or she wants to know from the consult. If guidance on specific cases is impractical, it is appropriate to ask the chair of the anesthesiology department, or several anesthesiologists collectively, for general guidance on what they look for from preoperative consults.
Anesthesiologists, like consultants, comprise a broad range of people, and it is always important to be sensitive to context. Generalists who work mainly on healthy patients or in a community setting may have forgotten some of their training in acute medicine and are more likely to appreciate advice on intraoperative care. On the other hand, an anesthesiologist who trained in a cardiothoracic subspecialty fellowship, who routinely deals with issues such as left ventricular assist devices and heart transplants, would not benefit from such advice.
WHAT A CONSULT SHOULD―AND SHOULD NOT―BE
The above advice can be distilled into a few principles:
- A consult is an opportunity for the medical consultant to provide helpful management suggestions to the operative team.
- A fundamental objective of a consult is to optimize a patient’s underlying disease before it is compounded by the insult of surgery.
- The purpose of a consult is never to “clear” a patient for surgery. Whether or not to proceed to surgery is a question for the anesthesiologist, surgeon, and patient to decide after weighing the risks and benefits once the patient’s comorbidities are optimally managed. The consult is an important contributing factor to this decision, but it should never be the mechanism of the decision. Though the note from the surgeon requesting a consult may routinely be written as, “Clear the patient for surgery,” consultants should recognize this for what it is—the surgeon’s attempt to avoid having the anesthesiologist cancel the operation—and refrain from weighing in on “clearance” one way or the other.
CASE STUDY: CARDIAC CONSULT REQUESTED BEFORE FEM-POP BYPASS SURGERY
Cardiovascular problems are the most common reasons for requesting preoperative consults. The following case illustrates a typical scenario for a cardiac consult request and presents examples of good and bad notes requesting consults and good and bad consults written in response.
The case
A 47-year-old man is scheduled for femoral-popliteal bypass surgery. His medical history is significant for diabetes, a myocardial infarction (MI) 3 years ago followed by placement of a stent, and a limited ability to assess exercise tolerance. Evidence of an anteroseptal MI is present on 12-lead electrocardiography. His medications include metoprolol 25 mg twice daily and an oral hypoglycemic agent. His blood pressure is 152/89 mm Hg, heart rate 81 beats per minute, respiratory rate 14 breaths per minute, and arterial oxygen saturation 96% on room air.
The consult request: Bad and good examples
A bad consult request in this case would be, “Clear the patient for surgery.” Although this type of request is routinely written, it is routinely useless.
For this complex surgery with significant fluid requirements, a much better consult request would include several specific requests and questions and might read as follows:
—Please evaluate patient’s post-MI therapy for his CAD. Is further therapy required to optimize CAD treatment?
—Do his blood pressure or diabetic regimens need modification? If so, can this be done postoperatively?
—Please evaluate patient’s myocardial function in light of a lack of info on exercise tolerance. Is an echo indicated?
—Are other tests, therapies, or interventions warranted pre- or postoperatively?
Example of a bad consult
A poorly written consult in a case like this may:
- Include a brief history repeating facts that are already known and noting that “the patient is at his baseline without obvious ischemia.”
- State that the patient is cleared for “spinal” anesthesia. “Clearing a patient for anesthesia” is useless to begin with, but clearing for a certain type of anesthesia places the anesthesiologist in a terrible medicolegal position if general anesthesia turns out to be needed. Moreover, there are no proven major outcome differences related to the type of anesthetic chosen.
- Recommend that “a pulmonary artery catheter might be indicated to monitor hemodynamics.” Besides the fact that such catheters probably do more harm than good, such a recommendation is unnecessary since the anesthesiologist is already expert in managing perioperative care.
- Recommend that “the anesthesia team should monitor the patient carefully in the perioperative arena for hypoxia and hypotension.” Qualitative advice, such as “avoid hypoxia, hypotension, and tachycardia,” is not useful, but quantitative information, such as “during ischemic stress testing, the patient exhibited ischemia when his heart rate went to 142,” can be very helpful.
- State that the patient be sent to the ICU following surgery. Mandating an ICU stay in advance makes no sense unless the operation itself demands ICU care, which is the call of the surgeon and anesthesiologist anyway.
A consult like this doesn’t tell the perioperative team anything that it didn’t already know. Such a consult not only is unhelpful but also actually creates more work since much of the advice needs to be “undone” lest a lawyer see the chart and it not be clear why the consultant’s recommendations were not followed.
Example of a good consult
In contrast, a good consult for this case would involve:
- A detailed history examining the potential for silent ischemia associated with the diabetes, as well as the relationship of the hypertension and beta-blocker therapy to episodes of ischemia. The level of ischemia should be clearly categorized. If it cannot be determined, this should be noted; if it can be determined only that the ischemia is not New York Heart Association class III or IV, note this as well (the perioperative outcomes literature suggests that no preoperative ischemia testing is needed with class I or II angina).
- Guidance on blood pressure optimization in light of the relative urgency of the procedure. Blood pressure need not be normalized preoperatively in this case, but if the operation were totally elective and the consultant felt it could make a difference, it would be appropriate to suggest that blood pressure be optimized beforehand.
- A recommendation on whether and when to change the beta-blocker dosage. If the dosage needs to be increased, the anesthesiologist will want to know how many doses are needed to reach a new steady state. Joint guidelines from the American College of Cardiology (ACC) and American Heart Association (AHA)2 recommend 7 to 30 days, but this time frame is unrealistic in this setting, so more practical advice would be appreciated. A good consult notes any deviation from established guidelines, however, and explains the rationale for such deviation.
- Evaluation of the myocardium at risk. This is especially important with left main disease, as it influences the decision whether to test or intervene versus proceeding with careful beta-blocker titration.
- Evaluation of myocardial function and, if appropriate, a therapy suggestion for optimization.
- Notation of the heart rate or blood pressure thresholds at which ischemia develops if a stress test is performed.
GOOD GUIDANCE FROM THE ACC/AHA GUIDELINES
Our advice here is broadly consistent with the aforementioned 2007 ACC/AHA guidelines on perioperative cardiovascular evaluation for noncardiac surgery.2 The following observation on cardiac evaluations from these guidelines applies to preoperative consults in general:
The purpose of preoperative evaluation is not to give medical clearance but rather to perform an evaluation of the patient’s current medical status; make recommendations concerning the evaluation, management, and risk of cardiac problems over the entire perioperative period; and provide a clinical risk profile that [can be used] in making treatment decisions that may influence short- and long-term cardiac outcomes.2
These guidelines contain a good description of the ideal preoperative evaluation and consult in a short section (Section II, “General Approach to the Patient”)2 that is worthy of wide dissemination.
DISCUSSION
Question from the audience: Many consults are written more for the surgical team than for the anesthesiologists, hence advice such as managing intraoperative diabetes. Isn’t that appropriate?
Dr. Lubarsky: There are a variety of users of the information in a consult note. I focused on the anesthesiologist, but certainly the surgical staff and house staff would benefit from suggestions about postoperative management. However, they would not benefit from suggestions on intraoperative management; surgeons simply do not need this information and the anesthesiologist will have his or her own regimen. But if there is a specific type of insulin infusion that’s been shown to be best in the specific patient at hand, then detailing that obviously is beneficial.
Question from the audience: We all agree that communication is key, but how does the consultant reach the anesthesiologist to find out what he or she wants to know when the anesthesiologist isn’t usually assigned to the case until a day before surgery?
Dr. Lubarsky: If no anesthesiologist is yet assigned to a case, the consultant can discuss the case with the chief of the anesthesiology department. The discussion should be documented in the note. But it’s important that the system be changed so that anesthesiologists are assigned to cases well in advance. I instituted such a policy at my previous hospital. Many hospitals schedule surgeries 3 months in advance, and many anesthesiology departments have schedules made at least 1 month and often 2 to 3 months in advance. The department could assign a specific anesthesiologist to a future scheduled case with ease.
Question from the audience: How do anesthesiologists educate all the various people we rely on for consults when we can’t get them in one place at one time?
Dr. Lubarsky: It’s a challenge. I try many things, such as going to cardiology rounds, but there are always new people coming through. A good monograph or a set of guidelines with examples would help. If each specialty educates the other and speaks at each other’s conferences more often, that should help. Anesthesiologists would benefit from hearing about the challenges medical consultants face; we may not be doing all we can to optimize perioperative care. There’s room for improvement through communication on both sides. I should also emphasize that we’re all trying to do the right thing. Doctors try to be accommodating, but that doesn’t always make for good decisions. Recently a consultant in my hospital did a preoperative stress test on a patient who didn’t need one. When I asked why, he said, “Because the surgeon asked me to.”
Question from the audience: But don’t you agree that many anesthesiologists would like to see that negative stress test, even if a stress test is not indicated by the guidelines? Cardiologists know that the anesthesiologists are often looking for that on the morning of surgery.
Dr. Lubarsky: The point is that physicians should be responsible for what they have expertise in. When I am asked to intubate a patient, my response as an expert in intubation might be, “Actually, he doesn’t need to be intubated right now.” In the case of this unnecessary stress test, the cardiologist probably should have called the surgeon and said, “It’s really not indicated because the patient had a negative stress test 2 years ago, there’s been no change in symptoms and no angina since then, and he operates well above 4 metabolic equivalents. There’s a clear-cut reason not to do it.” If the surgeon still wanted the test done just to be reassured, that’s simply a poor use of society’s resources. We depend on experts to identify the tests that are indicated to evaluate a patient’s disease and not just do tests for the sake of doing them.
The ideal preoperative medical consultation (consult) is useful to the whole surgical team, ensures maximal patient readiness for surgery, and promotes optimal perioperative care of the patient. Too often, however, consults are ignored or, even worse, set the stage for legal problems. This article identifies problems frequently seen in preoperative consults, particularly from the perspective of anesthesiologists, and gives guidance to those who write consults—hospitalists, internists, cardiologists, and other medical consultants—on providing the information that is most needed by those who use them.
A WIDE RANGE OF END USERS
Anesthesiologists are most often the primary users of the information in preoperative consults, but many other members of the surgical and perioperative team benefit from a well-developed consult, including surgeons, intensivists, nurses, and pain management specialists. Most important, patients stand to benefit, as a good consult helps to ensure that the full breadth of relevant patient-specific information is brought to bear to anticipate potential difficulties and promote optimal care.
Purpose of a consult is in the eye of the beholder
The literature on medical consults in the perioperative arena is scant. The only fairly recent assessment of physician attitudes toward the role of consults was reported by Katz et al in 1998.1 These researchers surveyed attitudes about the various perceived purposes of preoperative cardiology consults, and received rather different responses from anesthesiologists, cardiologists, and surgeons.
There was consensus among all three specialties that two particular functions of a consult are important:
- Treating an inadequately managed cardiac condition before surgery
- Providing data to use in anesthetic management.
Additionally, all three specialties deemed the suggestion of intraoperative treatment modalities to be reasonably important when such suggestions were specifically included in the consult request, although anesthesiologists assigned less importance to this function.1
In contrast, anesthesiologists considered suggestions about intraoperative treatment generally unimportant when not specifically requested, and they viewed suggestions of intraoperative monitoring and advice on the safest type of anesthesia as even less important. Anesthesiologists also deemed “clearing the patient for surgery” as an unimportant function of the consult. Cardiologists rated all of these functions as more important than anesthesiologists did and in some cases as considerably more important. To many of the survey questions, surgeons responded that a specific purpose of a consult was “neither important nor unimportant.”1 That may be because the surgeon’s purpose in obtaining the consult is often simply to address the concerns of the anesthesiologist, who might otherwise delay or cancel a needed surgery.
Consult deficiencies: Vagueness, illegibility, dictating anesthetic choice
The survey by Katz et al also assessed each specialty’s perceptions of the most common deficiencies of preoperative cardiology consults. The deficiencies deemed most common were failure to give specific facts, illegible handwriting, and attempts to dictate the type of anesthesia to be used. Anesthesiologists considered each of the deficiencies assessed as occurring more commonly than their cardiologist or surgeon colleagues did.1
The requester–user disconnect
The differing perceptions of preoperative consults by anesthesiologists and surgeons underscore a fundamental problem: the primary requesters of consults (surgeons) are different from the primary users of consults (anesthesiologists).
Ideally, preoperative consults should be requested by anesthesiologists. Unless and until the ordering of consults changes on a wide scale, however, our advice is for consultants to ask the anesthesiologist what he or she needs to know, in addition to any questions directed to the requesting surgeon. Communication between the surgeon and anesthesiologist should be encouraged as much as possible, and consultants should keep both the anesthesiologist and surgeon in mind when writing consult notes.
A final end user: The plaintiff’s attorney
It is wise to keep in mind one more potential user of your consult: a plaintiff’s attorney. A poorly written consult may benefit plaintiffs’ lawyers. Consults should never give absolute instructions; it is better to use such phrases as “Strongly consider…” or “The current literature strongly suggests…” Otherwise, the surgical team is placed in an awkward position if it does not follow your recommendations, even if for good reason. If a certain recommendation absolutely must be followed, then direct oral communication from the consultant to the attending anesthesiologist (or surgeon) is best.
CONSIDER THE PRIMARY USER: WHAT ANESTHESIOLOGISTS ALREADY KNOW
For the purpose of preoperative consults, it is helpful to think of anesthesiologists as experts in acute medical care. Their 4-year training consists of the following:
- 1 year of internship, often in medicine, including 6 months of basic patient care in the ward or clinic (the last time they will manage chronic disease)
- 4 months in the intensive care unit (ICU) and 1 month in the recovery room, which yields solid intensivist training
- 3 months in pain management, covering acute and chronic pain and regional blocks
- ~24 months in the operating room, often devoted to care of complex problems in surgical subspecialties (obstetric, pediatric, neurologic, cardiothoracic, vascular)
- 1 month of preoperative screening and consultations (a recent requirement).
An optional fifth year may be spent in a subspecialty.
Since the large part of anesthesiologists’ training is in acute care, they generally do not need advice about the acute treatment of any ailment. Consults should not advise anesthesiologists on subjects in which they have considerable expertise. They already have well-established ideas about addressing hypertension, myocardial ischemia, heart failure, arrhythmias (unless unusual therapies are needed), bronchospasm, glucose levels, and pain in the operating room, so they are apt to ignore advice on such topics.
There are several additional topics in which anesthesiologists have considerable expertise and do not need guidance in consults:
- Choice of anesthetic type and its impact on outcome
- Choice of invasive or noninvasive monitoring for any comorbidity and operation
- Postoperative patient disposition (ie, whether to send a patient home, to the postanesthesia care unit, to the ICU, or to a step-down unit)
- Impact of optimizing organ function on perioperative outcome
- Cardiovascular and respiratory physiology
- Pharmacology of intravenous agents.
WHAT ANESTHESIOLOGISTS MAY NOT KNOW―AND NEED FROM CONSULTANTS
How to manage chronic diseases
Follow-up care (eg, for poorly controlled diabetes or hypertension) often can wait until after the operation, and a consultant’s opinion about that is appreciated. It is especially helpful to know that the patient will be followed without the surgeon or anesthesiologist having to arrange for it.
New evidence-based guidance from the literature
One case when recommendations on acute medical management should be provided is when they involve new information from the literature—ie, important data or guidelines published within the prior 5 years or so. It can take time for new information and recommendations to reach all clinicians even within a single specialty. Moreover, important information, such as on the perioperative use of beta-blockers and statins, is not necessarily published in the anesthesiology literature. It is critical to relay information such as the recent recommendation not to withdraw statins prior to surgery, as the current editions of most anesthesiology textbooks have incorrect information suggesting discontinuation. Thus, consultants should include pertinent recent data and guideline recommendations, especially if they go against previous dogma.
Rare diseases, blood disorders, other special cases
As outlined in Table 1, advice on perioperative management is appreciated for patients with rare diseases, coagulation disorders or other blood disorders, and brittle diabetes and other endocrine disorders, as most anesthesiologists are not intimately familiar with these conditions. Anesthesiologists also need, but often do not get, basic details on coronary stents and other implanted devices (see Table 1), as well as guidance on the latest anticoagulation recommendations, with which it is difficult to keep up to date.
A sensitivity to audience and context
It is always appropriate to ask the surgeon requesting a consult—and the anesthesiologist assigned to the case, if known—what he or she wants to know from the consult. If guidance on specific cases is impractical, it is appropriate to ask the chair of the anesthesiology department, or several anesthesiologists collectively, for general guidance on what they look for from preoperative consults.
Anesthesiologists, like consultants, comprise a broad range of people, and it is always important to be sensitive to context. Generalists who work mainly on healthy patients or in a community setting may have forgotten some of their training in acute medicine and are more likely to appreciate advice on intraoperative care. On the other hand, an anesthesiologist who trained in a cardiothoracic subspecialty fellowship, who routinely deals with issues such as left ventricular assist devices and heart transplants, would not benefit from such advice.
WHAT A CONSULT SHOULD―AND SHOULD NOT―BE
The above advice can be distilled into a few principles:
- A consult is an opportunity for the medical consultant to provide helpful management suggestions to the operative team.
- A fundamental objective of a consult is to optimize a patient’s underlying disease before it is compounded by the insult of surgery.
- The purpose of a consult is never to “clear” a patient for surgery. Whether or not to proceed to surgery is a question for the anesthesiologist, surgeon, and patient to decide after weighing the risks and benefits once the patient’s comorbidities are optimally managed. The consult is an important contributing factor to this decision, but it should never be the mechanism of the decision. Though the note from the surgeon requesting a consult may routinely be written as, “Clear the patient for surgery,” consultants should recognize this for what it is—the surgeon’s attempt to avoid having the anesthesiologist cancel the operation—and refrain from weighing in on “clearance” one way or the other.
CASE STUDY: CARDIAC CONSULT REQUESTED BEFORE FEM-POP BYPASS SURGERY
Cardiovascular problems are the most common reasons for requesting preoperative consults. The following case illustrates a typical scenario for a cardiac consult request and presents examples of good and bad notes requesting consults and good and bad consults written in response.
The case
A 47-year-old man is scheduled for femoral-popliteal bypass surgery. His medical history is significant for diabetes, a myocardial infarction (MI) 3 years ago followed by placement of a stent, and a limited ability to assess exercise tolerance. Evidence of an anteroseptal MI is present on 12-lead electrocardiography. His medications include metoprolol 25 mg twice daily and an oral hypoglycemic agent. His blood pressure is 152/89 mm Hg, heart rate 81 beats per minute, respiratory rate 14 breaths per minute, and arterial oxygen saturation 96% on room air.
The consult request: Bad and good examples
A bad consult request in this case would be, “Clear the patient for surgery.” Although this type of request is routinely written, it is routinely useless.
For this complex surgery with significant fluid requirements, a much better consult request would include several specific requests and questions and might read as follows:
—Please evaluate patient’s post-MI therapy for his CAD. Is further therapy required to optimize CAD treatment?
—Do his blood pressure or diabetic regimens need modification? If so, can this be done postoperatively?
—Please evaluate patient’s myocardial function in light of a lack of info on exercise tolerance. Is an echo indicated?
—Are other tests, therapies, or interventions warranted pre- or postoperatively?
Example of a bad consult
A poorly written consult in a case like this may:
- Include a brief history repeating facts that are already known and noting that “the patient is at his baseline without obvious ischemia.”
- State that the patient is cleared for “spinal” anesthesia. “Clearing a patient for anesthesia” is useless to begin with, but clearing for a certain type of anesthesia places the anesthesiologist in a terrible medicolegal position if general anesthesia turns out to be needed. Moreover, there are no proven major outcome differences related to the type of anesthetic chosen.
- Recommend that “a pulmonary artery catheter might be indicated to monitor hemodynamics.” Besides the fact that such catheters probably do more harm than good, such a recommendation is unnecessary since the anesthesiologist is already expert in managing perioperative care.
- Recommend that “the anesthesia team should monitor the patient carefully in the perioperative arena for hypoxia and hypotension.” Qualitative advice, such as “avoid hypoxia, hypotension, and tachycardia,” is not useful, but quantitative information, such as “during ischemic stress testing, the patient exhibited ischemia when his heart rate went to 142,” can be very helpful.
- State that the patient be sent to the ICU following surgery. Mandating an ICU stay in advance makes no sense unless the operation itself demands ICU care, which is the call of the surgeon and anesthesiologist anyway.
A consult like this doesn’t tell the perioperative team anything that it didn’t already know. Such a consult not only is unhelpful but also actually creates more work since much of the advice needs to be “undone” lest a lawyer see the chart and it not be clear why the consultant’s recommendations were not followed.
Example of a good consult
In contrast, a good consult for this case would involve:
- A detailed history examining the potential for silent ischemia associated with the diabetes, as well as the relationship of the hypertension and beta-blocker therapy to episodes of ischemia. The level of ischemia should be clearly categorized. If it cannot be determined, this should be noted; if it can be determined only that the ischemia is not New York Heart Association class III or IV, note this as well (the perioperative outcomes literature suggests that no preoperative ischemia testing is needed with class I or II angina).
- Guidance on blood pressure optimization in light of the relative urgency of the procedure. Blood pressure need not be normalized preoperatively in this case, but if the operation were totally elective and the consultant felt it could make a difference, it would be appropriate to suggest that blood pressure be optimized beforehand.
- A recommendation on whether and when to change the beta-blocker dosage. If the dosage needs to be increased, the anesthesiologist will want to know how many doses are needed to reach a new steady state. Joint guidelines from the American College of Cardiology (ACC) and American Heart Association (AHA)2 recommend 7 to 30 days, but this time frame is unrealistic in this setting, so more practical advice would be appreciated. A good consult notes any deviation from established guidelines, however, and explains the rationale for such deviation.
- Evaluation of the myocardium at risk. This is especially important with left main disease, as it influences the decision whether to test or intervene versus proceeding with careful beta-blocker titration.
- Evaluation of myocardial function and, if appropriate, a therapy suggestion for optimization.
- Notation of the heart rate or blood pressure thresholds at which ischemia develops if a stress test is performed.
GOOD GUIDANCE FROM THE ACC/AHA GUIDELINES
Our advice here is broadly consistent with the aforementioned 2007 ACC/AHA guidelines on perioperative cardiovascular evaluation for noncardiac surgery.2 The following observation on cardiac evaluations from these guidelines applies to preoperative consults in general:
The purpose of preoperative evaluation is not to give medical clearance but rather to perform an evaluation of the patient’s current medical status; make recommendations concerning the evaluation, management, and risk of cardiac problems over the entire perioperative period; and provide a clinical risk profile that [can be used] in making treatment decisions that may influence short- and long-term cardiac outcomes.2
These guidelines contain a good description of the ideal preoperative evaluation and consult in a short section (Section II, “General Approach to the Patient”)2 that is worthy of wide dissemination.
DISCUSSION
Question from the audience: Many consults are written more for the surgical team than for the anesthesiologists, hence advice such as managing intraoperative diabetes. Isn’t that appropriate?
Dr. Lubarsky: There are a variety of users of the information in a consult note. I focused on the anesthesiologist, but certainly the surgical staff and house staff would benefit from suggestions about postoperative management. However, they would not benefit from suggestions on intraoperative management; surgeons simply do not need this information and the anesthesiologist will have his or her own regimen. But if there is a specific type of insulin infusion that’s been shown to be best in the specific patient at hand, then detailing that obviously is beneficial.
Question from the audience: We all agree that communication is key, but how does the consultant reach the anesthesiologist to find out what he or she wants to know when the anesthesiologist isn’t usually assigned to the case until a day before surgery?
Dr. Lubarsky: If no anesthesiologist is yet assigned to a case, the consultant can discuss the case with the chief of the anesthesiology department. The discussion should be documented in the note. But it’s important that the system be changed so that anesthesiologists are assigned to cases well in advance. I instituted such a policy at my previous hospital. Many hospitals schedule surgeries 3 months in advance, and many anesthesiology departments have schedules made at least 1 month and often 2 to 3 months in advance. The department could assign a specific anesthesiologist to a future scheduled case with ease.
Question from the audience: How do anesthesiologists educate all the various people we rely on for consults when we can’t get them in one place at one time?
Dr. Lubarsky: It’s a challenge. I try many things, such as going to cardiology rounds, but there are always new people coming through. A good monograph or a set of guidelines with examples would help. If each specialty educates the other and speaks at each other’s conferences more often, that should help. Anesthesiologists would benefit from hearing about the challenges medical consultants face; we may not be doing all we can to optimize perioperative care. There’s room for improvement through communication on both sides. I should also emphasize that we’re all trying to do the right thing. Doctors try to be accommodating, but that doesn’t always make for good decisions. Recently a consultant in my hospital did a preoperative stress test on a patient who didn’t need one. When I asked why, he said, “Because the surgeon asked me to.”
Question from the audience: But don’t you agree that many anesthesiologists would like to see that negative stress test, even if a stress test is not indicated by the guidelines? Cardiologists know that the anesthesiologists are often looking for that on the morning of surgery.
Dr. Lubarsky: The point is that physicians should be responsible for what they have expertise in. When I am asked to intubate a patient, my response as an expert in intubation might be, “Actually, he doesn’t need to be intubated right now.” In the case of this unnecessary stress test, the cardiologist probably should have called the surgeon and said, “It’s really not indicated because the patient had a negative stress test 2 years ago, there’s been no change in symptoms and no angina since then, and he operates well above 4 metabolic equivalents. There’s a clear-cut reason not to do it.” If the surgeon still wanted the test done just to be reassured, that’s simply a poor use of society’s resources. We depend on experts to identify the tests that are indicated to evaluate a patient’s disease and not just do tests for the sake of doing them.
- Katz RI, Barnhart JM, Ho G, et al. A survey on the intended purposes and perceived utility of preoperative cardiology consultations. Anesth Analg 1998; 87:830–836.
- Fleisher LA, Beckman JA, Brown KA, et al. ACC/AHA 2007 guidelines on perioperative cardiovascular evaluation and care for noncardiac surgery: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. Circulation 2007; 116:e418–e499.
- Katz RI, Barnhart JM, Ho G, et al. A survey on the intended purposes and perceived utility of preoperative cardiology consultations. Anesth Analg 1998; 87:830–836.
- Fleisher LA, Beckman JA, Brown KA, et al. ACC/AHA 2007 guidelines on perioperative cardiovascular evaluation and care for noncardiac surgery: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. Circulation 2007; 116:e418–e499.
KEY POINTS
- Consults that provide pertinent quantitative data about the patient are helpful—eg, the heart rate at which ischemia was exhibited during stress testing and the degree of ischemia.
- Anesthesiologists do not need assistance with managing intravenous drugs (with the exception of unusual agents), but they can use specific guidance on managing oral medications pre- and postoperatively to best achieve optimization and steady-state concentrations.
- Pertinent recent information (< 5 years old) from the nonanesthesiology literature should be provided.
- Medical consultants should arrange for follow-up care for patients with active conditions not addressed by the surgery.
- Absolute recommendations should be avoided in a consult: the surgical team may have good reason not to follow them, and legal repercussions could ensue. The words “consider” or “strongly consider” usually suffice, except where there is an absolute standard of care.
Perioperative management of warfarin and antiplatelet therapy
Perioperative management of surgical patients who require temporary discontinuation of vitamin K antagonists (warfarin) or antiplatelet drugs is complicated. The risk of a thrombotic event during interruption of anticoagulant or antiplatelet therapy must be weighed against the risk of bleeding when such therapy is used in close proximity to a surgical procedure. This balancing of risks is guided by the patient’s individual risk for thromboembolism or bleeding and underlying conditions such as the presence of a mechanical heart valve or a coronary stent.
High-profile adverse events have made anticoagulant and antiplatelet management one of the most highly litigated aspects of perioperative medicine. Moreover, there is a paucity of randomized clinical trial data and definitive guidelines to address the perioperative needs of patients on antithrombotic therapy. Treatment protocols vary depending on many underlying factors, such as the presence of mechanical heart valves, comorbidities, stent type and location, patient age and medical history, and type of surgical procedure. While recent attention has focused on genetic variations that result in higher or lower sensitivity to warfarin in some patients, routine genetic testing for warfarin sensitivity is controversial and not part of widespread practice at this time.
The first portion of this article explores key issues and principles in the perioperative management of surgical patients on warfarin therapy, and the second portion does the same for surgical patients on antiplatelet therapy.
ACCP RECOMMENDATIONS FOR PERIOPERATIVE ANTICOAGULANT MANAGEMENT
Patients with mechanical valves who are at high risk for perioperative thromboembolism include those with any mechanical mitral valve, an older valve, or a history of stroke or transient ischemic attack (TIA). Patients with atrial fibrillation who are at high risk include those with a recent stroke or TIA, rheumatic valvular heart disease, or a CHADS2 score of 5 or 6. (The CHADS2 scoring system assigns one point each for a history of congestive heart failure, hypertension, age greater than 75 years, or diabetes, and two points for history of stroke or TIA.) Patients with a history of VTE within the prior 3 months are also considered high risk.
Bridging anticoagulation (bridge therapy)—ie, the temporary use of intravenous unfractionated heparin (IV UFH) or low-molecular-weight heparin (LMWH) prior to surgery—is central to the ACCP’s recommendations for perioperative management in patients on long-term anticoagulant therapy. Key ACCP recommendations1 for these patients, according to their risk for thromboembolism (Table 1), are as follows:
- High risk—bridging anticoagulation with therapeutic-dose subcutaneous LMWH or IV UFH
- Moderate risk—bridging anticoagulation with therapeutic-dose subcutaneous LMWH, therapeutic-dose IV UFH, or low-dose subcutaneous LMWH
- Low risk—bridging anticoagulation with low-dose subcutaneous LMWH or no bridging.
ASSESSING RISKS: DETERMINING WHETHER TO BRIDGE
Patient-specific risk factors
Patient risk factors include the indication for anticoagulation, as detailed above, as well as other individual risks for thromboembolism, as discussed in the article by Michota on preventing VTE on page S45 of this supplement.
If anticoagulation is indicated because the patient has a mechanical heart valve, the valve type and position must be considered because these factors affect thromboembolic risk, as reflected in Table 1. For instance, the risk of thromboembolism is greater when the valve is in the mitral position than in the aortic position, and is also greater with an older caged-ball valve than with a newer-generation bileaflet valve.3
Procedure-related risk factors
Surgical risks factors include the type of surgery and its associated risks of bleeding and thromboembolism, as well as the expected time that anticoagulation will be interrupted. Estimating thromboembolic risk is complicated, however, and reliable results are generally not achieved with simplistic calculations or formulas. Such calculations tend not to appropriately account for the hypercoagulable state induced by surgery itself, as the risk of VTE is estimated to be 100 times greater during the perioperative period than in the nonoperative setting, owing to increased levels of plasminogen activator inhibitor-1. Moreover, multiple studies have demonstrated increases in coagulation factors that suggest that a “rebound hypercoagulability” may occur shortly after discontinuation of oral anticoagulant therapy.5–8
Net benefit vs risk in trials of bridge therapy
In an analysis of data from observational studies, Kearon and Hirsh estimated the relative risk reduction for thromboembolism with bridge therapy to be 66% to 80%, depending on the indication for anticoagulation.8 Thus, if a patient’s risk of developing thromboembolism is 1.5%, bridge therapy reduces the risk to 0.5% or less.
Weigh relative consequences of an event with the patient
Determining whether and how to initiate bridge therapy ultimately depends on the consequences of an event. Recurrent VTE is fatal in 5% to 10% of cases,15 and arterial thromboembolism is fatal in 20% of cases and causes permanent disability in at least 50% of cases.16 While 9% to 13% of major bleeding events are fatal, bleeding rarely causes permanent disability.17 Thus, whereas a patient who bleeds can be resuscitated, a patient who develops a thromboembolism may be permanently disabled. These considerations should be shared with the patient, and patient preference should factor into the management strategy. Though the risk of bleeding with anticoagulation may be much higher than the risk of stroke without it, many patients will be more concerned about stroke risk.
CHOICE OF AGENT FOR BRIDGE THERAPY
LMWH appears to offer cost advantage over UFH
For cost reasons, managed care organizations often recommend LMWH, which can be administered subcutaneously in outpatient settings, over IV UFH administered in the hospital. A retrospective analysis of medical costs from the 1990s in a managed care organization found that bridge therapy with LMWH prior to elective surgery cost an average of $13,114 less per patient (in total cost of care) than did bridge therapy with UFH.18
LMWH safety issues in valve patients are a myth
Clinical outcomes were not statistically significantly different for patients receiving LMWH or UFH in the above study.18 Nevertheless, there is a widely held notion that LMWH is not safe to use as bridge therapy for patients with mechanical heart valves. Recent prospective bridge studies do not support that view, demonstrating that LMWH used as bridge therapy is associated with low risks for thromboembolism and major bleeding even in patients with mechanical valves.9,10,12–14 In contrast, recent data on the use of IV UFH for bridging is minimal, with most bridge studies dating to the 1970s. Accordingly, the latest ACCP guidelines for perioperative management of patients on antithrombotic therapy recommend therapeutic-dose LMWH over IV UFH for bridge therapy, including in patients with mechanical heart valves.1 Likewise, 2006 guidelines from the American College of Cardiology and American Heart Association on management of patients with valvular heart disease endorse LMWH as an option for bridge therapy.19
A PRACTICAL APPROACH TO BRIDGE THERAPY
A bridge therapy protocol for patients receiving warfarin has been successfully used at the Cleveland Clinic, where I previously practiced. Essentials of the protocol20 are summarized here, followed by commentary that draws on additional sources.
Before surgery
- Discontinue warfarin 5 days before surgery (ie, hold four doses) if the preoperative international normalized ratio (INR) is 2 to 3, and 6 days before surgery (hold five doses) if the INR is 3 to 4.5.
- For bridge therapy, start LMWH (enoxaparin 1 mg/kg or dalteparin 100 IU/kg subcutaneously every 12 hours) beginning 36 hours after the last dose of warfarin.
- Give the last dose of LMWH approximately 24 hours prior to surgery.
After surgery
- For minor surgery, reinitiate LMWH at full dose approximately 24 hours after surgery. For major surgery and for patients at high risk of bleeding, consider using prophylactic doses on the first two postoperative days.
- Discuss the timing of anticoagulant reinitiation with the surgeon.
- Restart warfarin at preoperative dose 1 day after surgery.
- Order daily prothrombin time/INR tests until the patient is discharged and periodically after discharge until the INR is within the therapeutic range.
- Order a complete blood cell count with platelets on days 3 and 7.
- Discontinue LMWH when the INR is between 2 and 3 for 2 consecutive days.
Additionally, the plan should be discussed in advance with the patient, surgeon, and anesthesiologist, along with the risks and benefits associated with LMWH. The patient should receive written instructions for self-administration and information about signs and symptoms of bleeding and thromboembolism.
When to stop warfarin
Warfarin should be discontinued far enough in advance of surgery to achieve a preoperative target INR of less than 1.2.21 Patients with an initial INR of 2 to 3 tend to achieve that target after discontinuation of warfarin for about 5 days (four doses). A longer wait (6 days, or five doses) is necessary for patients with an initial INR of 3 to 4. Age is associated with a slower rate of decrease in the INR, and there is wide interpatient variation. The INR should always be checked prior to surgery.21
Warfarin need not be stopped for all procedures
If warfarin is stopped for minor procedures, bridging may be counterproductive
At the same time, a recent prospective observational study evaluated the effects of brief (≤ 5 days) interruption of warfarin among more than 1,000 patients undergoing minor outpatient procedures and found low rates of both thromboembolism (0.7%) and major bleeding (0.6%).27 The risk of major bleeding was significantly higher among the small proportion of patients who received bridge therapy with UFH or LMWH. The study concluded that interrupting warfarin for 5 days or less for minor outpatient procedures carries a low risk of thromboembolism and that the risk of clinically significant bleeding should be weighed before bridge therapy is considered in this setting.
When to stop bridge therapy preoperatively
Bridge therapy with LMWH is commonly discontinued 12 hours before surgery, but it is preferable to discontinue 24 hours before surgery. In a study of preoperative anticoagulant activity in 80 patients, LMWH (enoxaparin 1 mg/kg) was administered twice daily and discontinued the night before surgery.28 Blood anti–factor Xa levels were measured shortly before surgery, at which time 68% of patients still had therapeutic levels of anti–Xa. This suggests that discontinuing LMWH too close to the time of surgery can increase the risk of bleeding.
Consistent with these findings, consensus guidelines from the American Society of Regional Anesthesia and Pain Medicine (ASRA) recommend that needle placement for regional anesthesia take place 12 hours after the last dose of LMWH if prophylactic dosing is used and 24 hours after the last dose of LMWH if therapeutic dosing is used (ie, ≥ 1 mg/kg of enoxaparin every 12 hours).29
Dosing and timing of postoperative bridge therapy
Postoperative use of full-dose bridge therapy is associated with increased risks of bleeding, according to a multicenter study of approximately 500 patients who received various doses of UFH or LMWH for bridge therapy.14 Patients who received full-dose LMWH or UFH after surgery had a fivefold to sixfold increase in the incidence of major bleeding compared with patients who received prophylactic doses. The study centers that frequently used full-dose bridge protocols were four times as likely to report major bleeding events. In light of these findings, waiting a couple of days after surgery to initiate full-dose bridge therapy is recommended, and prophylactic dosing may be considered in the interim.
The ASRA consensus guidelines recommend that indwelling catheters be removed prior to postoperative reinitiation of twice-daily dosing of LMWH. The first dose of LMWH should be given no sooner than 2 hours after catheter removal. Once-daily dosing of LMWH (European dosing) is acceptable under the ASRA guidelines, but the first dose should be given 6 to 8 hours after surgery and the second dose no sooner than 24 hours later. The guidelines state that once-daily (but not twice-daily) LMWH dosing is acceptable in patients with indwelling catheters; neurological status should be monitored in these patients, and the catheter should be removed 12 to 24 hours after the last dose of LMWH.29
PERIOPERATIVE MANAGEMENT OF ANTIPLATELET THERAPY: TYPE OF AGENT MATTERS
Unlike the considerations with warfarin, the timing of preoperative discontinuation of antiplatelet therapy in patients undergoing noncardiac surgery depends on the type of agent used and its pharmacokinetic actions. Commonly used antiplatelet drugs include aspirin, the thienopyridine agent clopidogrel, and nonsteroidal anti-inflammatory drugs (NSAIDs).
Aspirin works by irreversibly inhibiting platelet cyclooxygenase. The circulating platelet pool is replaced every 7 to 10 days, so aspirin therapy should be discontinued 7 to 10 days before surgery.1
NSAIDs reversibly inhibit platelet cyclooxygenase. Knowing whether a patient is using short- or long-acting NSAIDs is important for determining when to discontinue therapy. For a short-acting NSAID such as ibuprofen, discontinuation 24 hours before surgery may be adequate to normalize platelet function.1,30
Thienopyridines inhibit adenosine diphosphate receptor–mediated platelet activation and aggregation. Short-acting thienopyridines may be discontinued 24 hours before surgery, but long-acting agents such as clopidogrel should be stopped 7 days prior to surgery (including when used with aspirin as dual antiplatelet therapy),1 although some outcomes data suggest that 5 days may be sufficient.31
All of these agents should be resumed as soon as adequate hemostasis is achieved after surgery. The ACCP guidelines on perioperative management of antithrombotic therapy recommend resumption of aspirin at the usual maintenance dose the day after surgery, but they make no specific recommendations on when to resume other antiplatelet drugs.1
ANTIPLATELET THERAPY: SPECIAL CONSIDERATIONS IN PATIENTS WITH STENTS
Patients who are on antiplatelet therapy because they have a coronary stent merit special consideration due to the high risk of thrombosis if therapy is interrupted. The risk of stent thrombosis is especially elevated in the postoperative period, particularly if surgery follows soon after stent placement.
- Relative risks and benefits of stopping versus continuing antiplatelet therapy
- Identification of patients at high risk for a perioperative event after cessation of antiplatelet therapy
- Identification of patients at high risk of bleeding.
Bleeding vs stent thrombosis: Consider relative consequences
The risk of bleeding varies by individual patient. No laboratory tests are available to determine individual bleeding risk, but the risk of perioperative bleeding increases when two or more antiplatelet agents are used, as in dual antiplatelet therapy with aspirin and clopidogrel.31
When balancing risks of bleeding versus thrombotic events, the relative consequences of each event again must be considered. Bleeding is rarely life-threatening in comparison with the potential consequences of stent thrombosis. In a prospective observational study of 2,229 patients who received drug-eluting stents, 29 (1.3%) developed stent thrombosis during 9-month follow-up.32 Among these patients, 20 (69%) had a nonfatal myocardial infarction and 13 (45%) died. The most significant independent risk factor for stent thrombosis was premature discontinuation of antiplatelet therapy (hazard ratio = 89.78 [95% CI, 29.90–260.60]; P < .001). Other independent risk factors included renal failure, bifurcation lesions, diabetes, and low ejection fraction.
Premature interruption of antiplatelet therapy: Why it matters
Abrupt discontinuation of antiplatelet therapy can lead to a rebound effect marked by an inflammatory prothrombotic state, increased platelet adhesion and aggregation, and excessive thromboxane A2 activity. Surgery further increases the prothrombotic and inflammatory state, which, combined with incompletely endothelialized drug-eluting stents, can lead to stent thrombosis and, consequently, myocardial infarction and/or death.33
Timing of surgery after stenting: Getting it right
The US Food and Drug Administration recommends that dual antiplatelet therapy be continued for at least 3 months after placement of a sirolimus-eluting stent and at least 6 months after placement of a paclitaxel-eluting stent. Recent data suggest, however, that this duration of antiplatelet therapy may not be sufficient and that at least 1 year of therapy may be needed.34
A recent joint science advisory from the American College of Cardiology (ACC) and the American Heart Association (AHA) emphasizes the importance of educating providers about the “potentially catastrophic” risks of premature stopping of thienopyridine therapy in patients with coronary stents.34 In addition to recommendations in this joint advisory, the ACC and AHA issued updated guidelines in 2007 on perioperative cardiovascular evaluation and care for noncardiac surgery.35 Below is a summary of recommendations on the timing of surgery following stenting in light of these and other sources:
- Following placement of a bare metal stent, elective and nonurgent procedures should be delayed for at least 1 month, according to the ACC/AHA joint advisory,34 or at least 6 weeks, according to the ACC/AHA guidelines.35 Newer data suggest that the optimal interval for delay is likely to be 3 months.36,37
- For patients with recent (< 6 weeks) bare metal stent placement who require urgent surgery, dual antiplatelet therapy should be continued during the perioperative period.1
- Following placement of a drug-eluting stent, elective and nonurgent procedures should be delayed for at least 12 months.34,35
- For patients with recent drug-eluting stent placement in whom surgery cannot be delayed, dual antiplatelet therapy should be continued without interruption if the stent was placed within the prior 6 months.1,35 If the stent was placed more than 6 months before urgent surgery, aspirin should be continued without interruption (at ≥ 81 mg/day) and clopidogrel should be continued until 5 days before surgery and resumed as soon as possible after surgery (at a loading dose of 300 mg followed by 75 mg/day). If the surgeon is comfortable continuing dual antiplatelet therapy in a patient whose stent was placed 6 to 12 months earlier, that course should be considered.1
It is important to note that the ACC/AHA joint advisory34 and other documents have medicolegal implications, so delaying nonurgent surgery for the periods recommended is the most prudent approach.
CONCLUSIONS
Perioperative management of anticoagulant and antiplatelet therapy is complicated by the paucity of randomized clinical trial data and the risk for serious adverse events. The underlying indications for anticoagulant and antiplatelet therapy vary widely, so the best approach to perioperative management is to involve all members of the health care team—hospitalist, surgeon, cardiologist, and anesthesiologist, together with the patient—to ensure that care is individualized and all relevant considerations are accounted for. Patient and surgical risks can be identified and quantified to some extent, but patients often have greater concerns about the risk of stroke than the risk of bleeding. Ideally, nonemergency surgeries should be scheduled to allow enough time to thoroughly plan the management protocol, reducing risks for bleeding and thrombotic events as much as possible.
DISCUSSION
Question from the audience: If a patient’s INR is 1.3 or 1.4, rather than the recommended 1.2, is it necessary to cancel a planned epidural?
Dr. Jaffer: It depends on how comfortable the surgeon or anesthesiologist is with the INR level. Generally, an INR less than 1.5 is probably acceptable, but it depends on the procedure. For a craniotomy, for example, 1.2 is recommended.
Question from the audience: Is it necessary to use anti–Xa levels to guide bridge therapy when administering LMWH or UFH in a patient with a mechanical heart valve?
Dr. Jaffer: It’s not generally necessary, except for pregnant women. For most patients, doses are calculated as milligrams of LMWH per kilogram body weight or as International Units of LMWH per kilogram.
Question from the audience: You mentioned medicolegal disputes arising from adverse events associated with bridge therapy, drug discontinuation, or related issues. Who has final responsibility for making decisions about discontinuation of antiplatelet therapy, for example?
Dr. Jaffer: I don’t know if it ultimately comes down to just one person. Several physicians should be involved in the decision, and communication protocols within an institution should be very clear. It’s important to make certain everyone involved in the decision is reviewing the same literature. The final decision has to be something everyone involved can accept and support.
- Douketis JD, Berger PB, Dunn AS, et al. The perioperative management of antithrombotic therapy: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest 2008; 133(suppl 6):299S–339S.
- du Breuil AL, Umland EM. Outpatient management of anticoagulation therapy. Am Fam Physician 2007; 75:1031–1042.
- Cannegieter SC, Rosendaal FR, Briët E. Thromboembolic and bleeding complications in patients with mechanical heart valve prostheses. Circulation 1994; 89:635–641.
- Snow V, Weiss KB, LeFevre M, et al. Management of newly detected atrial fibrillation: a clinical practice guideline from the American Academy of Family Physicians and the American College of Physicians. Ann Intern Med 2003; 139:1009–1017.
- Genewein U, Haeberli A, Straub PW, Beer JH. Rebound after cessation of oral anticoagulant therapy: the biochemical evidence. Br J Haematol 1996; 92:479–485.
- Grip L, Blombäck M, Schulman S. Hypercoagulable state and thromboembolism following warfarin withdrawal in post-myocardial-infarction patients. Eur Heart J 1991; 12:1225–1233.
- Palareti G, Legnani C, Guazzaloca G, et al. Activation of blood coagulation after abrupt or stepwise withdrawal of oral anticoagulants: a prospective study. Thromb Haemost 1994; 72:222–226.
- Kearon C, Hirsh J. Management of anticoagulation before and after elective surgery. N Engl J Med 1997; 336:1506–1511.
- Douketis JD, Johnson JA, Turpie AG. Low-molecular-weight heparin as bridging anticoagulation during interruption of warfarin: assessment of a standardized periprocedural anticoagulation regimen. Arch Intern Med 2004; 164:1319–1326.
- Kovacs MJ, Kearon C, Rodger M, et al. Single-arm study of bridging therapy with low-molecular-weight heparin for patients at risk of arterial embolism who require temporary interruption of warfarin. Circulation 2004; 110:1658–1663.
- Dunn AS, Spyropoulos AC, Turpie AG. Bridging therapy in patients on long-term oral anticoagulants who require surgery: the Prospective Peri-operative Enoxaparin Cohort Trial (PROSPECT). J Thromb Haemost 2007; 5:2211–2218.
- Spyropoulos AC, Turpie AG, Dunn AS, et al. Clinical outcomes with unfractionated heparin or low-molecular-weight heparin as bridging therapy in patients on long-term oral anticoagulants: the REGIMEN registry. J Thromb Haemost 2006; 4:1246–1252.
- Turpie AG, Douketis JD. Enoxaparin is effective and safe as bridging anticoagulation in patients with a mechanical prosthetic heart valve who require temporary interruption of warfarin because of surgery or an invasive procedure. Blood 2004:202s. Abstract 703.
- Jaffer AK, Brotman DJ, White RH. The perioperative and periprocedural management of warfarin: the PPMW study. J Thromb Haemost 2005; 3(suppl 1). Abstract OR309.
- Linkins LA, Choi PT, Douketis JD. Clinical impact of bleeding in patients taking oral anticoagulant therapy for venous thromboembolism: a meta-analysis. Ann Intern Med 2003; 139:893–900.
- Schulman S, Rhedin AS, Lindmarker P, et al. A comparison of six weeks with six months of oral anticoagulant therapy after a first episode of venous thromboembolism: duration of Anticoagulation Trial Study Group. N Engl J Med 1995; 332:1661–1665.
- Atrial Fibrillation Investigators. Risk factors for stroke and efficacy of antithrombotic therapy in atrial fibrillation: analysis of pooled data from five randomized controlled trials. Arch Intern Med 1994; 154:1449–1457.
- Spyropoulos AC, Frost FJ, Hurley JS, Roberts M. Costs and clinical outcomes associated with low-molecular-weight heparin vs unfractionated heparin for perioperative bridging in patients receiving long-term oral anticoagulant therapy. Chest 2004; 125:1642–1650.
- American College of Cardiology/American Heart Association Task Force on Practice Guidelines. ACC/AHA 2006 guidelines for the management of patients with valvular heart disease. Circulation 2006; 114:e84–e231.
- Jaffer AK, Brotman DJ, Chukwumerjie N. When patients on warfarin need surgery. Cleve Clin J Med 2003; 70:973–984.
- White RH, McKittrick T, Hutchinson R, Twitchell J. Temporary discontinuation of warfarin therapy: changes in the international normalized ratio. Ann Intern Med 1995; 122:40–42.
- Konstantatos A. Anticoagulation and cataract surgery: a review of the current literature. Anaesth Intensive Care 2001; 29:11–18.
- Weibert RT. Oral anticoagulant therapy in patients undergoing dental surgery. Clin Pharm 1992; 11:857–864.
- Billingsley EM, Maloney ME. Intraoperative and postoperative bleeding problems in patients taking warfarin, aspirin, and nonsteroidal antiinflammatory agents: a prospective study. Dermatol Surg 1997; 23:381–383.
- Kadakia SC, Angueira CE, Ward JA, Moore M. Gastrointestinal endoscopy in patients taking antiplatelet agents and anticoagulants: survey of ASGE members: American Society for Gastrointestinal Endoscopy. Gastrointest Endosc 1996; 44:309–316.
- Dunn AS, Turpie AG. Perioperative management of patients receiving oral anticoagulants: a systematic review. Arch Intern Med 2003; 163:901–908.
- Garcia DA, Regan S, Henault LE, et al. Risk of thromboembolism with short-term interruption of warfarin therapy. Arch Intern Med 2008; 168:63–69.
- O’Donnell MJ, Kearon C, Johnson J, et al. Brief communication: preoperative anticoagulant activity after bridging low-molecular-weight heparin for temporary interruption of warfarin. Ann Intern Med 2007; 146:184–187.
- Horlocker TT, Wedel DJ, Benzon H, et al. Regional anesthesia in the anticoagulated patient: defining the risks (the second ASRA Consensus Conference on Neuraxial Anesthesia and Anticoagulation). Reg Anesth Pain Med 2003; 28:172–197.
- Goldenberg NA, Jacobson L, Manco-Johnson MJ. Brief communication: duration of platelet dysfunction after a 7-day course of ibuprofen. Ann Intern Med 2005; 142:506–509.
- Lecompte T, Hardy JF. Antiplatelet agents and perioperative bleeding. Can J Anaesth 2006; 53(suppl 6):S103–S112.
- Iakovou I, Schmidt T, Bonizzoni E, et al. Incidence, predictors, and outcome of thrombosis after successful implantation of drug-eluting stents. JAMA 2005; 293:2126–2130.
- Newsome LT, Weller RS, Gerancher JC, Kutcher MA, Royster RL. Coronary artery stents: II. Perioperative considerations and management. Anesth Analg 2008; 107:570–590.
- Grines CL, Bonow RO, Casey DE Jr, et al. Prevention of premature discontinuation of dual antiplatelet therapy in patients with coronary artery stents: a science advisory from the American Heart Association, American College of Cardiology, Society for Cardiovascular Angiography and Interventions, American College of Surgeons, and American Dental Association, with representation from the American College of Physicians. Circulation 2007; 115:813–818.
- Fleisher LA, Beckman JA, Brown KA, et al. ACC/AHA 2007 guidelines on perioperative cardiovascular evaluation and care for noncardiac surgery: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines [published correction appears in J Am Coll Cardiol 2008; 52:794–797]. J Am Coll Cardiol 2007; 50:1707–1732.
- Brichon PY, Boitet P, Dujon A, et al. Perioperative in-stent thrombosis after lung resection performed within 3 months of coronary stenting. Eur J Cardiothorac Surg 2006; 30:793–796.
- Nuttall GA, Brown MJ, Stombaugh JW, et al. Time and cardiac risk of surgery after bare-metal stent percutaneous coronary intervention. Anesthesiology 2008; 109:588–595.
Perioperative management of surgical patients who require temporary discontinuation of vitamin K antagonists (warfarin) or antiplatelet drugs is complicated. The risk of a thrombotic event during interruption of anticoagulant or antiplatelet therapy must be weighed against the risk of bleeding when such therapy is used in close proximity to a surgical procedure. This balancing of risks is guided by the patient’s individual risk for thromboembolism or bleeding and underlying conditions such as the presence of a mechanical heart valve or a coronary stent.
High-profile adverse events have made anticoagulant and antiplatelet management one of the most highly litigated aspects of perioperative medicine. Moreover, there is a paucity of randomized clinical trial data and definitive guidelines to address the perioperative needs of patients on antithrombotic therapy. Treatment protocols vary depending on many underlying factors, such as the presence of mechanical heart valves, comorbidities, stent type and location, patient age and medical history, and type of surgical procedure. While recent attention has focused on genetic variations that result in higher or lower sensitivity to warfarin in some patients, routine genetic testing for warfarin sensitivity is controversial and not part of widespread practice at this time.
The first portion of this article explores key issues and principles in the perioperative management of surgical patients on warfarin therapy, and the second portion does the same for surgical patients on antiplatelet therapy.
ACCP RECOMMENDATIONS FOR PERIOPERATIVE ANTICOAGULANT MANAGEMENT
Patients with mechanical valves who are at high risk for perioperative thromboembolism include those with any mechanical mitral valve, an older valve, or a history of stroke or transient ischemic attack (TIA). Patients with atrial fibrillation who are at high risk include those with a recent stroke or TIA, rheumatic valvular heart disease, or a CHADS2 score of 5 or 6. (The CHADS2 scoring system assigns one point each for a history of congestive heart failure, hypertension, age greater than 75 years, or diabetes, and two points for history of stroke or TIA.) Patients with a history of VTE within the prior 3 months are also considered high risk.
Bridging anticoagulation (bridge therapy)—ie, the temporary use of intravenous unfractionated heparin (IV UFH) or low-molecular-weight heparin (LMWH) prior to surgery—is central to the ACCP’s recommendations for perioperative management in patients on long-term anticoagulant therapy. Key ACCP recommendations1 for these patients, according to their risk for thromboembolism (Table 1), are as follows:
- High risk—bridging anticoagulation with therapeutic-dose subcutaneous LMWH or IV UFH
- Moderate risk—bridging anticoagulation with therapeutic-dose subcutaneous LMWH, therapeutic-dose IV UFH, or low-dose subcutaneous LMWH
- Low risk—bridging anticoagulation with low-dose subcutaneous LMWH or no bridging.
ASSESSING RISKS: DETERMINING WHETHER TO BRIDGE
Patient-specific risk factors
Patient risk factors include the indication for anticoagulation, as detailed above, as well as other individual risks for thromboembolism, as discussed in the article by Michota on preventing VTE on page S45 of this supplement.
If anticoagulation is indicated because the patient has a mechanical heart valve, the valve type and position must be considered because these factors affect thromboembolic risk, as reflected in Table 1. For instance, the risk of thromboembolism is greater when the valve is in the mitral position than in the aortic position, and is also greater with an older caged-ball valve than with a newer-generation bileaflet valve.3
Procedure-related risk factors
Surgical risks factors include the type of surgery and its associated risks of bleeding and thromboembolism, as well as the expected time that anticoagulation will be interrupted. Estimating thromboembolic risk is complicated, however, and reliable results are generally not achieved with simplistic calculations or formulas. Such calculations tend not to appropriately account for the hypercoagulable state induced by surgery itself, as the risk of VTE is estimated to be 100 times greater during the perioperative period than in the nonoperative setting, owing to increased levels of plasminogen activator inhibitor-1. Moreover, multiple studies have demonstrated increases in coagulation factors that suggest that a “rebound hypercoagulability” may occur shortly after discontinuation of oral anticoagulant therapy.5–8
Net benefit vs risk in trials of bridge therapy
In an analysis of data from observational studies, Kearon and Hirsh estimated the relative risk reduction for thromboembolism with bridge therapy to be 66% to 80%, depending on the indication for anticoagulation.8 Thus, if a patient’s risk of developing thromboembolism is 1.5%, bridge therapy reduces the risk to 0.5% or less.
Weigh relative consequences of an event with the patient
Determining whether and how to initiate bridge therapy ultimately depends on the consequences of an event. Recurrent VTE is fatal in 5% to 10% of cases,15 and arterial thromboembolism is fatal in 20% of cases and causes permanent disability in at least 50% of cases.16 While 9% to 13% of major bleeding events are fatal, bleeding rarely causes permanent disability.17 Thus, whereas a patient who bleeds can be resuscitated, a patient who develops a thromboembolism may be permanently disabled. These considerations should be shared with the patient, and patient preference should factor into the management strategy. Though the risk of bleeding with anticoagulation may be much higher than the risk of stroke without it, many patients will be more concerned about stroke risk.
CHOICE OF AGENT FOR BRIDGE THERAPY
LMWH appears to offer cost advantage over UFH
For cost reasons, managed care organizations often recommend LMWH, which can be administered subcutaneously in outpatient settings, over IV UFH administered in the hospital. A retrospective analysis of medical costs from the 1990s in a managed care organization found that bridge therapy with LMWH prior to elective surgery cost an average of $13,114 less per patient (in total cost of care) than did bridge therapy with UFH.18
LMWH safety issues in valve patients are a myth
Clinical outcomes were not statistically significantly different for patients receiving LMWH or UFH in the above study.18 Nevertheless, there is a widely held notion that LMWH is not safe to use as bridge therapy for patients with mechanical heart valves. Recent prospective bridge studies do not support that view, demonstrating that LMWH used as bridge therapy is associated with low risks for thromboembolism and major bleeding even in patients with mechanical valves.9,10,12–14 In contrast, recent data on the use of IV UFH for bridging is minimal, with most bridge studies dating to the 1970s. Accordingly, the latest ACCP guidelines for perioperative management of patients on antithrombotic therapy recommend therapeutic-dose LMWH over IV UFH for bridge therapy, including in patients with mechanical heart valves.1 Likewise, 2006 guidelines from the American College of Cardiology and American Heart Association on management of patients with valvular heart disease endorse LMWH as an option for bridge therapy.19
A PRACTICAL APPROACH TO BRIDGE THERAPY
A bridge therapy protocol for patients receiving warfarin has been successfully used at the Cleveland Clinic, where I previously practiced. Essentials of the protocol20 are summarized here, followed by commentary that draws on additional sources.
Before surgery
- Discontinue warfarin 5 days before surgery (ie, hold four doses) if the preoperative international normalized ratio (INR) is 2 to 3, and 6 days before surgery (hold five doses) if the INR is 3 to 4.5.
- For bridge therapy, start LMWH (enoxaparin 1 mg/kg or dalteparin 100 IU/kg subcutaneously every 12 hours) beginning 36 hours after the last dose of warfarin.
- Give the last dose of LMWH approximately 24 hours prior to surgery.
After surgery
- For minor surgery, reinitiate LMWH at full dose approximately 24 hours after surgery. For major surgery and for patients at high risk of bleeding, consider using prophylactic doses on the first two postoperative days.
- Discuss the timing of anticoagulant reinitiation with the surgeon.
- Restart warfarin at preoperative dose 1 day after surgery.
- Order daily prothrombin time/INR tests until the patient is discharged and periodically after discharge until the INR is within the therapeutic range.
- Order a complete blood cell count with platelets on days 3 and 7.
- Discontinue LMWH when the INR is between 2 and 3 for 2 consecutive days.
Additionally, the plan should be discussed in advance with the patient, surgeon, and anesthesiologist, along with the risks and benefits associated with LMWH. The patient should receive written instructions for self-administration and information about signs and symptoms of bleeding and thromboembolism.
When to stop warfarin
Warfarin should be discontinued far enough in advance of surgery to achieve a preoperative target INR of less than 1.2.21 Patients with an initial INR of 2 to 3 tend to achieve that target after discontinuation of warfarin for about 5 days (four doses). A longer wait (6 days, or five doses) is necessary for patients with an initial INR of 3 to 4. Age is associated with a slower rate of decrease in the INR, and there is wide interpatient variation. The INR should always be checked prior to surgery.21
Warfarin need not be stopped for all procedures
If warfarin is stopped for minor procedures, bridging may be counterproductive
At the same time, a recent prospective observational study evaluated the effects of brief (≤ 5 days) interruption of warfarin among more than 1,000 patients undergoing minor outpatient procedures and found low rates of both thromboembolism (0.7%) and major bleeding (0.6%).27 The risk of major bleeding was significantly higher among the small proportion of patients who received bridge therapy with UFH or LMWH. The study concluded that interrupting warfarin for 5 days or less for minor outpatient procedures carries a low risk of thromboembolism and that the risk of clinically significant bleeding should be weighed before bridge therapy is considered in this setting.
When to stop bridge therapy preoperatively
Bridge therapy with LMWH is commonly discontinued 12 hours before surgery, but it is preferable to discontinue 24 hours before surgery. In a study of preoperative anticoagulant activity in 80 patients, LMWH (enoxaparin 1 mg/kg) was administered twice daily and discontinued the night before surgery.28 Blood anti–factor Xa levels were measured shortly before surgery, at which time 68% of patients still had therapeutic levels of anti–Xa. This suggests that discontinuing LMWH too close to the time of surgery can increase the risk of bleeding.
Consistent with these findings, consensus guidelines from the American Society of Regional Anesthesia and Pain Medicine (ASRA) recommend that needle placement for regional anesthesia take place 12 hours after the last dose of LMWH if prophylactic dosing is used and 24 hours after the last dose of LMWH if therapeutic dosing is used (ie, ≥ 1 mg/kg of enoxaparin every 12 hours).29
Dosing and timing of postoperative bridge therapy
Postoperative use of full-dose bridge therapy is associated with increased risks of bleeding, according to a multicenter study of approximately 500 patients who received various doses of UFH or LMWH for bridge therapy.14 Patients who received full-dose LMWH or UFH after surgery had a fivefold to sixfold increase in the incidence of major bleeding compared with patients who received prophylactic doses. The study centers that frequently used full-dose bridge protocols were four times as likely to report major bleeding events. In light of these findings, waiting a couple of days after surgery to initiate full-dose bridge therapy is recommended, and prophylactic dosing may be considered in the interim.
The ASRA consensus guidelines recommend that indwelling catheters be removed prior to postoperative reinitiation of twice-daily dosing of LMWH. The first dose of LMWH should be given no sooner than 2 hours after catheter removal. Once-daily dosing of LMWH (European dosing) is acceptable under the ASRA guidelines, but the first dose should be given 6 to 8 hours after surgery and the second dose no sooner than 24 hours later. The guidelines state that once-daily (but not twice-daily) LMWH dosing is acceptable in patients with indwelling catheters; neurological status should be monitored in these patients, and the catheter should be removed 12 to 24 hours after the last dose of LMWH.29
PERIOPERATIVE MANAGEMENT OF ANTIPLATELET THERAPY: TYPE OF AGENT MATTERS
Unlike the considerations with warfarin, the timing of preoperative discontinuation of antiplatelet therapy in patients undergoing noncardiac surgery depends on the type of agent used and its pharmacokinetic actions. Commonly used antiplatelet drugs include aspirin, the thienopyridine agent clopidogrel, and nonsteroidal anti-inflammatory drugs (NSAIDs).
Aspirin works by irreversibly inhibiting platelet cyclooxygenase. The circulating platelet pool is replaced every 7 to 10 days, so aspirin therapy should be discontinued 7 to 10 days before surgery.1
NSAIDs reversibly inhibit platelet cyclooxygenase. Knowing whether a patient is using short- or long-acting NSAIDs is important for determining when to discontinue therapy. For a short-acting NSAID such as ibuprofen, discontinuation 24 hours before surgery may be adequate to normalize platelet function.1,30
Thienopyridines inhibit adenosine diphosphate receptor–mediated platelet activation and aggregation. Short-acting thienopyridines may be discontinued 24 hours before surgery, but long-acting agents such as clopidogrel should be stopped 7 days prior to surgery (including when used with aspirin as dual antiplatelet therapy),1 although some outcomes data suggest that 5 days may be sufficient.31
All of these agents should be resumed as soon as adequate hemostasis is achieved after surgery. The ACCP guidelines on perioperative management of antithrombotic therapy recommend resumption of aspirin at the usual maintenance dose the day after surgery, but they make no specific recommendations on when to resume other antiplatelet drugs.1
ANTIPLATELET THERAPY: SPECIAL CONSIDERATIONS IN PATIENTS WITH STENTS
Patients who are on antiplatelet therapy because they have a coronary stent merit special consideration due to the high risk of thrombosis if therapy is interrupted. The risk of stent thrombosis is especially elevated in the postoperative period, particularly if surgery follows soon after stent placement.
- Relative risks and benefits of stopping versus continuing antiplatelet therapy
- Identification of patients at high risk for a perioperative event after cessation of antiplatelet therapy
- Identification of patients at high risk of bleeding.
Bleeding vs stent thrombosis: Consider relative consequences
The risk of bleeding varies by individual patient. No laboratory tests are available to determine individual bleeding risk, but the risk of perioperative bleeding increases when two or more antiplatelet agents are used, as in dual antiplatelet therapy with aspirin and clopidogrel.31
When balancing risks of bleeding versus thrombotic events, the relative consequences of each event again must be considered. Bleeding is rarely life-threatening in comparison with the potential consequences of stent thrombosis. In a prospective observational study of 2,229 patients who received drug-eluting stents, 29 (1.3%) developed stent thrombosis during 9-month follow-up.32 Among these patients, 20 (69%) had a nonfatal myocardial infarction and 13 (45%) died. The most significant independent risk factor for stent thrombosis was premature discontinuation of antiplatelet therapy (hazard ratio = 89.78 [95% CI, 29.90–260.60]; P < .001). Other independent risk factors included renal failure, bifurcation lesions, diabetes, and low ejection fraction.
Premature interruption of antiplatelet therapy: Why it matters
Abrupt discontinuation of antiplatelet therapy can lead to a rebound effect marked by an inflammatory prothrombotic state, increased platelet adhesion and aggregation, and excessive thromboxane A2 activity. Surgery further increases the prothrombotic and inflammatory state, which, combined with incompletely endothelialized drug-eluting stents, can lead to stent thrombosis and, consequently, myocardial infarction and/or death.33
Timing of surgery after stenting: Getting it right
The US Food and Drug Administration recommends that dual antiplatelet therapy be continued for at least 3 months after placement of a sirolimus-eluting stent and at least 6 months after placement of a paclitaxel-eluting stent. Recent data suggest, however, that this duration of antiplatelet therapy may not be sufficient and that at least 1 year of therapy may be needed.34
A recent joint science advisory from the American College of Cardiology (ACC) and the American Heart Association (AHA) emphasizes the importance of educating providers about the “potentially catastrophic” risks of premature stopping of thienopyridine therapy in patients with coronary stents.34 In addition to recommendations in this joint advisory, the ACC and AHA issued updated guidelines in 2007 on perioperative cardiovascular evaluation and care for noncardiac surgery.35 Below is a summary of recommendations on the timing of surgery following stenting in light of these and other sources:
- Following placement of a bare metal stent, elective and nonurgent procedures should be delayed for at least 1 month, according to the ACC/AHA joint advisory,34 or at least 6 weeks, according to the ACC/AHA guidelines.35 Newer data suggest that the optimal interval for delay is likely to be 3 months.36,37
- For patients with recent (< 6 weeks) bare metal stent placement who require urgent surgery, dual antiplatelet therapy should be continued during the perioperative period.1
- Following placement of a drug-eluting stent, elective and nonurgent procedures should be delayed for at least 12 months.34,35
- For patients with recent drug-eluting stent placement in whom surgery cannot be delayed, dual antiplatelet therapy should be continued without interruption if the stent was placed within the prior 6 months.1,35 If the stent was placed more than 6 months before urgent surgery, aspirin should be continued without interruption (at ≥ 81 mg/day) and clopidogrel should be continued until 5 days before surgery and resumed as soon as possible after surgery (at a loading dose of 300 mg followed by 75 mg/day). If the surgeon is comfortable continuing dual antiplatelet therapy in a patient whose stent was placed 6 to 12 months earlier, that course should be considered.1
It is important to note that the ACC/AHA joint advisory34 and other documents have medicolegal implications, so delaying nonurgent surgery for the periods recommended is the most prudent approach.
CONCLUSIONS
Perioperative management of anticoagulant and antiplatelet therapy is complicated by the paucity of randomized clinical trial data and the risk for serious adverse events. The underlying indications for anticoagulant and antiplatelet therapy vary widely, so the best approach to perioperative management is to involve all members of the health care team—hospitalist, surgeon, cardiologist, and anesthesiologist, together with the patient—to ensure that care is individualized and all relevant considerations are accounted for. Patient and surgical risks can be identified and quantified to some extent, but patients often have greater concerns about the risk of stroke than the risk of bleeding. Ideally, nonemergency surgeries should be scheduled to allow enough time to thoroughly plan the management protocol, reducing risks for bleeding and thrombotic events as much as possible.
DISCUSSION
Question from the audience: If a patient’s INR is 1.3 or 1.4, rather than the recommended 1.2, is it necessary to cancel a planned epidural?
Dr. Jaffer: It depends on how comfortable the surgeon or anesthesiologist is with the INR level. Generally, an INR less than 1.5 is probably acceptable, but it depends on the procedure. For a craniotomy, for example, 1.2 is recommended.
Question from the audience: Is it necessary to use anti–Xa levels to guide bridge therapy when administering LMWH or UFH in a patient with a mechanical heart valve?
Dr. Jaffer: It’s not generally necessary, except for pregnant women. For most patients, doses are calculated as milligrams of LMWH per kilogram body weight or as International Units of LMWH per kilogram.
Question from the audience: You mentioned medicolegal disputes arising from adverse events associated with bridge therapy, drug discontinuation, or related issues. Who has final responsibility for making decisions about discontinuation of antiplatelet therapy, for example?
Dr. Jaffer: I don’t know if it ultimately comes down to just one person. Several physicians should be involved in the decision, and communication protocols within an institution should be very clear. It’s important to make certain everyone involved in the decision is reviewing the same literature. The final decision has to be something everyone involved can accept and support.
Perioperative management of surgical patients who require temporary discontinuation of vitamin K antagonists (warfarin) or antiplatelet drugs is complicated. The risk of a thrombotic event during interruption of anticoagulant or antiplatelet therapy must be weighed against the risk of bleeding when such therapy is used in close proximity to a surgical procedure. This balancing of risks is guided by the patient’s individual risk for thromboembolism or bleeding and underlying conditions such as the presence of a mechanical heart valve or a coronary stent.
High-profile adverse events have made anticoagulant and antiplatelet management one of the most highly litigated aspects of perioperative medicine. Moreover, there is a paucity of randomized clinical trial data and definitive guidelines to address the perioperative needs of patients on antithrombotic therapy. Treatment protocols vary depending on many underlying factors, such as the presence of mechanical heart valves, comorbidities, stent type and location, patient age and medical history, and type of surgical procedure. While recent attention has focused on genetic variations that result in higher or lower sensitivity to warfarin in some patients, routine genetic testing for warfarin sensitivity is controversial and not part of widespread practice at this time.
The first portion of this article explores key issues and principles in the perioperative management of surgical patients on warfarin therapy, and the second portion does the same for surgical patients on antiplatelet therapy.
ACCP RECOMMENDATIONS FOR PERIOPERATIVE ANTICOAGULANT MANAGEMENT
Patients with mechanical valves who are at high risk for perioperative thromboembolism include those with any mechanical mitral valve, an older valve, or a history of stroke or transient ischemic attack (TIA). Patients with atrial fibrillation who are at high risk include those with a recent stroke or TIA, rheumatic valvular heart disease, or a CHADS2 score of 5 or 6. (The CHADS2 scoring system assigns one point each for a history of congestive heart failure, hypertension, age greater than 75 years, or diabetes, and two points for history of stroke or TIA.) Patients with a history of VTE within the prior 3 months are also considered high risk.
Bridging anticoagulation (bridge therapy)—ie, the temporary use of intravenous unfractionated heparin (IV UFH) or low-molecular-weight heparin (LMWH) prior to surgery—is central to the ACCP’s recommendations for perioperative management in patients on long-term anticoagulant therapy. Key ACCP recommendations1 for these patients, according to their risk for thromboembolism (Table 1), are as follows:
- High risk—bridging anticoagulation with therapeutic-dose subcutaneous LMWH or IV UFH
- Moderate risk—bridging anticoagulation with therapeutic-dose subcutaneous LMWH, therapeutic-dose IV UFH, or low-dose subcutaneous LMWH
- Low risk—bridging anticoagulation with low-dose subcutaneous LMWH or no bridging.
ASSESSING RISKS: DETERMINING WHETHER TO BRIDGE
Patient-specific risk factors
Patient risk factors include the indication for anticoagulation, as detailed above, as well as other individual risks for thromboembolism, as discussed in the article by Michota on preventing VTE on page S45 of this supplement.
If anticoagulation is indicated because the patient has a mechanical heart valve, the valve type and position must be considered because these factors affect thromboembolic risk, as reflected in Table 1. For instance, the risk of thromboembolism is greater when the valve is in the mitral position than in the aortic position, and is also greater with an older caged-ball valve than with a newer-generation bileaflet valve.3
Procedure-related risk factors
Surgical risks factors include the type of surgery and its associated risks of bleeding and thromboembolism, as well as the expected time that anticoagulation will be interrupted. Estimating thromboembolic risk is complicated, however, and reliable results are generally not achieved with simplistic calculations or formulas. Such calculations tend not to appropriately account for the hypercoagulable state induced by surgery itself, as the risk of VTE is estimated to be 100 times greater during the perioperative period than in the nonoperative setting, owing to increased levels of plasminogen activator inhibitor-1. Moreover, multiple studies have demonstrated increases in coagulation factors that suggest that a “rebound hypercoagulability” may occur shortly after discontinuation of oral anticoagulant therapy.5–8
Net benefit vs risk in trials of bridge therapy
In an analysis of data from observational studies, Kearon and Hirsh estimated the relative risk reduction for thromboembolism with bridge therapy to be 66% to 80%, depending on the indication for anticoagulation.8 Thus, if a patient’s risk of developing thromboembolism is 1.5%, bridge therapy reduces the risk to 0.5% or less.
Weigh relative consequences of an event with the patient
Determining whether and how to initiate bridge therapy ultimately depends on the consequences of an event. Recurrent VTE is fatal in 5% to 10% of cases,15 and arterial thromboembolism is fatal in 20% of cases and causes permanent disability in at least 50% of cases.16 While 9% to 13% of major bleeding events are fatal, bleeding rarely causes permanent disability.17 Thus, whereas a patient who bleeds can be resuscitated, a patient who develops a thromboembolism may be permanently disabled. These considerations should be shared with the patient, and patient preference should factor into the management strategy. Though the risk of bleeding with anticoagulation may be much higher than the risk of stroke without it, many patients will be more concerned about stroke risk.
CHOICE OF AGENT FOR BRIDGE THERAPY
LMWH appears to offer cost advantage over UFH
For cost reasons, managed care organizations often recommend LMWH, which can be administered subcutaneously in outpatient settings, over IV UFH administered in the hospital. A retrospective analysis of medical costs from the 1990s in a managed care organization found that bridge therapy with LMWH prior to elective surgery cost an average of $13,114 less per patient (in total cost of care) than did bridge therapy with UFH.18
LMWH safety issues in valve patients are a myth
Clinical outcomes were not statistically significantly different for patients receiving LMWH or UFH in the above study.18 Nevertheless, there is a widely held notion that LMWH is not safe to use as bridge therapy for patients with mechanical heart valves. Recent prospective bridge studies do not support that view, demonstrating that LMWH used as bridge therapy is associated with low risks for thromboembolism and major bleeding even in patients with mechanical valves.9,10,12–14 In contrast, recent data on the use of IV UFH for bridging is minimal, with most bridge studies dating to the 1970s. Accordingly, the latest ACCP guidelines for perioperative management of patients on antithrombotic therapy recommend therapeutic-dose LMWH over IV UFH for bridge therapy, including in patients with mechanical heart valves.1 Likewise, 2006 guidelines from the American College of Cardiology and American Heart Association on management of patients with valvular heart disease endorse LMWH as an option for bridge therapy.19
A PRACTICAL APPROACH TO BRIDGE THERAPY
A bridge therapy protocol for patients receiving warfarin has been successfully used at the Cleveland Clinic, where I previously practiced. Essentials of the protocol20 are summarized here, followed by commentary that draws on additional sources.
Before surgery
- Discontinue warfarin 5 days before surgery (ie, hold four doses) if the preoperative international normalized ratio (INR) is 2 to 3, and 6 days before surgery (hold five doses) if the INR is 3 to 4.5.
- For bridge therapy, start LMWH (enoxaparin 1 mg/kg or dalteparin 100 IU/kg subcutaneously every 12 hours) beginning 36 hours after the last dose of warfarin.
- Give the last dose of LMWH approximately 24 hours prior to surgery.
After surgery
- For minor surgery, reinitiate LMWH at full dose approximately 24 hours after surgery. For major surgery and for patients at high risk of bleeding, consider using prophylactic doses on the first two postoperative days.
- Discuss the timing of anticoagulant reinitiation with the surgeon.
- Restart warfarin at preoperative dose 1 day after surgery.
- Order daily prothrombin time/INR tests until the patient is discharged and periodically after discharge until the INR is within the therapeutic range.
- Order a complete blood cell count with platelets on days 3 and 7.
- Discontinue LMWH when the INR is between 2 and 3 for 2 consecutive days.
Additionally, the plan should be discussed in advance with the patient, surgeon, and anesthesiologist, along with the risks and benefits associated with LMWH. The patient should receive written instructions for self-administration and information about signs and symptoms of bleeding and thromboembolism.
When to stop warfarin
Warfarin should be discontinued far enough in advance of surgery to achieve a preoperative target INR of less than 1.2.21 Patients with an initial INR of 2 to 3 tend to achieve that target after discontinuation of warfarin for about 5 days (four doses). A longer wait (6 days, or five doses) is necessary for patients with an initial INR of 3 to 4. Age is associated with a slower rate of decrease in the INR, and there is wide interpatient variation. The INR should always be checked prior to surgery.21
Warfarin need not be stopped for all procedures
If warfarin is stopped for minor procedures, bridging may be counterproductive
At the same time, a recent prospective observational study evaluated the effects of brief (≤ 5 days) interruption of warfarin among more than 1,000 patients undergoing minor outpatient procedures and found low rates of both thromboembolism (0.7%) and major bleeding (0.6%).27 The risk of major bleeding was significantly higher among the small proportion of patients who received bridge therapy with UFH or LMWH. The study concluded that interrupting warfarin for 5 days or less for minor outpatient procedures carries a low risk of thromboembolism and that the risk of clinically significant bleeding should be weighed before bridge therapy is considered in this setting.
When to stop bridge therapy preoperatively
Bridge therapy with LMWH is commonly discontinued 12 hours before surgery, but it is preferable to discontinue 24 hours before surgery. In a study of preoperative anticoagulant activity in 80 patients, LMWH (enoxaparin 1 mg/kg) was administered twice daily and discontinued the night before surgery.28 Blood anti–factor Xa levels were measured shortly before surgery, at which time 68% of patients still had therapeutic levels of anti–Xa. This suggests that discontinuing LMWH too close to the time of surgery can increase the risk of bleeding.
Consistent with these findings, consensus guidelines from the American Society of Regional Anesthesia and Pain Medicine (ASRA) recommend that needle placement for regional anesthesia take place 12 hours after the last dose of LMWH if prophylactic dosing is used and 24 hours after the last dose of LMWH if therapeutic dosing is used (ie, ≥ 1 mg/kg of enoxaparin every 12 hours).29
Dosing and timing of postoperative bridge therapy
Postoperative use of full-dose bridge therapy is associated with increased risks of bleeding, according to a multicenter study of approximately 500 patients who received various doses of UFH or LMWH for bridge therapy.14 Patients who received full-dose LMWH or UFH after surgery had a fivefold to sixfold increase in the incidence of major bleeding compared with patients who received prophylactic doses. The study centers that frequently used full-dose bridge protocols were four times as likely to report major bleeding events. In light of these findings, waiting a couple of days after surgery to initiate full-dose bridge therapy is recommended, and prophylactic dosing may be considered in the interim.
The ASRA consensus guidelines recommend that indwelling catheters be removed prior to postoperative reinitiation of twice-daily dosing of LMWH. The first dose of LMWH should be given no sooner than 2 hours after catheter removal. Once-daily dosing of LMWH (European dosing) is acceptable under the ASRA guidelines, but the first dose should be given 6 to 8 hours after surgery and the second dose no sooner than 24 hours later. The guidelines state that once-daily (but not twice-daily) LMWH dosing is acceptable in patients with indwelling catheters; neurological status should be monitored in these patients, and the catheter should be removed 12 to 24 hours after the last dose of LMWH.29
PERIOPERATIVE MANAGEMENT OF ANTIPLATELET THERAPY: TYPE OF AGENT MATTERS
Unlike the considerations with warfarin, the timing of preoperative discontinuation of antiplatelet therapy in patients undergoing noncardiac surgery depends on the type of agent used and its pharmacokinetic actions. Commonly used antiplatelet drugs include aspirin, the thienopyridine agent clopidogrel, and nonsteroidal anti-inflammatory drugs (NSAIDs).
Aspirin works by irreversibly inhibiting platelet cyclooxygenase. The circulating platelet pool is replaced every 7 to 10 days, so aspirin therapy should be discontinued 7 to 10 days before surgery.1
NSAIDs reversibly inhibit platelet cyclooxygenase. Knowing whether a patient is using short- or long-acting NSAIDs is important for determining when to discontinue therapy. For a short-acting NSAID such as ibuprofen, discontinuation 24 hours before surgery may be adequate to normalize platelet function.1,30
Thienopyridines inhibit adenosine diphosphate receptor–mediated platelet activation and aggregation. Short-acting thienopyridines may be discontinued 24 hours before surgery, but long-acting agents such as clopidogrel should be stopped 7 days prior to surgery (including when used with aspirin as dual antiplatelet therapy),1 although some outcomes data suggest that 5 days may be sufficient.31
All of these agents should be resumed as soon as adequate hemostasis is achieved after surgery. The ACCP guidelines on perioperative management of antithrombotic therapy recommend resumption of aspirin at the usual maintenance dose the day after surgery, but they make no specific recommendations on when to resume other antiplatelet drugs.1
ANTIPLATELET THERAPY: SPECIAL CONSIDERATIONS IN PATIENTS WITH STENTS
Patients who are on antiplatelet therapy because they have a coronary stent merit special consideration due to the high risk of thrombosis if therapy is interrupted. The risk of stent thrombosis is especially elevated in the postoperative period, particularly if surgery follows soon after stent placement.
- Relative risks and benefits of stopping versus continuing antiplatelet therapy
- Identification of patients at high risk for a perioperative event after cessation of antiplatelet therapy
- Identification of patients at high risk of bleeding.
Bleeding vs stent thrombosis: Consider relative consequences
The risk of bleeding varies by individual patient. No laboratory tests are available to determine individual bleeding risk, but the risk of perioperative bleeding increases when two or more antiplatelet agents are used, as in dual antiplatelet therapy with aspirin and clopidogrel.31
When balancing risks of bleeding versus thrombotic events, the relative consequences of each event again must be considered. Bleeding is rarely life-threatening in comparison with the potential consequences of stent thrombosis. In a prospective observational study of 2,229 patients who received drug-eluting stents, 29 (1.3%) developed stent thrombosis during 9-month follow-up.32 Among these patients, 20 (69%) had a nonfatal myocardial infarction and 13 (45%) died. The most significant independent risk factor for stent thrombosis was premature discontinuation of antiplatelet therapy (hazard ratio = 89.78 [95% CI, 29.90–260.60]; P < .001). Other independent risk factors included renal failure, bifurcation lesions, diabetes, and low ejection fraction.
Premature interruption of antiplatelet therapy: Why it matters
Abrupt discontinuation of antiplatelet therapy can lead to a rebound effect marked by an inflammatory prothrombotic state, increased platelet adhesion and aggregation, and excessive thromboxane A2 activity. Surgery further increases the prothrombotic and inflammatory state, which, combined with incompletely endothelialized drug-eluting stents, can lead to stent thrombosis and, consequently, myocardial infarction and/or death.33
Timing of surgery after stenting: Getting it right
The US Food and Drug Administration recommends that dual antiplatelet therapy be continued for at least 3 months after placement of a sirolimus-eluting stent and at least 6 months after placement of a paclitaxel-eluting stent. Recent data suggest, however, that this duration of antiplatelet therapy may not be sufficient and that at least 1 year of therapy may be needed.34
A recent joint science advisory from the American College of Cardiology (ACC) and the American Heart Association (AHA) emphasizes the importance of educating providers about the “potentially catastrophic” risks of premature stopping of thienopyridine therapy in patients with coronary stents.34 In addition to recommendations in this joint advisory, the ACC and AHA issued updated guidelines in 2007 on perioperative cardiovascular evaluation and care for noncardiac surgery.35 Below is a summary of recommendations on the timing of surgery following stenting in light of these and other sources:
- Following placement of a bare metal stent, elective and nonurgent procedures should be delayed for at least 1 month, according to the ACC/AHA joint advisory,34 or at least 6 weeks, according to the ACC/AHA guidelines.35 Newer data suggest that the optimal interval for delay is likely to be 3 months.36,37
- For patients with recent (< 6 weeks) bare metal stent placement who require urgent surgery, dual antiplatelet therapy should be continued during the perioperative period.1
- Following placement of a drug-eluting stent, elective and nonurgent procedures should be delayed for at least 12 months.34,35
- For patients with recent drug-eluting stent placement in whom surgery cannot be delayed, dual antiplatelet therapy should be continued without interruption if the stent was placed within the prior 6 months.1,35 If the stent was placed more than 6 months before urgent surgery, aspirin should be continued without interruption (at ≥ 81 mg/day) and clopidogrel should be continued until 5 days before surgery and resumed as soon as possible after surgery (at a loading dose of 300 mg followed by 75 mg/day). If the surgeon is comfortable continuing dual antiplatelet therapy in a patient whose stent was placed 6 to 12 months earlier, that course should be considered.1
It is important to note that the ACC/AHA joint advisory34 and other documents have medicolegal implications, so delaying nonurgent surgery for the periods recommended is the most prudent approach.
CONCLUSIONS
Perioperative management of anticoagulant and antiplatelet therapy is complicated by the paucity of randomized clinical trial data and the risk for serious adverse events. The underlying indications for anticoagulant and antiplatelet therapy vary widely, so the best approach to perioperative management is to involve all members of the health care team—hospitalist, surgeon, cardiologist, and anesthesiologist, together with the patient—to ensure that care is individualized and all relevant considerations are accounted for. Patient and surgical risks can be identified and quantified to some extent, but patients often have greater concerns about the risk of stroke than the risk of bleeding. Ideally, nonemergency surgeries should be scheduled to allow enough time to thoroughly plan the management protocol, reducing risks for bleeding and thrombotic events as much as possible.
DISCUSSION
Question from the audience: If a patient’s INR is 1.3 or 1.4, rather than the recommended 1.2, is it necessary to cancel a planned epidural?
Dr. Jaffer: It depends on how comfortable the surgeon or anesthesiologist is with the INR level. Generally, an INR less than 1.5 is probably acceptable, but it depends on the procedure. For a craniotomy, for example, 1.2 is recommended.
Question from the audience: Is it necessary to use anti–Xa levels to guide bridge therapy when administering LMWH or UFH in a patient with a mechanical heart valve?
Dr. Jaffer: It’s not generally necessary, except for pregnant women. For most patients, doses are calculated as milligrams of LMWH per kilogram body weight or as International Units of LMWH per kilogram.
Question from the audience: You mentioned medicolegal disputes arising from adverse events associated with bridge therapy, drug discontinuation, or related issues. Who has final responsibility for making decisions about discontinuation of antiplatelet therapy, for example?
Dr. Jaffer: I don’t know if it ultimately comes down to just one person. Several physicians should be involved in the decision, and communication protocols within an institution should be very clear. It’s important to make certain everyone involved in the decision is reviewing the same literature. The final decision has to be something everyone involved can accept and support.
- Douketis JD, Berger PB, Dunn AS, et al. The perioperative management of antithrombotic therapy: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest 2008; 133(suppl 6):299S–339S.
- du Breuil AL, Umland EM. Outpatient management of anticoagulation therapy. Am Fam Physician 2007; 75:1031–1042.
- Cannegieter SC, Rosendaal FR, Briët E. Thromboembolic and bleeding complications in patients with mechanical heart valve prostheses. Circulation 1994; 89:635–641.
- Snow V, Weiss KB, LeFevre M, et al. Management of newly detected atrial fibrillation: a clinical practice guideline from the American Academy of Family Physicians and the American College of Physicians. Ann Intern Med 2003; 139:1009–1017.
- Genewein U, Haeberli A, Straub PW, Beer JH. Rebound after cessation of oral anticoagulant therapy: the biochemical evidence. Br J Haematol 1996; 92:479–485.
- Grip L, Blombäck M, Schulman S. Hypercoagulable state and thromboembolism following warfarin withdrawal in post-myocardial-infarction patients. Eur Heart J 1991; 12:1225–1233.
- Palareti G, Legnani C, Guazzaloca G, et al. Activation of blood coagulation after abrupt or stepwise withdrawal of oral anticoagulants: a prospective study. Thromb Haemost 1994; 72:222–226.
- Kearon C, Hirsh J. Management of anticoagulation before and after elective surgery. N Engl J Med 1997; 336:1506–1511.
- Douketis JD, Johnson JA, Turpie AG. Low-molecular-weight heparin as bridging anticoagulation during interruption of warfarin: assessment of a standardized periprocedural anticoagulation regimen. Arch Intern Med 2004; 164:1319–1326.
- Kovacs MJ, Kearon C, Rodger M, et al. Single-arm study of bridging therapy with low-molecular-weight heparin for patients at risk of arterial embolism who require temporary interruption of warfarin. Circulation 2004; 110:1658–1663.
- Dunn AS, Spyropoulos AC, Turpie AG. Bridging therapy in patients on long-term oral anticoagulants who require surgery: the Prospective Peri-operative Enoxaparin Cohort Trial (PROSPECT). J Thromb Haemost 2007; 5:2211–2218.
- Spyropoulos AC, Turpie AG, Dunn AS, et al. Clinical outcomes with unfractionated heparin or low-molecular-weight heparin as bridging therapy in patients on long-term oral anticoagulants: the REGIMEN registry. J Thromb Haemost 2006; 4:1246–1252.
- Turpie AG, Douketis JD. Enoxaparin is effective and safe as bridging anticoagulation in patients with a mechanical prosthetic heart valve who require temporary interruption of warfarin because of surgery or an invasive procedure. Blood 2004:202s. Abstract 703.
- Jaffer AK, Brotman DJ, White RH. The perioperative and periprocedural management of warfarin: the PPMW study. J Thromb Haemost 2005; 3(suppl 1). Abstract OR309.
- Linkins LA, Choi PT, Douketis JD. Clinical impact of bleeding in patients taking oral anticoagulant therapy for venous thromboembolism: a meta-analysis. Ann Intern Med 2003; 139:893–900.
- Schulman S, Rhedin AS, Lindmarker P, et al. A comparison of six weeks with six months of oral anticoagulant therapy after a first episode of venous thromboembolism: duration of Anticoagulation Trial Study Group. N Engl J Med 1995; 332:1661–1665.
- Atrial Fibrillation Investigators. Risk factors for stroke and efficacy of antithrombotic therapy in atrial fibrillation: analysis of pooled data from five randomized controlled trials. Arch Intern Med 1994; 154:1449–1457.
- Spyropoulos AC, Frost FJ, Hurley JS, Roberts M. Costs and clinical outcomes associated with low-molecular-weight heparin vs unfractionated heparin for perioperative bridging in patients receiving long-term oral anticoagulant therapy. Chest 2004; 125:1642–1650.
- American College of Cardiology/American Heart Association Task Force on Practice Guidelines. ACC/AHA 2006 guidelines for the management of patients with valvular heart disease. Circulation 2006; 114:e84–e231.
- Jaffer AK, Brotman DJ, Chukwumerjie N. When patients on warfarin need surgery. Cleve Clin J Med 2003; 70:973–984.
- White RH, McKittrick T, Hutchinson R, Twitchell J. Temporary discontinuation of warfarin therapy: changes in the international normalized ratio. Ann Intern Med 1995; 122:40–42.
- Konstantatos A. Anticoagulation and cataract surgery: a review of the current literature. Anaesth Intensive Care 2001; 29:11–18.
- Weibert RT. Oral anticoagulant therapy in patients undergoing dental surgery. Clin Pharm 1992; 11:857–864.
- Billingsley EM, Maloney ME. Intraoperative and postoperative bleeding problems in patients taking warfarin, aspirin, and nonsteroidal antiinflammatory agents: a prospective study. Dermatol Surg 1997; 23:381–383.
- Kadakia SC, Angueira CE, Ward JA, Moore M. Gastrointestinal endoscopy in patients taking antiplatelet agents and anticoagulants: survey of ASGE members: American Society for Gastrointestinal Endoscopy. Gastrointest Endosc 1996; 44:309–316.
- Dunn AS, Turpie AG. Perioperative management of patients receiving oral anticoagulants: a systematic review. Arch Intern Med 2003; 163:901–908.
- Garcia DA, Regan S, Henault LE, et al. Risk of thromboembolism with short-term interruption of warfarin therapy. Arch Intern Med 2008; 168:63–69.
- O’Donnell MJ, Kearon C, Johnson J, et al. Brief communication: preoperative anticoagulant activity after bridging low-molecular-weight heparin for temporary interruption of warfarin. Ann Intern Med 2007; 146:184–187.
- Horlocker TT, Wedel DJ, Benzon H, et al. Regional anesthesia in the anticoagulated patient: defining the risks (the second ASRA Consensus Conference on Neuraxial Anesthesia and Anticoagulation). Reg Anesth Pain Med 2003; 28:172–197.
- Goldenberg NA, Jacobson L, Manco-Johnson MJ. Brief communication: duration of platelet dysfunction after a 7-day course of ibuprofen. Ann Intern Med 2005; 142:506–509.
- Lecompte T, Hardy JF. Antiplatelet agents and perioperative bleeding. Can J Anaesth 2006; 53(suppl 6):S103–S112.
- Iakovou I, Schmidt T, Bonizzoni E, et al. Incidence, predictors, and outcome of thrombosis after successful implantation of drug-eluting stents. JAMA 2005; 293:2126–2130.
- Newsome LT, Weller RS, Gerancher JC, Kutcher MA, Royster RL. Coronary artery stents: II. Perioperative considerations and management. Anesth Analg 2008; 107:570–590.
- Grines CL, Bonow RO, Casey DE Jr, et al. Prevention of premature discontinuation of dual antiplatelet therapy in patients with coronary artery stents: a science advisory from the American Heart Association, American College of Cardiology, Society for Cardiovascular Angiography and Interventions, American College of Surgeons, and American Dental Association, with representation from the American College of Physicians. Circulation 2007; 115:813–818.
- Fleisher LA, Beckman JA, Brown KA, et al. ACC/AHA 2007 guidelines on perioperative cardiovascular evaluation and care for noncardiac surgery: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines [published correction appears in J Am Coll Cardiol 2008; 52:794–797]. J Am Coll Cardiol 2007; 50:1707–1732.
- Brichon PY, Boitet P, Dujon A, et al. Perioperative in-stent thrombosis after lung resection performed within 3 months of coronary stenting. Eur J Cardiothorac Surg 2006; 30:793–796.
- Nuttall GA, Brown MJ, Stombaugh JW, et al. Time and cardiac risk of surgery after bare-metal stent percutaneous coronary intervention. Anesthesiology 2008; 109:588–595.
- Douketis JD, Berger PB, Dunn AS, et al. The perioperative management of antithrombotic therapy: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest 2008; 133(suppl 6):299S–339S.
- du Breuil AL, Umland EM. Outpatient management of anticoagulation therapy. Am Fam Physician 2007; 75:1031–1042.
- Cannegieter SC, Rosendaal FR, Briët E. Thromboembolic and bleeding complications in patients with mechanical heart valve prostheses. Circulation 1994; 89:635–641.
- Snow V, Weiss KB, LeFevre M, et al. Management of newly detected atrial fibrillation: a clinical practice guideline from the American Academy of Family Physicians and the American College of Physicians. Ann Intern Med 2003; 139:1009–1017.
- Genewein U, Haeberli A, Straub PW, Beer JH. Rebound after cessation of oral anticoagulant therapy: the biochemical evidence. Br J Haematol 1996; 92:479–485.
- Grip L, Blombäck M, Schulman S. Hypercoagulable state and thromboembolism following warfarin withdrawal in post-myocardial-infarction patients. Eur Heart J 1991; 12:1225–1233.
- Palareti G, Legnani C, Guazzaloca G, et al. Activation of blood coagulation after abrupt or stepwise withdrawal of oral anticoagulants: a prospective study. Thromb Haemost 1994; 72:222–226.
- Kearon C, Hirsh J. Management of anticoagulation before and after elective surgery. N Engl J Med 1997; 336:1506–1511.
- Douketis JD, Johnson JA, Turpie AG. Low-molecular-weight heparin as bridging anticoagulation during interruption of warfarin: assessment of a standardized periprocedural anticoagulation regimen. Arch Intern Med 2004; 164:1319–1326.
- Kovacs MJ, Kearon C, Rodger M, et al. Single-arm study of bridging therapy with low-molecular-weight heparin for patients at risk of arterial embolism who require temporary interruption of warfarin. Circulation 2004; 110:1658–1663.
- Dunn AS, Spyropoulos AC, Turpie AG. Bridging therapy in patients on long-term oral anticoagulants who require surgery: the Prospective Peri-operative Enoxaparin Cohort Trial (PROSPECT). J Thromb Haemost 2007; 5:2211–2218.
- Spyropoulos AC, Turpie AG, Dunn AS, et al. Clinical outcomes with unfractionated heparin or low-molecular-weight heparin as bridging therapy in patients on long-term oral anticoagulants: the REGIMEN registry. J Thromb Haemost 2006; 4:1246–1252.
- Turpie AG, Douketis JD. Enoxaparin is effective and safe as bridging anticoagulation in patients with a mechanical prosthetic heart valve who require temporary interruption of warfarin because of surgery or an invasive procedure. Blood 2004:202s. Abstract 703.
- Jaffer AK, Brotman DJ, White RH. The perioperative and periprocedural management of warfarin: the PPMW study. J Thromb Haemost 2005; 3(suppl 1). Abstract OR309.
- Linkins LA, Choi PT, Douketis JD. Clinical impact of bleeding in patients taking oral anticoagulant therapy for venous thromboembolism: a meta-analysis. Ann Intern Med 2003; 139:893–900.
- Schulman S, Rhedin AS, Lindmarker P, et al. A comparison of six weeks with six months of oral anticoagulant therapy after a first episode of venous thromboembolism: duration of Anticoagulation Trial Study Group. N Engl J Med 1995; 332:1661–1665.
- Atrial Fibrillation Investigators. Risk factors for stroke and efficacy of antithrombotic therapy in atrial fibrillation: analysis of pooled data from five randomized controlled trials. Arch Intern Med 1994; 154:1449–1457.
- Spyropoulos AC, Frost FJ, Hurley JS, Roberts M. Costs and clinical outcomes associated with low-molecular-weight heparin vs unfractionated heparin for perioperative bridging in patients receiving long-term oral anticoagulant therapy. Chest 2004; 125:1642–1650.
- American College of Cardiology/American Heart Association Task Force on Practice Guidelines. ACC/AHA 2006 guidelines for the management of patients with valvular heart disease. Circulation 2006; 114:e84–e231.
- Jaffer AK, Brotman DJ, Chukwumerjie N. When patients on warfarin need surgery. Cleve Clin J Med 2003; 70:973–984.
- White RH, McKittrick T, Hutchinson R, Twitchell J. Temporary discontinuation of warfarin therapy: changes in the international normalized ratio. Ann Intern Med 1995; 122:40–42.
- Konstantatos A. Anticoagulation and cataract surgery: a review of the current literature. Anaesth Intensive Care 2001; 29:11–18.
- Weibert RT. Oral anticoagulant therapy in patients undergoing dental surgery. Clin Pharm 1992; 11:857–864.
- Billingsley EM, Maloney ME. Intraoperative and postoperative bleeding problems in patients taking warfarin, aspirin, and nonsteroidal antiinflammatory agents: a prospective study. Dermatol Surg 1997; 23:381–383.
- Kadakia SC, Angueira CE, Ward JA, Moore M. Gastrointestinal endoscopy in patients taking antiplatelet agents and anticoagulants: survey of ASGE members: American Society for Gastrointestinal Endoscopy. Gastrointest Endosc 1996; 44:309–316.
- Dunn AS, Turpie AG. Perioperative management of patients receiving oral anticoagulants: a systematic review. Arch Intern Med 2003; 163:901–908.
- Garcia DA, Regan S, Henault LE, et al. Risk of thromboembolism with short-term interruption of warfarin therapy. Arch Intern Med 2008; 168:63–69.
- O’Donnell MJ, Kearon C, Johnson J, et al. Brief communication: preoperative anticoagulant activity after bridging low-molecular-weight heparin for temporary interruption of warfarin. Ann Intern Med 2007; 146:184–187.
- Horlocker TT, Wedel DJ, Benzon H, et al. Regional anesthesia in the anticoagulated patient: defining the risks (the second ASRA Consensus Conference on Neuraxial Anesthesia and Anticoagulation). Reg Anesth Pain Med 2003; 28:172–197.
- Goldenberg NA, Jacobson L, Manco-Johnson MJ. Brief communication: duration of platelet dysfunction after a 7-day course of ibuprofen. Ann Intern Med 2005; 142:506–509.
- Lecompte T, Hardy JF. Antiplatelet agents and perioperative bleeding. Can J Anaesth 2006; 53(suppl 6):S103–S112.
- Iakovou I, Schmidt T, Bonizzoni E, et al. Incidence, predictors, and outcome of thrombosis after successful implantation of drug-eluting stents. JAMA 2005; 293:2126–2130.
- Newsome LT, Weller RS, Gerancher JC, Kutcher MA, Royster RL. Coronary artery stents: II. Perioperative considerations and management. Anesth Analg 2008; 107:570–590.
- Grines CL, Bonow RO, Casey DE Jr, et al. Prevention of premature discontinuation of dual antiplatelet therapy in patients with coronary artery stents: a science advisory from the American Heart Association, American College of Cardiology, Society for Cardiovascular Angiography and Interventions, American College of Surgeons, and American Dental Association, with representation from the American College of Physicians. Circulation 2007; 115:813–818.
- Fleisher LA, Beckman JA, Brown KA, et al. ACC/AHA 2007 guidelines on perioperative cardiovascular evaluation and care for noncardiac surgery: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines [published correction appears in J Am Coll Cardiol 2008; 52:794–797]. J Am Coll Cardiol 2007; 50:1707–1732.
- Brichon PY, Boitet P, Dujon A, et al. Perioperative in-stent thrombosis after lung resection performed within 3 months of coronary stenting. Eur J Cardiothorac Surg 2006; 30:793–796.
- Nuttall GA, Brown MJ, Stombaugh JW, et al. Time and cardiac risk of surgery after bare-metal stent percutaneous coronary intervention. Anesthesiology 2008; 109:588–595.
KEY POINTS
- Determining when and how to use bridge anticoagulation therapy depends on the patient’s risk for thromboembolism, which is in turn based on the indication for warfarin—ie, a mechanical heart valve, atrial fibrillation, or prior venous thromboembolism.
- Factor patient preference into whether and how to use bridge therapy: many patients are more concerned about stroke risk than bleeding risk, regardless of the relative frequency of these events.
- Anticoagulation with warfarin often does not need to be interrupted for patients undergoing minor surgery, such as some ophthalmic, dental, dermatologic, and gastrointestinal procedures.
- Premature discontinuation of antiplatelet therapy in surgical patients with recent coronary stent placement significantly raises the risk of catastrophic perioperative stent thrombosis.
Prevention of venous thromboembolism after surgery
Most surgical patients who require hospitalization should be considered at high risk for venous thromboembolism (VTE) and be given appropriate prophylaxis. For lower-risk procedures such as knee arthroscopy, prophylaxis is needed for those with individual risk factors such as morbid obesity, limited mobility after surgery, or a history of deep vein thrombosis (DVT) or malignancy. Too often, however, prophylaxis is not provided appropriately or not given at all.
This review surveys the essentials of perioperative VTE prophylaxis and important new developments in the field, which include the 2008 release of new evidence-based clinical practice guidelines on antithrombotic and thrombolytic therapy from the American College of Chest Physicians (ACCP). This 8th edition of the guidelines updates the previous edition, published in 2004, and includes a section by Geerts et al devoted to VTE prevention.1 Other major guidelines are also discussed, as are developments in VTE-related quality measurement, management of special patient populations (those with renal impairment or morbid obesity), and emerging therapies for VTE prophylaxis.
IMPETUS FOR QUALITY IMPROVEMENT IN VTE
A new seriousness about VTE quality measures
The 8th edition of the ACCP guidelines recommends that every hospital develop a formal, active strategy to consistently identify medical and surgical patients at risk for VTE and to prevent VTE occurrence.1 Although prior editions of the ACCP guidelines have made this recommendation for more than 2 decades, fewer than 1 in 10 acute care hospitals had any such strategy in place as recently as 5 years ago. Now, however, most US hospitals have implemented such a strategy, thanks to the growing national emphasis on health care quality measurement in recent years.
The Surgical Care Improvement Project (SCIP) has been at the forefront of this recent quality measures movement. SCIP, a joint project of the American Medical Association and federal government agencies, set a goal to reduce surgical complications in the United States by 25% from 2005 to 2010.2 Two SCIP process measures relate to improving VTE prophylaxis2,3:
- The proportion of surgical patients for whom recommended VTE prophylaxis is ordered
- The proportion of surgical patients who actually receive appropriate VTE prophylaxis within 24 hours before or after surgery.
The Joint Commission and the National Quality Forum recently endorsed these two SCIP performance measures for perioperative VTE prophylaxis along with several others relating to VTE treatment.
CMS raises the stakes with reimbursement restrictions
More significantly, the federal government’s Centers for Medicare and Medicaid Services (CMS) will soon refuse to reimburse for hospital treatment of a primary diagnosis of DVT or pulmonary embolism (PE) following recent (within 30 days) total hip or knee arthroplasty. Effective October 1, 2009, a primary VTE diagnosis following these joint replacement procedures will be added to CMS’ current list of “never events,” or hospital-acquired conditions for which CMS will not provide reimbursement because they are considered the result of preventable medical errors. (Notably, treatment of DVT or PE as a secondary diagnosis will still be reimbursed—for example, if a joint replacement patient develops nosocomial pneumonia, is transferred to the intensive care unit, and then develops VTE.) This addition of DVT and PE to the list is highly controversial since these events sometimes develop even if prophylactic therapy is appropriate and aggressive.
Strategies to promote best practices
In the update for the new 8th edition of its guidelines, the ACCP added recommendations on specific ways for hospitals to identify patients at high risk for VTE and ensure that they receive appropriate prophylaxis. These include the use of computer decision-support systems, preprinted orders, and periodic audit and feedback.1
Researchers at Brigham and Women’s Hospital evaluated the effectiveness of a computer alert system for notifying physicians of newly hospitalized patients at risk for DVT who were not receiving prevention therapy within the first 24 hours of hospital admission.4 These patients presumably “fell through the cracks” and warranted prophylaxis but were otherwise not recognized by the health care team. Risk was determined by a scoring system based on multiple variables, including malignancy, previous DVT or PE, hypercoagulability, major surgery, advanced age, obesity, ordered bed rest, and treatment with hormone replacement therapy or oral contraceptives. Study physicians had to acknowledge having received the alert but could choose whether or not to order VTE prophylaxis. Prophylaxis was used in considerably more patients from the intervention group than from a control group of high-risk patients whose physicians did not receive alerts (34% vs 14%, respectively); accordingly, the risk of a symptomatic DVT or PE event at 90 days was reduced by 41% in the intervention group.
Despite this evidence of improved practice under the alert system, the study begs the question of why the percentage of patients at risk for VTE who were given prophylaxis was still so low (34%), demonstrating how much progress in improving practice remains to be achieved.
PROPHYLAXIS STRATEGIES: MATCHING THERAPY TO RISK
NONPHARMACOLOGIC PROPHYLAXIS STRATEGIES
Does ambulation prevent DVT?
Although it is commonly accepted that walking prevents DVT, this has never been directly tested. Walking may simply be a marker of health, and healthy people are less prone to develop thromboses. We have almost no evidence to show that forcing an unhealthy person to walk helps prevent DVT. Early ambulation offers many benefits and should be encouraged, but it should not be considered DVT prophylaxis; it is simply good hospital care.
Mechanical devices: Adherence is key
Amaragiri and Lees conducted a systematic literature review of randomized controlled trials evaluating the effectiveness of graduated compression stockings (elastic stockings) for preventing DVT in various groups of hospitalized patients.6 The analysis demonstrated a statistically significant reduction in DVT incidence with graduated compression stockings compared with control both among the nine trials in which stockings were used alone (odds ratio = 0.34) and among the seven trials in which stockings were used in addition to another method of thromboprophylaxis (odds ratio = 0.24). Although benefit was demonstrated, many of the trials in this review involved patients undergoing gynecologic surgery and date from the 1970s and 1980s (when obesity was less prevalent), so the applicability of their results today may be limited.
The 8th edition of the ACCP guidelines recommends that mechanical methods of VTE prophylaxis be used primarily in patients who are at high risk of bleeding and that careful attention be directed to ensuring their proper use and optimal adherence.1 The latter point about adherence cannot be emphasized enough, as graduated compression stockings and other mechanical devices have been shown not to be effective unless they are worn at least 18 to 20 hours a day. This degree of adherence is difficult to achieve, as it can severely limit patient mobility and leave patients susceptible to development of pressure ulcers.
Mechanical compression should be initiated prior to induction of anesthesia and continue intraoperatively and then into the postanesthesia care unit. Orders for use of mechanical devices should include instructions in the patient’s medical chart specifying how—and for how many hours per day—they are to be worn. Not doing so leaves the physician vulnerable to litigation, especially as the ACCP guidelines include language on optimal adherence to these devices (“they should be removed for only a short time each day when the patient is actually walking or for bathing”1).
Continuous external compression therapy
Newer mechanical device options include a continuous external compression therapy system that allows patients to be mobile while wearing it and provides rhythmic compression that results in good peak venous flows. Ideally such a device could be put on the patient preoperatively and worn during surgery, throughout the hospital stay, and even at home during recovery. Anecdotally, however, I see patients turn these new devices off at the side of the bed just as often as they do with traditional devices.
Vena caval interruption
Vena caval interruption involves placement of a retrievable vena cava filter before surgery and removal some time later; it offers the potential for VTE prophylaxis in patients who could not tolerate even minor amounts of bleeding, such as certain trauma patients. The Eastern Association for the Surgery of Trauma has put forth a consensus recommendation to consider vena caval interruption in high-risk trauma patients who cannot receive pharmacologic prophylaxis.7 A randomized trial evaluating the usefulness of vena caval interruption for patients undergoing surgery is needed. For now, this intervention should be regarded as experimental and considered only on a highly individualized basis.
PHARMACOLOGIC PROPHYLAXIS
Timing of initiation
Pharmacologic VTE prophylaxis generally should begin 8 to 24 hours postoperatively. Of course, adequate hemostasis is required before initiation, and the net risk/benefit tradeoff with regard to timing of anticoagulant initiation has still not been well studied in many surgical patient populations.
Extended prophylaxis
In the update for the 8th edition of its guidelines, the ACCP added an explicit recommendation for extended outpatient prophylaxis with low-molecular-weight heparin (LMWH) for up to 28 days postoperatively in selected high-risk patients undergoing general or gynecologic surgery, including those with cancer or a history of VTE.1 This recommendation was based largely on studies of extended prophylaxis in patients with cancer undergoing colorectal surgery.8
Increased appreciation of the value of extended VTE prophylaxis after discharge is linked to a growing recognition that DVT and PE episodes in the community setting are often related to a recent hospital stay for either medical illness or surgery. A population-based study found that 59% of all community cases of a first lifetime VTE event in residents of Olmsted County, Minn., over a 15-year period could be linked to current or recent (< 30 days) hospitalization or nursing home residence.9 A similar population-based study in the Worcester, Mass., area found that three-fourths of all VTE events in a 3-year period occurred in the outpatient setting.10 Among patients with these outpatient VTE events, a large proportion had undergone surgery (23%) or hospitalization (37%) in the prior 3 months; among those, 67% experienced their VTE within 1 month of their time in the hospital.
These findings are no surprise, since surgery induces a hypercoagulable state that, when combined with individual risk factors such as obesity, old age, or poor heart function, cannot be assumed to return to baseline on postoperative day 4 or 5 just because the patient is being discharged.
Orthopedic surgery
For patients undergoing major orthopedic procedures, the ACCP guidelines recommend against routine screening for VTE with Doppler ultrasonography before discharge if the patient is asymptomatic.1 Such screening is not considered cost-effective because asymptomatic clots often are found, for which treatment is uncertain, and proximal clots may be missed, giving a false sense of security.
New to the ACCP guidelines in the 8th edition is the recommendation that patients undergoing knee arthroscopy who have risk factors for VTE (or whose procedure is complicated) should receive 1 week of prophylaxis with LMWH.1 Also new are recommendations for patients with risk factors undergoing single- or multilevel laminectomy (Table 4).
Recommendations unchanged in neurosurgery, spinal injury, trauma, burns
Recommendations for neurosurgery remain unchanged from the prior (2004) edition of the ACCP guidelines and are still based on the 2000 meta-analysis by Iorio and Agnelli of LMWH prophylaxis in neurosurgery cases.11 In the United States, the standard is overwhelmingly to use mechanical devices for thromboprophylaxis in neurosurgery, even for patients with cancer.
For prophylaxis in surgical patients with spinal cord injury, multisystem trauma, or burns, LMWH is predominantly used, and the ACCP recommendations are unchanged from 2004.
Drug-specific considerations
LMWH vs vitamin K antagonist. Although vitamin K antagonists (warfarin) still appear in the latest ACCP recommendations,1 LMWH is preferable. A 2004 meta-analysis of studies comparing vitamin K antagonists with LMWH for prophylaxis in patients undergoing orthopedic surgery found that vitamin K antagonists were associated with more episodes of total DVT (relative risk [RR] = 1.51; 95% CI, 1.27–1.79) and proximal DVT (RR = 1.51; 95% CI, 1.04–2.17) compared with LMWH.12 No difference was found in rates of wound hematoma or major bleeding. This finding of inferiority for vitamin K antagonists came despite the likelihood that warfarin was more often administered correctly (ie, with dose adjustment to achieve an international normalized ratio [INR] of 2.0 to 3.0 within 72 hours after surgery) in the studies in this analysis than it is in real-world practice.
Fondaparinux. The indirect factor Xa–specific inhibitor fondaparinux has had a surprisingly limited clinical adoption despite having been widely studied and found to be safe and effective. This is likely attributable in part to its 17-hour half-life, which raises concerns that it may take 3 days for its effects to stop if a patient begins to bleed. Large phase 3 studies have found fondaparinux to be equivalent to LMWH in VTE prevention after hip replacement, marginally superior to LMWH after knee replacement, and superior to LMWH following hip fracture repair.13 Fondaparinux was associated with an increase in bleeding events and instances of transfusion requirement, but only in one of the studies, which was in the setting of knee replacement surgery.14
Aspirin not recommended by ACCP. Although aspirin reduces the risk of VTE, practice guidelines from both the ACCP1 and the International Union of Angiology15 contain no recommendation for its use as prophylaxis because it is considered less effective and more risky than other therapies. In contrast, clinical practice guidelines from the American Academy of Orthopaedic Surgeons suggest that aspirin is reasonable for VTE prophylaxis.16 The varying recommendations reflect differences in perspective among these different specialties.
Aspirin has the advantages of ease of use and low cost, but it is clearly not the best evidence-based approach for VTE prophylaxis. The only recent randomized trial evidence in support of aspirin comes from the Pulmonary Embolism Prevention trial, a study with a flawed design involving more than 13,000 patients undergoing surgery for hip fracture or elective arthroplasty in five countries.17 Patients were randomized to receive aspirin 160 mg daily or placebo for 35 days along with any other prophylaxis deemed necessary (an important potential confounder). Aspirin was associated with an absolute reduction in symptomatic events of less than 1% relative to placebo, and no benefit was observed within the first week. The best results with aspirin were among patients with hip fracture. No benefit was shown among patients undergoing hip arthroplasty or knee arthroplasty; in those groups, both the aspirin and placebo recipients were also treated with LMWH. An absolute increase in rates of wound bleeding (0.6% increase) and gastrointestinal bleeding (1.0% increase) was observed in the aspirin group. The absolute increase in complications was greater than the absolute reduction in episodes of symptomatic DVT: for every episode of symptomatic DVT averted, one wound bleed and 10 gastrointestinal bleeds occurred.
SPECIAL PATIENT POPULATIONS
Renal impairment
The 8th edition of the ACCP guidelines recommends that renal function be kept in mind when considering LMWH, fondaparinux, and other antithrombotic drugs that are cleared by the kidneys. Fondaparinux and LMWH can bioaccumulate in patients with renal insufficiency, who have a higher risk of bleeding to begin with, thereby compounding the risk. Options for patients with renal compromise include avoiding drugs that bioaccumulate, using a lower dosage, and monitoring the drug level or anticoagulant effect.1
Fondaparinux is explicitly contraindicated in patients with low body weight (< 50 kg) or renal impairment (creatinine clearance < 30 mL/min). Renal function should be assessed periodically in any patients receiving the drug.18
I also would not use fondaparinux or LMWH in patients with rapidly changing renal function. For patients with chronic, stable renal impairment, one can reduce the dose of LMWH empirically; one LMWH, enoxaparin, has specific dosing guidelines in its package insert (one-third reduction in dose), but this option does not hold for patients with rapidly changing renal function.19
Obesity
The 8th edition of the ACCP guidelines recommends weight-based dosing of thromboprophylactic agents in obese patients. The guidelines particularly recommend that patients undergoing inpatient bariatric surgery be given higher doses of LMWH or unfractionated heparin.1,20
Frederiksen et al measured the anticoagulant effect of a single fixed dose of a LMWH (using anti-factor Xa heparin activity levels) and found that it was dependent on body weight.21 This suggests that fixed doses that are effective in normal-weight patients may have no detectable anti-coagulant effect in patients with very high body weight.
Weight-based dosing: mounting nonprospective evidence. Weight-based dosage adjustment for the morbidly obese has not been directly studied in a prospective, randomized fashion. A nonrandomized study by Scholten et al compared two regimens of enoxaparin (30 mg twice daily vs 40 mg twice daily) among 481 obese patients undergoing bariatric surgery; each regimen was used along with mechanical thromboprophylaxis.22 They found that the higher-dose regimen was associated with significantly fewer postoperative DVT complications (0.6% vs 5.4%; P < .01) without an increase in bleeding complications.
Separately, Shepherd et al used weight-adjusted doses of unfractionated heparin (started on the evening of surgery) to achieve subtherapeutic peak anti–factor Xa heparin activity levels of 0.11 to 0.25 IU/mL in a series of 700 patients undergoing laparoscopic gastric bypass surgery.23 The resulting doses were greater than those in traditional fixed-rate protocols, but rates of bleeding and VTE events were low and comparable to those reported in patients receiving standard doses.
Don’t rule out multimodal approaches. Multimodal prophylaxis can also be used in obese patients and need not be abandoned as a result of size considerations. For instance, two intermittent compression therapy devices can be pieced together with a Velcro binder if a single device is too small to be worn.
EMERGING ANTICOAGULANT OPTIONS
For many years, unfractionated heparin was the only available parenteral anticoagulant. While heparin has broad anticoagulant properties, it also has well-established limitations, including the need for parenteral delivery, recent problems related to contamination (it is derived from pig intestines), and of course heparin-induced thrombocytopenia (HIT). HIT is an immune-mediated form of platelet activation that can lead to widespread thrombosis throughout the body. It is more commonly associated with venous thrombosis, but arterial events with limb-threatening ischemia may also occur. LMWH is associated with a reduced risk of HIT, but LMWH does not avoid the risk entirely.
Beyond the issue of avoiding HIT, newer anticoagulant therapies are being developed with the aim of oral administration and more targeted inhibition of coagulation factors IIa (thrombin) and Xa.24
Oral direct thrombin inhibitors
One of the two most promising classes of emerging anticoagulants is the direct thrombin inhibitors, most of which are being developed for oral administration. There were high hopes for the initial compound in this class, ximelagatran, but it was abandoned about 5 years ago because of hepatotoxicity.
Dabigatran is the direct thrombin inhibitor furthest along in development today. Currently approved in Europe for prevention of VTE in patients undergoing total hip or knee replacement surgery, dabigatran is likely to be available soon in the United States. It is administered orally, has a rapid onset of action (< 1 hour), and has a predictable anticoagulant response that requires no monitoring.24 Because dabigatran is excreted essentially unchanged by the kidneys and may bioaccumulate, it should not be used in patients with renal impairment or rapidly changing renal function.
In phase 3 clinical trials for VTE prevention in knee replacement surgery, dabigatran was at least as effective as enoxaparin 40 mg once daily and had a comparable safety profile,25 but it was slightly less effective than enoxaparin 30 mg twice daily.26 In a phase 3 trial in patients undergoing hip replacement surgery, dabigatran was equivalent in efficacy and safety to enoxaparin 40 mg once daily.27
Oral direct factor Xa inhibitors
A key rationale for direct inhibition of factor Xa is that it results in inhibition of thrombin production on the activated platelet. Whereas fondaparinux is an indirect inhibitor of factor Xa, direct factor Xa inhibitors offer an advantage in that they inhibit factor Xa within the prothrombinase complex, which occurs on the surface of a platelet and is the main site for thrombin development (very little thrombin is actually produced on endothelial cells). Recall the adage that “thrombin begets more thrombin”: it activates not only platelets but the intrinsic and extrinsic pathways.28
Factor Xa may be a better target than thrombin for a number of other reasons:
- Factor Xa is believed to have few functions (compared with thrombin) outside of coagulation
- In vitro studies show that factor Xa has a wider therapeutic window than thrombin, which translates to greater separation between drug levels that will confer efficacy and bleeding
- Thrombin inhibitors are associated with rebound thrombin generation (there is no evidence of this with factor Xa inhibitors)
- The efficacy of heparin-based anticoagulants improves as selectivity for factor Xa increases (unfractionated heparin is less effective than LMWH, which is less effective than fondaparinux).
Two direct factor Xa inhibitors—both administered orally—are far along in development, as detailed below.
Apixaban has shown promise, but the phase 3 ADVANCE-1 study of apixaban for VTE prevention in patients undergoing knee surgery did not meet statistical criteria for noninferiority compared with enoxaparin 30 mg twice daily.29 This prompted a delay in regulatory filings for apixaban in the United States, and the drug’s prospects for approval for VTE prevention may be unclear until release of results from two other comparative phase 3 trials with enoxaparin in 2009 and 2010.
Rivaroxaban is more likely to become clinically available soon, in light of recent results from the phase 3 RECORD4 trial demonstrating that it was significantly superior to enoxaparin 30 mg twice daily in preventing VTE following knee replacement surgery with comparable rates of major bleeding.30
DISCUSSION
Question from the audience: Some surgeons in my hospital prescribe warfarin immediately after surgery without a bridge of LMWH. Is that appropriate?
Dr. Michota: Warfarin is an option for prophylaxis in orthopedic surgery, beginning on the day of surgery. It could even be started the day before surgery, but the dose should be monitored to achieve an INR between 2.0 and 3.0 within 72 hours of the procedure. If the INR is not in this optimum range, prophylactic doses of LMWH can be given until it is therapeutic.
Follow-up question: In practice, do you actually encourage INR monitoring? Usually we just put patients on a certain dose without monitoring. When we do check the INR, it’s usually 1.4 or 1.5.
Dr. Michota: Warfarin was shown to be effective in reducing VTE risk in orthopedic surgery with dose adjustment based on INR monitoring. On that basis, warfarin remains in the guideline recommendations. Unmonitored, warfarin has not been shown to reduce risk, so to give it that way would not be evidence-based.
Question from the audience: I work with several plastic surgeons who use compression stockings intraoperatively because they’ve heard of several patients who developed a PE during surgery. Is there any benefit to using compression stockings for 2 to 3 hours and then sending the patient home?
Dr. Michota: I don’t know. Theoretically, a device that is on and working before induction may reduce stasis.
The plastic surgery societies do have guidelines. Risk depends on the type of plastic surgery procedure; for example, risk probably increases due to inflammation in procedures that involve scraping the fat pads.
This is an area where we don’t have much data. These patients may be at risk, but we don’t know the best way to mitigate it. It is important that risks be discussed with patients in the informed-consent process and be documented. If the surgeon thinks it is reasonable to give pharmacologic prophylaxis after surgery, I wouldn’t hesitate to do that, but any form of bleeding in the setting of plastic surgery is catastrophic because it defeats the reason for which the surgery was done in the first place.
Question from the audience: How do the guidelines address being aggressive with pharmacologic thromboprophylaxis when a patient is already taking dual antiplatelet therapy?
Dr. Michota: For patients with an indication for VTE prophylaxis in a setting for which there is a specific strategy, the ACCP guidelines recommend that they be put on that regimen whether they are on antiplatelet agents or not. For example, consider a high-risk patient having colorectal surgery who should get unfractionated heparin or LMWH postoperatively and who is currently taking clopidogrel and aspirin. There is no evidence that the dual aspirin–clopidogrel therapy alone is effective in decreasing the risk of DVT. However, we do know that if we add on additional agents, the risk of bleeding is increased. The guidelines consider risk and benefit, and they recommend adding the agents that we know work to prevent DVT.
Question from the audience: You briefly mentioned prophylaxis for knee arthroscopy, which is the most frequently performed orthopedic procedure. Do these recommendations apply to all patients undergoing knee arthroscopy?
Dr. Michota: No. Prophylaxis is indicated only for patients with what the ACCP considers to be additional risk factors for thrombosis. They didn’t specify which risk factors, but good indications for prophylaxis would include morbid obesity, limited mobility after the procedure, a personal history of DVT, features of stasis noted on physical examination, stasis dermatitis (or other features that could indicate prior thrombosis), advanced age, and malignancy. If a patient undergoing knee arthroscopy has other nonmodifiable risk factors, you should also think about prophylaxis. But the vast majority of patients do not need it.
Question from the audience: I’m an academic hospitalist who works closely with orthopedic surgeons. Certain surgeons will only use aspirin for prophylaxis, and it is nonnegotiable. Where does that leave me from a medicolegal standpoint? Our model is to follow ACCP recommendations, but these orthopedic surgeons still use only aspirin.
Dr. Michota: You must do everything you can to come to a consensus with your surgeon colleagues. If you are uncomfortable, as a group you must say to the surgeons, “We are uncomfortable. This is how we view the data. How do you view the data?” If they answer, “We’re doing it because it’s easy, and the American Academy of Orthopaedic Surgeons says we can do it,” I don’t have a good response. But it is more likely that their use of aspirin is based on their own observations; they may not see many clots. Of course, the problem with observational data is that the numbers are not large and they are not generated in a randomized and prospective fashion. Perhaps you can come to some middle ground, but you could always make the difficult choice and say, “I’m just not going to follow your patients.”
- Geerts WH, Bergqvist D, Pineo GF, et al; American College of Chest Physicians. Prevention of venous thromboembolism: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest 2008; 133:381S–453S.
- Medicare Quality Improvement Community (MedQIC) Web site. http://www.medqic.org. Accessed June 1, 2009.
- Surgical Care Improvement Project (SCIP). Colorado Foundation for Medical Care Web site. http://www.cfmc.org/hospital/hospital_scip.htm. Accessed June 1, 2009.
- Kucher N, Koo S, Quiroz R, et al. Electronic alerts to prevent venous thromboembolism among hospitalized patients. N Engl J Med 2005; 352:969–977.
- Anderson FA Jr, Spencer FA. Risk factors for venous thromboembolism. Circulation 2003; 107:I-9–I-16.
- Amaragiri SV, Lees TA. Elastic compression stockings for prevention of deep vein thrombosis. Cochrane Database Syst Rev 2000; (3):CD001484.
- Rogers FB, Cipolle MD, Velmahos G, et al. Practice management guidelines for the prevention of venous thromboembolism in trauma patients: the EAST Practice Management Work Group. J Trauma 2002; 53:142–164.
- Bergqvist D, Agnelli G, Cohen AT, et al. Duration of prophylaxis against venous thromboembolism with enoxaparin after surgery for cancer. N Engl J Med 2002; 346:975–980.
- Heit JA, O’Fallon WM, Petterson TM, et al. Relative impact of risk factors for deep vein thrombosis and pulmonary embolism: a population-based study. Arch Intern Med 2002; 162:1245–1248.
- Spencer FA, Lessard D, Emery C, et al. Venous thromboembolism in the outpatient setting. Arch Intern Med 2007; 167:1471–1475.
- Iorio A, Agnelli G. Low-molecular-weight and unfractionated heparin for prevention of venous thromboembolism in neurosurgery: a meta-analysis. Arch Intern Med 2000; 160:2327–2332.
- Mismetti P, Laporte S, Zufferey P, et al. Prevention of venous thromboembolism in orthopedic surgery with vitamin K antagonists: a meta-analysis. J Thromb Haemost 2004; 2:1058–1070.
- Turpie AG, Bauer KA, Eriksson BI, Lassen MR. Fondaparinux vs enoxaparin for the prevention of venous thromboembolism in major orthopedic surgery: a meta-analysis of 4 randomized double-blind studies. Arch Intern Med 2002; 162:1833–1840.
- Bauer KA, Eriksson BI, Lassen MR, Turpie AG; Steering Committee of the Pentasaccharide in Major Knee Surgery Study. Fondaparinux compared with enoxaparin for the prevention of venous thromboembolism after elective major knee surgery. N Engl J Med 2001; 345:1305–1310.
- Cardiovascular Disease Educational and Research Trust; Cyprus Cardiovascular Disease Educational and Research Trust; European Venous Forum; International Surgical Thrombosis Forum; International Union of Angiology; Union Internationale de Phlébologie. Prevention and treatment of venous thromboembolism. International Consensus Statement (guidelines according to scientific evidence). Int Angiol 2006; 25:101–161.
- American Academy of Orthopaedic Surgeons clinical guideline on prevention of symptomatic pulmonary embolism in patients undergoing total hip or knee arthroplasty. http://www.aaos.org/research/guidelines/PE_summary.pdf. Accessed June 5, 2009.
- Prevention of pulmonary embolism and deep vein thrombosis with low dose aspirin: Pulmonary Embolism Prevention (PEP) trial. Lancet 2000; 355:1295-1302.
- Arixtra injection [package insert]. Research Triangle Park, NC: GlaxoSmithKline; 2008.
- Sanderink GJ, Guimart C, Jariwala N, et al. Enoxaparin pharmacokinetics and pharmacodynamics in renal impairment. J Am Coll Cardiol 2001; 37(suppl A):229A. Abstract.
- Hirsh J, Bauer KA, Donati MB, et al; American College of Chest Physicians. Parenteral anticoagulants: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition) [published correction appears in Chest 2008; 134:473]. Chest 2008; 133:141S–159S.
- Frederiksen SG, Hedenbro JL, Norgren L. Enoxaparin effect depends on body-weight and current doses may be inadequate in obese patients. Br J Surg 2003; 90:547–548.
- Scholten DJ, Hoedema RM, Scholten SE. A comparison of two different prophylactic dose regimens of low molecular weight heparin in bariatric surgery. Obes Surg 2002; 12:19–24.
- Shepherd MF, Rosborough TK, Schwartz ML. Heparin thromboprophylaxis in gastric bypass surgery. Obes Surg 2003; 13:249–253.
- Weitz JI, Bates SM. New anticoagulants. J Thromb Haemost 2005; 3:1843–1853.
- Eriksson BI, Dahl OE, Rosencher N, et al. Oral dabigatran etexilate vs. subcutaneous enoxaparin for the prevention of venous thromboembolism after total knee replacement: the RE-MODEL randomized trial. J Thromb Haemost 2007; 5:2178–2185.
- RE-MOBILIZE Writing Committee, Ginsberg JS, Davidson BL, et al. Oral thrombin inhibitor dabigatran etexilate vs North American enoxaparin regimen for prevention of venous thromboembolism after knee arthroplasty surgery. J Arthroplasty 2009; 24:1–9.
- Eriksson BI, Dahl OE, Rosencher N, et al; RE-NOVATE Study Group. Dabigatran etexilate versus enoxaparin for prevention of venous thromboembolism after total hip replacement: a randomised, double-blind, non-inferiority trial [published correction appears in Lancet 2007; 370:2004]. Lancet 2007: 370:949–956.
- Hoffman M, Monroe DM 3rd, Roberts HR. Activated factor VII activates factors IX and X on the surface of activated platelets: thoughts on the mechanism of action of high-dose activated factor VII. Blood Coagul Fibrinolysis 1998; 9(suppl 1):S61–S65.
- Bristol-Myers Squibb and Pfizer provide update on apixaban clinical development program [press release]. New York, NY: August 27, 2008.
- Turpie AG, Lassen MR, Davidson BL, et al. Rivaroxaban versus enoxaparin for thromboprophylaxis after total knee arthroplasty (RECORD4): a randomised trial. Lancet 2009; 373:1673–1680.
Most surgical patients who require hospitalization should be considered at high risk for venous thromboembolism (VTE) and be given appropriate prophylaxis. For lower-risk procedures such as knee arthroscopy, prophylaxis is needed for those with individual risk factors such as morbid obesity, limited mobility after surgery, or a history of deep vein thrombosis (DVT) or malignancy. Too often, however, prophylaxis is not provided appropriately or not given at all.
This review surveys the essentials of perioperative VTE prophylaxis and important new developments in the field, which include the 2008 release of new evidence-based clinical practice guidelines on antithrombotic and thrombolytic therapy from the American College of Chest Physicians (ACCP). This 8th edition of the guidelines updates the previous edition, published in 2004, and includes a section by Geerts et al devoted to VTE prevention.1 Other major guidelines are also discussed, as are developments in VTE-related quality measurement, management of special patient populations (those with renal impairment or morbid obesity), and emerging therapies for VTE prophylaxis.
IMPETUS FOR QUALITY IMPROVEMENT IN VTE
A new seriousness about VTE quality measures
The 8th edition of the ACCP guidelines recommends that every hospital develop a formal, active strategy to consistently identify medical and surgical patients at risk for VTE and to prevent VTE occurrence.1 Although prior editions of the ACCP guidelines have made this recommendation for more than 2 decades, fewer than 1 in 10 acute care hospitals had any such strategy in place as recently as 5 years ago. Now, however, most US hospitals have implemented such a strategy, thanks to the growing national emphasis on health care quality measurement in recent years.
The Surgical Care Improvement Project (SCIP) has been at the forefront of this recent quality measures movement. SCIP, a joint project of the American Medical Association and federal government agencies, set a goal to reduce surgical complications in the United States by 25% from 2005 to 2010.2 Two SCIP process measures relate to improving VTE prophylaxis2,3:
- The proportion of surgical patients for whom recommended VTE prophylaxis is ordered
- The proportion of surgical patients who actually receive appropriate VTE prophylaxis within 24 hours before or after surgery.
The Joint Commission and the National Quality Forum recently endorsed these two SCIP performance measures for perioperative VTE prophylaxis along with several others relating to VTE treatment.
CMS raises the stakes with reimbursement restrictions
More significantly, the federal government’s Centers for Medicare and Medicaid Services (CMS) will soon refuse to reimburse for hospital treatment of a primary diagnosis of DVT or pulmonary embolism (PE) following recent (within 30 days) total hip or knee arthroplasty. Effective October 1, 2009, a primary VTE diagnosis following these joint replacement procedures will be added to CMS’ current list of “never events,” or hospital-acquired conditions for which CMS will not provide reimbursement because they are considered the result of preventable medical errors. (Notably, treatment of DVT or PE as a secondary diagnosis will still be reimbursed—for example, if a joint replacement patient develops nosocomial pneumonia, is transferred to the intensive care unit, and then develops VTE.) This addition of DVT and PE to the list is highly controversial since these events sometimes develop even if prophylactic therapy is appropriate and aggressive.
Strategies to promote best practices
In the update for the new 8th edition of its guidelines, the ACCP added recommendations on specific ways for hospitals to identify patients at high risk for VTE and ensure that they receive appropriate prophylaxis. These include the use of computer decision-support systems, preprinted orders, and periodic audit and feedback.1
Researchers at Brigham and Women’s Hospital evaluated the effectiveness of a computer alert system for notifying physicians of newly hospitalized patients at risk for DVT who were not receiving prevention therapy within the first 24 hours of hospital admission.4 These patients presumably “fell through the cracks” and warranted prophylaxis but were otherwise not recognized by the health care team. Risk was determined by a scoring system based on multiple variables, including malignancy, previous DVT or PE, hypercoagulability, major surgery, advanced age, obesity, ordered bed rest, and treatment with hormone replacement therapy or oral contraceptives. Study physicians had to acknowledge having received the alert but could choose whether or not to order VTE prophylaxis. Prophylaxis was used in considerably more patients from the intervention group than from a control group of high-risk patients whose physicians did not receive alerts (34% vs 14%, respectively); accordingly, the risk of a symptomatic DVT or PE event at 90 days was reduced by 41% in the intervention group.
Despite this evidence of improved practice under the alert system, the study begs the question of why the percentage of patients at risk for VTE who were given prophylaxis was still so low (34%), demonstrating how much progress in improving practice remains to be achieved.
PROPHYLAXIS STRATEGIES: MATCHING THERAPY TO RISK
NONPHARMACOLOGIC PROPHYLAXIS STRATEGIES
Does ambulation prevent DVT?
Although it is commonly accepted that walking prevents DVT, this has never been directly tested. Walking may simply be a marker of health, and healthy people are less prone to develop thromboses. We have almost no evidence to show that forcing an unhealthy person to walk helps prevent DVT. Early ambulation offers many benefits and should be encouraged, but it should not be considered DVT prophylaxis; it is simply good hospital care.
Mechanical devices: Adherence is key
Amaragiri and Lees conducted a systematic literature review of randomized controlled trials evaluating the effectiveness of graduated compression stockings (elastic stockings) for preventing DVT in various groups of hospitalized patients.6 The analysis demonstrated a statistically significant reduction in DVT incidence with graduated compression stockings compared with control both among the nine trials in which stockings were used alone (odds ratio = 0.34) and among the seven trials in which stockings were used in addition to another method of thromboprophylaxis (odds ratio = 0.24). Although benefit was demonstrated, many of the trials in this review involved patients undergoing gynecologic surgery and date from the 1970s and 1980s (when obesity was less prevalent), so the applicability of their results today may be limited.
The 8th edition of the ACCP guidelines recommends that mechanical methods of VTE prophylaxis be used primarily in patients who are at high risk of bleeding and that careful attention be directed to ensuring their proper use and optimal adherence.1 The latter point about adherence cannot be emphasized enough, as graduated compression stockings and other mechanical devices have been shown not to be effective unless they are worn at least 18 to 20 hours a day. This degree of adherence is difficult to achieve, as it can severely limit patient mobility and leave patients susceptible to development of pressure ulcers.
Mechanical compression should be initiated prior to induction of anesthesia and continue intraoperatively and then into the postanesthesia care unit. Orders for use of mechanical devices should include instructions in the patient’s medical chart specifying how—and for how many hours per day—they are to be worn. Not doing so leaves the physician vulnerable to litigation, especially as the ACCP guidelines include language on optimal adherence to these devices (“they should be removed for only a short time each day when the patient is actually walking or for bathing”1).
Continuous external compression therapy
Newer mechanical device options include a continuous external compression therapy system that allows patients to be mobile while wearing it and provides rhythmic compression that results in good peak venous flows. Ideally such a device could be put on the patient preoperatively and worn during surgery, throughout the hospital stay, and even at home during recovery. Anecdotally, however, I see patients turn these new devices off at the side of the bed just as often as they do with traditional devices.
Vena caval interruption
Vena caval interruption involves placement of a retrievable vena cava filter before surgery and removal some time later; it offers the potential for VTE prophylaxis in patients who could not tolerate even minor amounts of bleeding, such as certain trauma patients. The Eastern Association for the Surgery of Trauma has put forth a consensus recommendation to consider vena caval interruption in high-risk trauma patients who cannot receive pharmacologic prophylaxis.7 A randomized trial evaluating the usefulness of vena caval interruption for patients undergoing surgery is needed. For now, this intervention should be regarded as experimental and considered only on a highly individualized basis.
PHARMACOLOGIC PROPHYLAXIS
Timing of initiation
Pharmacologic VTE prophylaxis generally should begin 8 to 24 hours postoperatively. Of course, adequate hemostasis is required before initiation, and the net risk/benefit tradeoff with regard to timing of anticoagulant initiation has still not been well studied in many surgical patient populations.
Extended prophylaxis
In the update for the 8th edition of its guidelines, the ACCP added an explicit recommendation for extended outpatient prophylaxis with low-molecular-weight heparin (LMWH) for up to 28 days postoperatively in selected high-risk patients undergoing general or gynecologic surgery, including those with cancer or a history of VTE.1 This recommendation was based largely on studies of extended prophylaxis in patients with cancer undergoing colorectal surgery.8
Increased appreciation of the value of extended VTE prophylaxis after discharge is linked to a growing recognition that DVT and PE episodes in the community setting are often related to a recent hospital stay for either medical illness or surgery. A population-based study found that 59% of all community cases of a first lifetime VTE event in residents of Olmsted County, Minn., over a 15-year period could be linked to current or recent (< 30 days) hospitalization or nursing home residence.9 A similar population-based study in the Worcester, Mass., area found that three-fourths of all VTE events in a 3-year period occurred in the outpatient setting.10 Among patients with these outpatient VTE events, a large proportion had undergone surgery (23%) or hospitalization (37%) in the prior 3 months; among those, 67% experienced their VTE within 1 month of their time in the hospital.
These findings are no surprise, since surgery induces a hypercoagulable state that, when combined with individual risk factors such as obesity, old age, or poor heart function, cannot be assumed to return to baseline on postoperative day 4 or 5 just because the patient is being discharged.
Orthopedic surgery
For patients undergoing major orthopedic procedures, the ACCP guidelines recommend against routine screening for VTE with Doppler ultrasonography before discharge if the patient is asymptomatic.1 Such screening is not considered cost-effective because asymptomatic clots often are found, for which treatment is uncertain, and proximal clots may be missed, giving a false sense of security.
New to the ACCP guidelines in the 8th edition is the recommendation that patients undergoing knee arthroscopy who have risk factors for VTE (or whose procedure is complicated) should receive 1 week of prophylaxis with LMWH.1 Also new are recommendations for patients with risk factors undergoing single- or multilevel laminectomy (Table 4).
Recommendations unchanged in neurosurgery, spinal injury, trauma, burns
Recommendations for neurosurgery remain unchanged from the prior (2004) edition of the ACCP guidelines and are still based on the 2000 meta-analysis by Iorio and Agnelli of LMWH prophylaxis in neurosurgery cases.11 In the United States, the standard is overwhelmingly to use mechanical devices for thromboprophylaxis in neurosurgery, even for patients with cancer.
For prophylaxis in surgical patients with spinal cord injury, multisystem trauma, or burns, LMWH is predominantly used, and the ACCP recommendations are unchanged from 2004.
Drug-specific considerations
LMWH vs vitamin K antagonist. Although vitamin K antagonists (warfarin) still appear in the latest ACCP recommendations,1 LMWH is preferable. A 2004 meta-analysis of studies comparing vitamin K antagonists with LMWH for prophylaxis in patients undergoing orthopedic surgery found that vitamin K antagonists were associated with more episodes of total DVT (relative risk [RR] = 1.51; 95% CI, 1.27–1.79) and proximal DVT (RR = 1.51; 95% CI, 1.04–2.17) compared with LMWH.12 No difference was found in rates of wound hematoma or major bleeding. This finding of inferiority for vitamin K antagonists came despite the likelihood that warfarin was more often administered correctly (ie, with dose adjustment to achieve an international normalized ratio [INR] of 2.0 to 3.0 within 72 hours after surgery) in the studies in this analysis than it is in real-world practice.
Fondaparinux. The indirect factor Xa–specific inhibitor fondaparinux has had a surprisingly limited clinical adoption despite having been widely studied and found to be safe and effective. This is likely attributable in part to its 17-hour half-life, which raises concerns that it may take 3 days for its effects to stop if a patient begins to bleed. Large phase 3 studies have found fondaparinux to be equivalent to LMWH in VTE prevention after hip replacement, marginally superior to LMWH after knee replacement, and superior to LMWH following hip fracture repair.13 Fondaparinux was associated with an increase in bleeding events and instances of transfusion requirement, but only in one of the studies, which was in the setting of knee replacement surgery.14
Aspirin not recommended by ACCP. Although aspirin reduces the risk of VTE, practice guidelines from both the ACCP1 and the International Union of Angiology15 contain no recommendation for its use as prophylaxis because it is considered less effective and more risky than other therapies. In contrast, clinical practice guidelines from the American Academy of Orthopaedic Surgeons suggest that aspirin is reasonable for VTE prophylaxis.16 The varying recommendations reflect differences in perspective among these different specialties.
Aspirin has the advantages of ease of use and low cost, but it is clearly not the best evidence-based approach for VTE prophylaxis. The only recent randomized trial evidence in support of aspirin comes from the Pulmonary Embolism Prevention trial, a study with a flawed design involving more than 13,000 patients undergoing surgery for hip fracture or elective arthroplasty in five countries.17 Patients were randomized to receive aspirin 160 mg daily or placebo for 35 days along with any other prophylaxis deemed necessary (an important potential confounder). Aspirin was associated with an absolute reduction in symptomatic events of less than 1% relative to placebo, and no benefit was observed within the first week. The best results with aspirin were among patients with hip fracture. No benefit was shown among patients undergoing hip arthroplasty or knee arthroplasty; in those groups, both the aspirin and placebo recipients were also treated with LMWH. An absolute increase in rates of wound bleeding (0.6% increase) and gastrointestinal bleeding (1.0% increase) was observed in the aspirin group. The absolute increase in complications was greater than the absolute reduction in episodes of symptomatic DVT: for every episode of symptomatic DVT averted, one wound bleed and 10 gastrointestinal bleeds occurred.
SPECIAL PATIENT POPULATIONS
Renal impairment
The 8th edition of the ACCP guidelines recommends that renal function be kept in mind when considering LMWH, fondaparinux, and other antithrombotic drugs that are cleared by the kidneys. Fondaparinux and LMWH can bioaccumulate in patients with renal insufficiency, who have a higher risk of bleeding to begin with, thereby compounding the risk. Options for patients with renal compromise include avoiding drugs that bioaccumulate, using a lower dosage, and monitoring the drug level or anticoagulant effect.1
Fondaparinux is explicitly contraindicated in patients with low body weight (< 50 kg) or renal impairment (creatinine clearance < 30 mL/min). Renal function should be assessed periodically in any patients receiving the drug.18
I also would not use fondaparinux or LMWH in patients with rapidly changing renal function. For patients with chronic, stable renal impairment, one can reduce the dose of LMWH empirically; one LMWH, enoxaparin, has specific dosing guidelines in its package insert (one-third reduction in dose), but this option does not hold for patients with rapidly changing renal function.19
Obesity
The 8th edition of the ACCP guidelines recommends weight-based dosing of thromboprophylactic agents in obese patients. The guidelines particularly recommend that patients undergoing inpatient bariatric surgery be given higher doses of LMWH or unfractionated heparin.1,20
Frederiksen et al measured the anticoagulant effect of a single fixed dose of a LMWH (using anti-factor Xa heparin activity levels) and found that it was dependent on body weight.21 This suggests that fixed doses that are effective in normal-weight patients may have no detectable anti-coagulant effect in patients with very high body weight.
Weight-based dosing: mounting nonprospective evidence. Weight-based dosage adjustment for the morbidly obese has not been directly studied in a prospective, randomized fashion. A nonrandomized study by Scholten et al compared two regimens of enoxaparin (30 mg twice daily vs 40 mg twice daily) among 481 obese patients undergoing bariatric surgery; each regimen was used along with mechanical thromboprophylaxis.22 They found that the higher-dose regimen was associated with significantly fewer postoperative DVT complications (0.6% vs 5.4%; P < .01) without an increase in bleeding complications.
Separately, Shepherd et al used weight-adjusted doses of unfractionated heparin (started on the evening of surgery) to achieve subtherapeutic peak anti–factor Xa heparin activity levels of 0.11 to 0.25 IU/mL in a series of 700 patients undergoing laparoscopic gastric bypass surgery.23 The resulting doses were greater than those in traditional fixed-rate protocols, but rates of bleeding and VTE events were low and comparable to those reported in patients receiving standard doses.
Don’t rule out multimodal approaches. Multimodal prophylaxis can also be used in obese patients and need not be abandoned as a result of size considerations. For instance, two intermittent compression therapy devices can be pieced together with a Velcro binder if a single device is too small to be worn.
EMERGING ANTICOAGULANT OPTIONS
For many years, unfractionated heparin was the only available parenteral anticoagulant. While heparin has broad anticoagulant properties, it also has well-established limitations, including the need for parenteral delivery, recent problems related to contamination (it is derived from pig intestines), and of course heparin-induced thrombocytopenia (HIT). HIT is an immune-mediated form of platelet activation that can lead to widespread thrombosis throughout the body. It is more commonly associated with venous thrombosis, but arterial events with limb-threatening ischemia may also occur. LMWH is associated with a reduced risk of HIT, but LMWH does not avoid the risk entirely.
Beyond the issue of avoiding HIT, newer anticoagulant therapies are being developed with the aim of oral administration and more targeted inhibition of coagulation factors IIa (thrombin) and Xa.24
Oral direct thrombin inhibitors
One of the two most promising classes of emerging anticoagulants is the direct thrombin inhibitors, most of which are being developed for oral administration. There were high hopes for the initial compound in this class, ximelagatran, but it was abandoned about 5 years ago because of hepatotoxicity.
Dabigatran is the direct thrombin inhibitor furthest along in development today. Currently approved in Europe for prevention of VTE in patients undergoing total hip or knee replacement surgery, dabigatran is likely to be available soon in the United States. It is administered orally, has a rapid onset of action (< 1 hour), and has a predictable anticoagulant response that requires no monitoring.24 Because dabigatran is excreted essentially unchanged by the kidneys and may bioaccumulate, it should not be used in patients with renal impairment or rapidly changing renal function.
In phase 3 clinical trials for VTE prevention in knee replacement surgery, dabigatran was at least as effective as enoxaparin 40 mg once daily and had a comparable safety profile,25 but it was slightly less effective than enoxaparin 30 mg twice daily.26 In a phase 3 trial in patients undergoing hip replacement surgery, dabigatran was equivalent in efficacy and safety to enoxaparin 40 mg once daily.27
Oral direct factor Xa inhibitors
A key rationale for direct inhibition of factor Xa is that it results in inhibition of thrombin production on the activated platelet. Whereas fondaparinux is an indirect inhibitor of factor Xa, direct factor Xa inhibitors offer an advantage in that they inhibit factor Xa within the prothrombinase complex, which occurs on the surface of a platelet and is the main site for thrombin development (very little thrombin is actually produced on endothelial cells). Recall the adage that “thrombin begets more thrombin”: it activates not only platelets but the intrinsic and extrinsic pathways.28
Factor Xa may be a better target than thrombin for a number of other reasons:
- Factor Xa is believed to have few functions (compared with thrombin) outside of coagulation
- In vitro studies show that factor Xa has a wider therapeutic window than thrombin, which translates to greater separation between drug levels that will confer efficacy and bleeding
- Thrombin inhibitors are associated with rebound thrombin generation (there is no evidence of this with factor Xa inhibitors)
- The efficacy of heparin-based anticoagulants improves as selectivity for factor Xa increases (unfractionated heparin is less effective than LMWH, which is less effective than fondaparinux).
Two direct factor Xa inhibitors—both administered orally—are far along in development, as detailed below.
Apixaban has shown promise, but the phase 3 ADVANCE-1 study of apixaban for VTE prevention in patients undergoing knee surgery did not meet statistical criteria for noninferiority compared with enoxaparin 30 mg twice daily.29 This prompted a delay in regulatory filings for apixaban in the United States, and the drug’s prospects for approval for VTE prevention may be unclear until release of results from two other comparative phase 3 trials with enoxaparin in 2009 and 2010.
Rivaroxaban is more likely to become clinically available soon, in light of recent results from the phase 3 RECORD4 trial demonstrating that it was significantly superior to enoxaparin 30 mg twice daily in preventing VTE following knee replacement surgery with comparable rates of major bleeding.30
DISCUSSION
Question from the audience: Some surgeons in my hospital prescribe warfarin immediately after surgery without a bridge of LMWH. Is that appropriate?
Dr. Michota: Warfarin is an option for prophylaxis in orthopedic surgery, beginning on the day of surgery. It could even be started the day before surgery, but the dose should be monitored to achieve an INR between 2.0 and 3.0 within 72 hours of the procedure. If the INR is not in this optimum range, prophylactic doses of LMWH can be given until it is therapeutic.
Follow-up question: In practice, do you actually encourage INR monitoring? Usually we just put patients on a certain dose without monitoring. When we do check the INR, it’s usually 1.4 or 1.5.
Dr. Michota: Warfarin was shown to be effective in reducing VTE risk in orthopedic surgery with dose adjustment based on INR monitoring. On that basis, warfarin remains in the guideline recommendations. Unmonitored, warfarin has not been shown to reduce risk, so to give it that way would not be evidence-based.
Question from the audience: I work with several plastic surgeons who use compression stockings intraoperatively because they’ve heard of several patients who developed a PE during surgery. Is there any benefit to using compression stockings for 2 to 3 hours and then sending the patient home?
Dr. Michota: I don’t know. Theoretically, a device that is on and working before induction may reduce stasis.
The plastic surgery societies do have guidelines. Risk depends on the type of plastic surgery procedure; for example, risk probably increases due to inflammation in procedures that involve scraping the fat pads.
This is an area where we don’t have much data. These patients may be at risk, but we don’t know the best way to mitigate it. It is important that risks be discussed with patients in the informed-consent process and be documented. If the surgeon thinks it is reasonable to give pharmacologic prophylaxis after surgery, I wouldn’t hesitate to do that, but any form of bleeding in the setting of plastic surgery is catastrophic because it defeats the reason for which the surgery was done in the first place.
Question from the audience: How do the guidelines address being aggressive with pharmacologic thromboprophylaxis when a patient is already taking dual antiplatelet therapy?
Dr. Michota: For patients with an indication for VTE prophylaxis in a setting for which there is a specific strategy, the ACCP guidelines recommend that they be put on that regimen whether they are on antiplatelet agents or not. For example, consider a high-risk patient having colorectal surgery who should get unfractionated heparin or LMWH postoperatively and who is currently taking clopidogrel and aspirin. There is no evidence that the dual aspirin–clopidogrel therapy alone is effective in decreasing the risk of DVT. However, we do know that if we add on additional agents, the risk of bleeding is increased. The guidelines consider risk and benefit, and they recommend adding the agents that we know work to prevent DVT.
Question from the audience: You briefly mentioned prophylaxis for knee arthroscopy, which is the most frequently performed orthopedic procedure. Do these recommendations apply to all patients undergoing knee arthroscopy?
Dr. Michota: No. Prophylaxis is indicated only for patients with what the ACCP considers to be additional risk factors for thrombosis. They didn’t specify which risk factors, but good indications for prophylaxis would include morbid obesity, limited mobility after the procedure, a personal history of DVT, features of stasis noted on physical examination, stasis dermatitis (or other features that could indicate prior thrombosis), advanced age, and malignancy. If a patient undergoing knee arthroscopy has other nonmodifiable risk factors, you should also think about prophylaxis. But the vast majority of patients do not need it.
Question from the audience: I’m an academic hospitalist who works closely with orthopedic surgeons. Certain surgeons will only use aspirin for prophylaxis, and it is nonnegotiable. Where does that leave me from a medicolegal standpoint? Our model is to follow ACCP recommendations, but these orthopedic surgeons still use only aspirin.
Dr. Michota: You must do everything you can to come to a consensus with your surgeon colleagues. If you are uncomfortable, as a group you must say to the surgeons, “We are uncomfortable. This is how we view the data. How do you view the data?” If they answer, “We’re doing it because it’s easy, and the American Academy of Orthopaedic Surgeons says we can do it,” I don’t have a good response. But it is more likely that their use of aspirin is based on their own observations; they may not see many clots. Of course, the problem with observational data is that the numbers are not large and they are not generated in a randomized and prospective fashion. Perhaps you can come to some middle ground, but you could always make the difficult choice and say, “I’m just not going to follow your patients.”
Most surgical patients who require hospitalization should be considered at high risk for venous thromboembolism (VTE) and be given appropriate prophylaxis. For lower-risk procedures such as knee arthroscopy, prophylaxis is needed for those with individual risk factors such as morbid obesity, limited mobility after surgery, or a history of deep vein thrombosis (DVT) or malignancy. Too often, however, prophylaxis is not provided appropriately or not given at all.
This review surveys the essentials of perioperative VTE prophylaxis and important new developments in the field, which include the 2008 release of new evidence-based clinical practice guidelines on antithrombotic and thrombolytic therapy from the American College of Chest Physicians (ACCP). This 8th edition of the guidelines updates the previous edition, published in 2004, and includes a section by Geerts et al devoted to VTE prevention.1 Other major guidelines are also discussed, as are developments in VTE-related quality measurement, management of special patient populations (those with renal impairment or morbid obesity), and emerging therapies for VTE prophylaxis.
IMPETUS FOR QUALITY IMPROVEMENT IN VTE
A new seriousness about VTE quality measures
The 8th edition of the ACCP guidelines recommends that every hospital develop a formal, active strategy to consistently identify medical and surgical patients at risk for VTE and to prevent VTE occurrence.1 Although prior editions of the ACCP guidelines have made this recommendation for more than 2 decades, fewer than 1 in 10 acute care hospitals had any such strategy in place as recently as 5 years ago. Now, however, most US hospitals have implemented such a strategy, thanks to the growing national emphasis on health care quality measurement in recent years.
The Surgical Care Improvement Project (SCIP) has been at the forefront of this recent quality measures movement. SCIP, a joint project of the American Medical Association and federal government agencies, set a goal to reduce surgical complications in the United States by 25% from 2005 to 2010.2 Two SCIP process measures relate to improving VTE prophylaxis2,3:
- The proportion of surgical patients for whom recommended VTE prophylaxis is ordered
- The proportion of surgical patients who actually receive appropriate VTE prophylaxis within 24 hours before or after surgery.
The Joint Commission and the National Quality Forum recently endorsed these two SCIP performance measures for perioperative VTE prophylaxis along with several others relating to VTE treatment.
CMS raises the stakes with reimbursement restrictions
More significantly, the federal government’s Centers for Medicare and Medicaid Services (CMS) will soon refuse to reimburse for hospital treatment of a primary diagnosis of DVT or pulmonary embolism (PE) following recent (within 30 days) total hip or knee arthroplasty. Effective October 1, 2009, a primary VTE diagnosis following these joint replacement procedures will be added to CMS’ current list of “never events,” or hospital-acquired conditions for which CMS will not provide reimbursement because they are considered the result of preventable medical errors. (Notably, treatment of DVT or PE as a secondary diagnosis will still be reimbursed—for example, if a joint replacement patient develops nosocomial pneumonia, is transferred to the intensive care unit, and then develops VTE.) This addition of DVT and PE to the list is highly controversial since these events sometimes develop even if prophylactic therapy is appropriate and aggressive.
Strategies to promote best practices
In the update for the new 8th edition of its guidelines, the ACCP added recommendations on specific ways for hospitals to identify patients at high risk for VTE and ensure that they receive appropriate prophylaxis. These include the use of computer decision-support systems, preprinted orders, and periodic audit and feedback.1
Researchers at Brigham and Women’s Hospital evaluated the effectiveness of a computer alert system for notifying physicians of newly hospitalized patients at risk for DVT who were not receiving prevention therapy within the first 24 hours of hospital admission.4 These patients presumably “fell through the cracks” and warranted prophylaxis but were otherwise not recognized by the health care team. Risk was determined by a scoring system based on multiple variables, including malignancy, previous DVT or PE, hypercoagulability, major surgery, advanced age, obesity, ordered bed rest, and treatment with hormone replacement therapy or oral contraceptives. Study physicians had to acknowledge having received the alert but could choose whether or not to order VTE prophylaxis. Prophylaxis was used in considerably more patients from the intervention group than from a control group of high-risk patients whose physicians did not receive alerts (34% vs 14%, respectively); accordingly, the risk of a symptomatic DVT or PE event at 90 days was reduced by 41% in the intervention group.
Despite this evidence of improved practice under the alert system, the study begs the question of why the percentage of patients at risk for VTE who were given prophylaxis was still so low (34%), demonstrating how much progress in improving practice remains to be achieved.
PROPHYLAXIS STRATEGIES: MATCHING THERAPY TO RISK
NONPHARMACOLOGIC PROPHYLAXIS STRATEGIES
Does ambulation prevent DVT?
Although it is commonly accepted that walking prevents DVT, this has never been directly tested. Walking may simply be a marker of health, and healthy people are less prone to develop thromboses. We have almost no evidence to show that forcing an unhealthy person to walk helps prevent DVT. Early ambulation offers many benefits and should be encouraged, but it should not be considered DVT prophylaxis; it is simply good hospital care.
Mechanical devices: Adherence is key
Amaragiri and Lees conducted a systematic literature review of randomized controlled trials evaluating the effectiveness of graduated compression stockings (elastic stockings) for preventing DVT in various groups of hospitalized patients.6 The analysis demonstrated a statistically significant reduction in DVT incidence with graduated compression stockings compared with control both among the nine trials in which stockings were used alone (odds ratio = 0.34) and among the seven trials in which stockings were used in addition to another method of thromboprophylaxis (odds ratio = 0.24). Although benefit was demonstrated, many of the trials in this review involved patients undergoing gynecologic surgery and date from the 1970s and 1980s (when obesity was less prevalent), so the applicability of their results today may be limited.
The 8th edition of the ACCP guidelines recommends that mechanical methods of VTE prophylaxis be used primarily in patients who are at high risk of bleeding and that careful attention be directed to ensuring their proper use and optimal adherence.1 The latter point about adherence cannot be emphasized enough, as graduated compression stockings and other mechanical devices have been shown not to be effective unless they are worn at least 18 to 20 hours a day. This degree of adherence is difficult to achieve, as it can severely limit patient mobility and leave patients susceptible to development of pressure ulcers.
Mechanical compression should be initiated prior to induction of anesthesia and continue intraoperatively and then into the postanesthesia care unit. Orders for use of mechanical devices should include instructions in the patient’s medical chart specifying how—and for how many hours per day—they are to be worn. Not doing so leaves the physician vulnerable to litigation, especially as the ACCP guidelines include language on optimal adherence to these devices (“they should be removed for only a short time each day when the patient is actually walking or for bathing”1).
Continuous external compression therapy
Newer mechanical device options include a continuous external compression therapy system that allows patients to be mobile while wearing it and provides rhythmic compression that results in good peak venous flows. Ideally such a device could be put on the patient preoperatively and worn during surgery, throughout the hospital stay, and even at home during recovery. Anecdotally, however, I see patients turn these new devices off at the side of the bed just as often as they do with traditional devices.
Vena caval interruption
Vena caval interruption involves placement of a retrievable vena cava filter before surgery and removal some time later; it offers the potential for VTE prophylaxis in patients who could not tolerate even minor amounts of bleeding, such as certain trauma patients. The Eastern Association for the Surgery of Trauma has put forth a consensus recommendation to consider vena caval interruption in high-risk trauma patients who cannot receive pharmacologic prophylaxis.7 A randomized trial evaluating the usefulness of vena caval interruption for patients undergoing surgery is needed. For now, this intervention should be regarded as experimental and considered only on a highly individualized basis.
PHARMACOLOGIC PROPHYLAXIS
Timing of initiation
Pharmacologic VTE prophylaxis generally should begin 8 to 24 hours postoperatively. Of course, adequate hemostasis is required before initiation, and the net risk/benefit tradeoff with regard to timing of anticoagulant initiation has still not been well studied in many surgical patient populations.
Extended prophylaxis
In the update for the 8th edition of its guidelines, the ACCP added an explicit recommendation for extended outpatient prophylaxis with low-molecular-weight heparin (LMWH) for up to 28 days postoperatively in selected high-risk patients undergoing general or gynecologic surgery, including those with cancer or a history of VTE.1 This recommendation was based largely on studies of extended prophylaxis in patients with cancer undergoing colorectal surgery.8
Increased appreciation of the value of extended VTE prophylaxis after discharge is linked to a growing recognition that DVT and PE episodes in the community setting are often related to a recent hospital stay for either medical illness or surgery. A population-based study found that 59% of all community cases of a first lifetime VTE event in residents of Olmsted County, Minn., over a 15-year period could be linked to current or recent (< 30 days) hospitalization or nursing home residence.9 A similar population-based study in the Worcester, Mass., area found that three-fourths of all VTE events in a 3-year period occurred in the outpatient setting.10 Among patients with these outpatient VTE events, a large proportion had undergone surgery (23%) or hospitalization (37%) in the prior 3 months; among those, 67% experienced their VTE within 1 month of their time in the hospital.
These findings are no surprise, since surgery induces a hypercoagulable state that, when combined with individual risk factors such as obesity, old age, or poor heart function, cannot be assumed to return to baseline on postoperative day 4 or 5 just because the patient is being discharged.
Orthopedic surgery
For patients undergoing major orthopedic procedures, the ACCP guidelines recommend against routine screening for VTE with Doppler ultrasonography before discharge if the patient is asymptomatic.1 Such screening is not considered cost-effective because asymptomatic clots often are found, for which treatment is uncertain, and proximal clots may be missed, giving a false sense of security.
New to the ACCP guidelines in the 8th edition is the recommendation that patients undergoing knee arthroscopy who have risk factors for VTE (or whose procedure is complicated) should receive 1 week of prophylaxis with LMWH.1 Also new are recommendations for patients with risk factors undergoing single- or multilevel laminectomy (Table 4).
Recommendations unchanged in neurosurgery, spinal injury, trauma, burns
Recommendations for neurosurgery remain unchanged from the prior (2004) edition of the ACCP guidelines and are still based on the 2000 meta-analysis by Iorio and Agnelli of LMWH prophylaxis in neurosurgery cases.11 In the United States, the standard is overwhelmingly to use mechanical devices for thromboprophylaxis in neurosurgery, even for patients with cancer.
For prophylaxis in surgical patients with spinal cord injury, multisystem trauma, or burns, LMWH is predominantly used, and the ACCP recommendations are unchanged from 2004.
Drug-specific considerations
LMWH vs vitamin K antagonist. Although vitamin K antagonists (warfarin) still appear in the latest ACCP recommendations,1 LMWH is preferable. A 2004 meta-analysis of studies comparing vitamin K antagonists with LMWH for prophylaxis in patients undergoing orthopedic surgery found that vitamin K antagonists were associated with more episodes of total DVT (relative risk [RR] = 1.51; 95% CI, 1.27–1.79) and proximal DVT (RR = 1.51; 95% CI, 1.04–2.17) compared with LMWH.12 No difference was found in rates of wound hematoma or major bleeding. This finding of inferiority for vitamin K antagonists came despite the likelihood that warfarin was more often administered correctly (ie, with dose adjustment to achieve an international normalized ratio [INR] of 2.0 to 3.0 within 72 hours after surgery) in the studies in this analysis than it is in real-world practice.
Fondaparinux. The indirect factor Xa–specific inhibitor fondaparinux has had a surprisingly limited clinical adoption despite having been widely studied and found to be safe and effective. This is likely attributable in part to its 17-hour half-life, which raises concerns that it may take 3 days for its effects to stop if a patient begins to bleed. Large phase 3 studies have found fondaparinux to be equivalent to LMWH in VTE prevention after hip replacement, marginally superior to LMWH after knee replacement, and superior to LMWH following hip fracture repair.13 Fondaparinux was associated with an increase in bleeding events and instances of transfusion requirement, but only in one of the studies, which was in the setting of knee replacement surgery.14
Aspirin not recommended by ACCP. Although aspirin reduces the risk of VTE, practice guidelines from both the ACCP1 and the International Union of Angiology15 contain no recommendation for its use as prophylaxis because it is considered less effective and more risky than other therapies. In contrast, clinical practice guidelines from the American Academy of Orthopaedic Surgeons suggest that aspirin is reasonable for VTE prophylaxis.16 The varying recommendations reflect differences in perspective among these different specialties.
Aspirin has the advantages of ease of use and low cost, but it is clearly not the best evidence-based approach for VTE prophylaxis. The only recent randomized trial evidence in support of aspirin comes from the Pulmonary Embolism Prevention trial, a study with a flawed design involving more than 13,000 patients undergoing surgery for hip fracture or elective arthroplasty in five countries.17 Patients were randomized to receive aspirin 160 mg daily or placebo for 35 days along with any other prophylaxis deemed necessary (an important potential confounder). Aspirin was associated with an absolute reduction in symptomatic events of less than 1% relative to placebo, and no benefit was observed within the first week. The best results with aspirin were among patients with hip fracture. No benefit was shown among patients undergoing hip arthroplasty or knee arthroplasty; in those groups, both the aspirin and placebo recipients were also treated with LMWH. An absolute increase in rates of wound bleeding (0.6% increase) and gastrointestinal bleeding (1.0% increase) was observed in the aspirin group. The absolute increase in complications was greater than the absolute reduction in episodes of symptomatic DVT: for every episode of symptomatic DVT averted, one wound bleed and 10 gastrointestinal bleeds occurred.
SPECIAL PATIENT POPULATIONS
Renal impairment
The 8th edition of the ACCP guidelines recommends that renal function be kept in mind when considering LMWH, fondaparinux, and other antithrombotic drugs that are cleared by the kidneys. Fondaparinux and LMWH can bioaccumulate in patients with renal insufficiency, who have a higher risk of bleeding to begin with, thereby compounding the risk. Options for patients with renal compromise include avoiding drugs that bioaccumulate, using a lower dosage, and monitoring the drug level or anticoagulant effect.1
Fondaparinux is explicitly contraindicated in patients with low body weight (< 50 kg) or renal impairment (creatinine clearance < 30 mL/min). Renal function should be assessed periodically in any patients receiving the drug.18
I also would not use fondaparinux or LMWH in patients with rapidly changing renal function. For patients with chronic, stable renal impairment, one can reduce the dose of LMWH empirically; one LMWH, enoxaparin, has specific dosing guidelines in its package insert (one-third reduction in dose), but this option does not hold for patients with rapidly changing renal function.19
Obesity
The 8th edition of the ACCP guidelines recommends weight-based dosing of thromboprophylactic agents in obese patients. The guidelines particularly recommend that patients undergoing inpatient bariatric surgery be given higher doses of LMWH or unfractionated heparin.1,20
Frederiksen et al measured the anticoagulant effect of a single fixed dose of a LMWH (using anti-factor Xa heparin activity levels) and found that it was dependent on body weight.21 This suggests that fixed doses that are effective in normal-weight patients may have no detectable anti-coagulant effect in patients with very high body weight.
Weight-based dosing: mounting nonprospective evidence. Weight-based dosage adjustment for the morbidly obese has not been directly studied in a prospective, randomized fashion. A nonrandomized study by Scholten et al compared two regimens of enoxaparin (30 mg twice daily vs 40 mg twice daily) among 481 obese patients undergoing bariatric surgery; each regimen was used along with mechanical thromboprophylaxis.22 They found that the higher-dose regimen was associated with significantly fewer postoperative DVT complications (0.6% vs 5.4%; P < .01) without an increase in bleeding complications.
Separately, Shepherd et al used weight-adjusted doses of unfractionated heparin (started on the evening of surgery) to achieve subtherapeutic peak anti–factor Xa heparin activity levels of 0.11 to 0.25 IU/mL in a series of 700 patients undergoing laparoscopic gastric bypass surgery.23 The resulting doses were greater than those in traditional fixed-rate protocols, but rates of bleeding and VTE events were low and comparable to those reported in patients receiving standard doses.
Don’t rule out multimodal approaches. Multimodal prophylaxis can also be used in obese patients and need not be abandoned as a result of size considerations. For instance, two intermittent compression therapy devices can be pieced together with a Velcro binder if a single device is too small to be worn.
EMERGING ANTICOAGULANT OPTIONS
For many years, unfractionated heparin was the only available parenteral anticoagulant. While heparin has broad anticoagulant properties, it also has well-established limitations, including the need for parenteral delivery, recent problems related to contamination (it is derived from pig intestines), and of course heparin-induced thrombocytopenia (HIT). HIT is an immune-mediated form of platelet activation that can lead to widespread thrombosis throughout the body. It is more commonly associated with venous thrombosis, but arterial events with limb-threatening ischemia may also occur. LMWH is associated with a reduced risk of HIT, but LMWH does not avoid the risk entirely.
Beyond the issue of avoiding HIT, newer anticoagulant therapies are being developed with the aim of oral administration and more targeted inhibition of coagulation factors IIa (thrombin) and Xa.24
Oral direct thrombin inhibitors
One of the two most promising classes of emerging anticoagulants is the direct thrombin inhibitors, most of which are being developed for oral administration. There were high hopes for the initial compound in this class, ximelagatran, but it was abandoned about 5 years ago because of hepatotoxicity.
Dabigatran is the direct thrombin inhibitor furthest along in development today. Currently approved in Europe for prevention of VTE in patients undergoing total hip or knee replacement surgery, dabigatran is likely to be available soon in the United States. It is administered orally, has a rapid onset of action (< 1 hour), and has a predictable anticoagulant response that requires no monitoring.24 Because dabigatran is excreted essentially unchanged by the kidneys and may bioaccumulate, it should not be used in patients with renal impairment or rapidly changing renal function.
In phase 3 clinical trials for VTE prevention in knee replacement surgery, dabigatran was at least as effective as enoxaparin 40 mg once daily and had a comparable safety profile,25 but it was slightly less effective than enoxaparin 30 mg twice daily.26 In a phase 3 trial in patients undergoing hip replacement surgery, dabigatran was equivalent in efficacy and safety to enoxaparin 40 mg once daily.27
Oral direct factor Xa inhibitors
A key rationale for direct inhibition of factor Xa is that it results in inhibition of thrombin production on the activated platelet. Whereas fondaparinux is an indirect inhibitor of factor Xa, direct factor Xa inhibitors offer an advantage in that they inhibit factor Xa within the prothrombinase complex, which occurs on the surface of a platelet and is the main site for thrombin development (very little thrombin is actually produced on endothelial cells). Recall the adage that “thrombin begets more thrombin”: it activates not only platelets but the intrinsic and extrinsic pathways.28
Factor Xa may be a better target than thrombin for a number of other reasons:
- Factor Xa is believed to have few functions (compared with thrombin) outside of coagulation
- In vitro studies show that factor Xa has a wider therapeutic window than thrombin, which translates to greater separation between drug levels that will confer efficacy and bleeding
- Thrombin inhibitors are associated with rebound thrombin generation (there is no evidence of this with factor Xa inhibitors)
- The efficacy of heparin-based anticoagulants improves as selectivity for factor Xa increases (unfractionated heparin is less effective than LMWH, which is less effective than fondaparinux).
Two direct factor Xa inhibitors—both administered orally—are far along in development, as detailed below.
Apixaban has shown promise, but the phase 3 ADVANCE-1 study of apixaban for VTE prevention in patients undergoing knee surgery did not meet statistical criteria for noninferiority compared with enoxaparin 30 mg twice daily.29 This prompted a delay in regulatory filings for apixaban in the United States, and the drug’s prospects for approval for VTE prevention may be unclear until release of results from two other comparative phase 3 trials with enoxaparin in 2009 and 2010.
Rivaroxaban is more likely to become clinically available soon, in light of recent results from the phase 3 RECORD4 trial demonstrating that it was significantly superior to enoxaparin 30 mg twice daily in preventing VTE following knee replacement surgery with comparable rates of major bleeding.30
DISCUSSION
Question from the audience: Some surgeons in my hospital prescribe warfarin immediately after surgery without a bridge of LMWH. Is that appropriate?
Dr. Michota: Warfarin is an option for prophylaxis in orthopedic surgery, beginning on the day of surgery. It could even be started the day before surgery, but the dose should be monitored to achieve an INR between 2.0 and 3.0 within 72 hours of the procedure. If the INR is not in this optimum range, prophylactic doses of LMWH can be given until it is therapeutic.
Follow-up question: In practice, do you actually encourage INR monitoring? Usually we just put patients on a certain dose without monitoring. When we do check the INR, it’s usually 1.4 or 1.5.
Dr. Michota: Warfarin was shown to be effective in reducing VTE risk in orthopedic surgery with dose adjustment based on INR monitoring. On that basis, warfarin remains in the guideline recommendations. Unmonitored, warfarin has not been shown to reduce risk, so to give it that way would not be evidence-based.
Question from the audience: I work with several plastic surgeons who use compression stockings intraoperatively because they’ve heard of several patients who developed a PE during surgery. Is there any benefit to using compression stockings for 2 to 3 hours and then sending the patient home?
Dr. Michota: I don’t know. Theoretically, a device that is on and working before induction may reduce stasis.
The plastic surgery societies do have guidelines. Risk depends on the type of plastic surgery procedure; for example, risk probably increases due to inflammation in procedures that involve scraping the fat pads.
This is an area where we don’t have much data. These patients may be at risk, but we don’t know the best way to mitigate it. It is important that risks be discussed with patients in the informed-consent process and be documented. If the surgeon thinks it is reasonable to give pharmacologic prophylaxis after surgery, I wouldn’t hesitate to do that, but any form of bleeding in the setting of plastic surgery is catastrophic because it defeats the reason for which the surgery was done in the first place.
Question from the audience: How do the guidelines address being aggressive with pharmacologic thromboprophylaxis when a patient is already taking dual antiplatelet therapy?
Dr. Michota: For patients with an indication for VTE prophylaxis in a setting for which there is a specific strategy, the ACCP guidelines recommend that they be put on that regimen whether they are on antiplatelet agents or not. For example, consider a high-risk patient having colorectal surgery who should get unfractionated heparin or LMWH postoperatively and who is currently taking clopidogrel and aspirin. There is no evidence that the dual aspirin–clopidogrel therapy alone is effective in decreasing the risk of DVT. However, we do know that if we add on additional agents, the risk of bleeding is increased. The guidelines consider risk and benefit, and they recommend adding the agents that we know work to prevent DVT.
Question from the audience: You briefly mentioned prophylaxis for knee arthroscopy, which is the most frequently performed orthopedic procedure. Do these recommendations apply to all patients undergoing knee arthroscopy?
Dr. Michota: No. Prophylaxis is indicated only for patients with what the ACCP considers to be additional risk factors for thrombosis. They didn’t specify which risk factors, but good indications for prophylaxis would include morbid obesity, limited mobility after the procedure, a personal history of DVT, features of stasis noted on physical examination, stasis dermatitis (or other features that could indicate prior thrombosis), advanced age, and malignancy. If a patient undergoing knee arthroscopy has other nonmodifiable risk factors, you should also think about prophylaxis. But the vast majority of patients do not need it.
Question from the audience: I’m an academic hospitalist who works closely with orthopedic surgeons. Certain surgeons will only use aspirin for prophylaxis, and it is nonnegotiable. Where does that leave me from a medicolegal standpoint? Our model is to follow ACCP recommendations, but these orthopedic surgeons still use only aspirin.
Dr. Michota: You must do everything you can to come to a consensus with your surgeon colleagues. If you are uncomfortable, as a group you must say to the surgeons, “We are uncomfortable. This is how we view the data. How do you view the data?” If they answer, “We’re doing it because it’s easy, and the American Academy of Orthopaedic Surgeons says we can do it,” I don’t have a good response. But it is more likely that their use of aspirin is based on their own observations; they may not see many clots. Of course, the problem with observational data is that the numbers are not large and they are not generated in a randomized and prospective fashion. Perhaps you can come to some middle ground, but you could always make the difficult choice and say, “I’m just not going to follow your patients.”
- Geerts WH, Bergqvist D, Pineo GF, et al; American College of Chest Physicians. Prevention of venous thromboembolism: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest 2008; 133:381S–453S.
- Medicare Quality Improvement Community (MedQIC) Web site. http://www.medqic.org. Accessed June 1, 2009.
- Surgical Care Improvement Project (SCIP). Colorado Foundation for Medical Care Web site. http://www.cfmc.org/hospital/hospital_scip.htm. Accessed June 1, 2009.
- Kucher N, Koo S, Quiroz R, et al. Electronic alerts to prevent venous thromboembolism among hospitalized patients. N Engl J Med 2005; 352:969–977.
- Anderson FA Jr, Spencer FA. Risk factors for venous thromboembolism. Circulation 2003; 107:I-9–I-16.
- Amaragiri SV, Lees TA. Elastic compression stockings for prevention of deep vein thrombosis. Cochrane Database Syst Rev 2000; (3):CD001484.
- Rogers FB, Cipolle MD, Velmahos G, et al. Practice management guidelines for the prevention of venous thromboembolism in trauma patients: the EAST Practice Management Work Group. J Trauma 2002; 53:142–164.
- Bergqvist D, Agnelli G, Cohen AT, et al. Duration of prophylaxis against venous thromboembolism with enoxaparin after surgery for cancer. N Engl J Med 2002; 346:975–980.
- Heit JA, O’Fallon WM, Petterson TM, et al. Relative impact of risk factors for deep vein thrombosis and pulmonary embolism: a population-based study. Arch Intern Med 2002; 162:1245–1248.
- Spencer FA, Lessard D, Emery C, et al. Venous thromboembolism in the outpatient setting. Arch Intern Med 2007; 167:1471–1475.
- Iorio A, Agnelli G. Low-molecular-weight and unfractionated heparin for prevention of venous thromboembolism in neurosurgery: a meta-analysis. Arch Intern Med 2000; 160:2327–2332.
- Mismetti P, Laporte S, Zufferey P, et al. Prevention of venous thromboembolism in orthopedic surgery with vitamin K antagonists: a meta-analysis. J Thromb Haemost 2004; 2:1058–1070.
- Turpie AG, Bauer KA, Eriksson BI, Lassen MR. Fondaparinux vs enoxaparin for the prevention of venous thromboembolism in major orthopedic surgery: a meta-analysis of 4 randomized double-blind studies. Arch Intern Med 2002; 162:1833–1840.
- Bauer KA, Eriksson BI, Lassen MR, Turpie AG; Steering Committee of the Pentasaccharide in Major Knee Surgery Study. Fondaparinux compared with enoxaparin for the prevention of venous thromboembolism after elective major knee surgery. N Engl J Med 2001; 345:1305–1310.
- Cardiovascular Disease Educational and Research Trust; Cyprus Cardiovascular Disease Educational and Research Trust; European Venous Forum; International Surgical Thrombosis Forum; International Union of Angiology; Union Internationale de Phlébologie. Prevention and treatment of venous thromboembolism. International Consensus Statement (guidelines according to scientific evidence). Int Angiol 2006; 25:101–161.
- American Academy of Orthopaedic Surgeons clinical guideline on prevention of symptomatic pulmonary embolism in patients undergoing total hip or knee arthroplasty. http://www.aaos.org/research/guidelines/PE_summary.pdf. Accessed June 5, 2009.
- Prevention of pulmonary embolism and deep vein thrombosis with low dose aspirin: Pulmonary Embolism Prevention (PEP) trial. Lancet 2000; 355:1295-1302.
- Arixtra injection [package insert]. Research Triangle Park, NC: GlaxoSmithKline; 2008.
- Sanderink GJ, Guimart C, Jariwala N, et al. Enoxaparin pharmacokinetics and pharmacodynamics in renal impairment. J Am Coll Cardiol 2001; 37(suppl A):229A. Abstract.
- Hirsh J, Bauer KA, Donati MB, et al; American College of Chest Physicians. Parenteral anticoagulants: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition) [published correction appears in Chest 2008; 134:473]. Chest 2008; 133:141S–159S.
- Frederiksen SG, Hedenbro JL, Norgren L. Enoxaparin effect depends on body-weight and current doses may be inadequate in obese patients. Br J Surg 2003; 90:547–548.
- Scholten DJ, Hoedema RM, Scholten SE. A comparison of two different prophylactic dose regimens of low molecular weight heparin in bariatric surgery. Obes Surg 2002; 12:19–24.
- Shepherd MF, Rosborough TK, Schwartz ML. Heparin thromboprophylaxis in gastric bypass surgery. Obes Surg 2003; 13:249–253.
- Weitz JI, Bates SM. New anticoagulants. J Thromb Haemost 2005; 3:1843–1853.
- Eriksson BI, Dahl OE, Rosencher N, et al. Oral dabigatran etexilate vs. subcutaneous enoxaparin for the prevention of venous thromboembolism after total knee replacement: the RE-MODEL randomized trial. J Thromb Haemost 2007; 5:2178–2185.
- RE-MOBILIZE Writing Committee, Ginsberg JS, Davidson BL, et al. Oral thrombin inhibitor dabigatran etexilate vs North American enoxaparin regimen for prevention of venous thromboembolism after knee arthroplasty surgery. J Arthroplasty 2009; 24:1–9.
- Eriksson BI, Dahl OE, Rosencher N, et al; RE-NOVATE Study Group. Dabigatran etexilate versus enoxaparin for prevention of venous thromboembolism after total hip replacement: a randomised, double-blind, non-inferiority trial [published correction appears in Lancet 2007; 370:2004]. Lancet 2007: 370:949–956.
- Hoffman M, Monroe DM 3rd, Roberts HR. Activated factor VII activates factors IX and X on the surface of activated platelets: thoughts on the mechanism of action of high-dose activated factor VII. Blood Coagul Fibrinolysis 1998; 9(suppl 1):S61–S65.
- Bristol-Myers Squibb and Pfizer provide update on apixaban clinical development program [press release]. New York, NY: August 27, 2008.
- Turpie AG, Lassen MR, Davidson BL, et al. Rivaroxaban versus enoxaparin for thromboprophylaxis after total knee arthroplasty (RECORD4): a randomised trial. Lancet 2009; 373:1673–1680.
- Geerts WH, Bergqvist D, Pineo GF, et al; American College of Chest Physicians. Prevention of venous thromboembolism: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest 2008; 133:381S–453S.
- Medicare Quality Improvement Community (MedQIC) Web site. http://www.medqic.org. Accessed June 1, 2009.
- Surgical Care Improvement Project (SCIP). Colorado Foundation for Medical Care Web site. http://www.cfmc.org/hospital/hospital_scip.htm. Accessed June 1, 2009.
- Kucher N, Koo S, Quiroz R, et al. Electronic alerts to prevent venous thromboembolism among hospitalized patients. N Engl J Med 2005; 352:969–977.
- Anderson FA Jr, Spencer FA. Risk factors for venous thromboembolism. Circulation 2003; 107:I-9–I-16.
- Amaragiri SV, Lees TA. Elastic compression stockings for prevention of deep vein thrombosis. Cochrane Database Syst Rev 2000; (3):CD001484.
- Rogers FB, Cipolle MD, Velmahos G, et al. Practice management guidelines for the prevention of venous thromboembolism in trauma patients: the EAST Practice Management Work Group. J Trauma 2002; 53:142–164.
- Bergqvist D, Agnelli G, Cohen AT, et al. Duration of prophylaxis against venous thromboembolism with enoxaparin after surgery for cancer. N Engl J Med 2002; 346:975–980.
- Heit JA, O’Fallon WM, Petterson TM, et al. Relative impact of risk factors for deep vein thrombosis and pulmonary embolism: a population-based study. Arch Intern Med 2002; 162:1245–1248.
- Spencer FA, Lessard D, Emery C, et al. Venous thromboembolism in the outpatient setting. Arch Intern Med 2007; 167:1471–1475.
- Iorio A, Agnelli G. Low-molecular-weight and unfractionated heparin for prevention of venous thromboembolism in neurosurgery: a meta-analysis. Arch Intern Med 2000; 160:2327–2332.
- Mismetti P, Laporte S, Zufferey P, et al. Prevention of venous thromboembolism in orthopedic surgery with vitamin K antagonists: a meta-analysis. J Thromb Haemost 2004; 2:1058–1070.
- Turpie AG, Bauer KA, Eriksson BI, Lassen MR. Fondaparinux vs enoxaparin for the prevention of venous thromboembolism in major orthopedic surgery: a meta-analysis of 4 randomized double-blind studies. Arch Intern Med 2002; 162:1833–1840.
- Bauer KA, Eriksson BI, Lassen MR, Turpie AG; Steering Committee of the Pentasaccharide in Major Knee Surgery Study. Fondaparinux compared with enoxaparin for the prevention of venous thromboembolism after elective major knee surgery. N Engl J Med 2001; 345:1305–1310.
- Cardiovascular Disease Educational and Research Trust; Cyprus Cardiovascular Disease Educational and Research Trust; European Venous Forum; International Surgical Thrombosis Forum; International Union of Angiology; Union Internationale de Phlébologie. Prevention and treatment of venous thromboembolism. International Consensus Statement (guidelines according to scientific evidence). Int Angiol 2006; 25:101–161.
- American Academy of Orthopaedic Surgeons clinical guideline on prevention of symptomatic pulmonary embolism in patients undergoing total hip or knee arthroplasty. http://www.aaos.org/research/guidelines/PE_summary.pdf. Accessed June 5, 2009.
- Prevention of pulmonary embolism and deep vein thrombosis with low dose aspirin: Pulmonary Embolism Prevention (PEP) trial. Lancet 2000; 355:1295-1302.
- Arixtra injection [package insert]. Research Triangle Park, NC: GlaxoSmithKline; 2008.
- Sanderink GJ, Guimart C, Jariwala N, et al. Enoxaparin pharmacokinetics and pharmacodynamics in renal impairment. J Am Coll Cardiol 2001; 37(suppl A):229A. Abstract.
- Hirsh J, Bauer KA, Donati MB, et al; American College of Chest Physicians. Parenteral anticoagulants: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition) [published correction appears in Chest 2008; 134:473]. Chest 2008; 133:141S–159S.
- Frederiksen SG, Hedenbro JL, Norgren L. Enoxaparin effect depends on body-weight and current doses may be inadequate in obese patients. Br J Surg 2003; 90:547–548.
- Scholten DJ, Hoedema RM, Scholten SE. A comparison of two different prophylactic dose regimens of low molecular weight heparin in bariatric surgery. Obes Surg 2002; 12:19–24.
- Shepherd MF, Rosborough TK, Schwartz ML. Heparin thromboprophylaxis in gastric bypass surgery. Obes Surg 2003; 13:249–253.
- Weitz JI, Bates SM. New anticoagulants. J Thromb Haemost 2005; 3:1843–1853.
- Eriksson BI, Dahl OE, Rosencher N, et al. Oral dabigatran etexilate vs. subcutaneous enoxaparin for the prevention of venous thromboembolism after total knee replacement: the RE-MODEL randomized trial. J Thromb Haemost 2007; 5:2178–2185.
- RE-MOBILIZE Writing Committee, Ginsberg JS, Davidson BL, et al. Oral thrombin inhibitor dabigatran etexilate vs North American enoxaparin regimen for prevention of venous thromboembolism after knee arthroplasty surgery. J Arthroplasty 2009; 24:1–9.
- Eriksson BI, Dahl OE, Rosencher N, et al; RE-NOVATE Study Group. Dabigatran etexilate versus enoxaparin for prevention of venous thromboembolism after total hip replacement: a randomised, double-blind, non-inferiority trial [published correction appears in Lancet 2007; 370:2004]. Lancet 2007: 370:949–956.
- Hoffman M, Monroe DM 3rd, Roberts HR. Activated factor VII activates factors IX and X on the surface of activated platelets: thoughts on the mechanism of action of high-dose activated factor VII. Blood Coagul Fibrinolysis 1998; 9(suppl 1):S61–S65.
- Bristol-Myers Squibb and Pfizer provide update on apixaban clinical development program [press release]. New York, NY: August 27, 2008.
- Turpie AG, Lassen MR, Davidson BL, et al. Rivaroxaban versus enoxaparin for thromboprophylaxis after total knee arthroplasty (RECORD4): a randomised trial. Lancet 2009; 373:1673–1680.
KEY POINTS
- Effective October 1, 2009, the Centers for Medicare and Medicaid Services is refusing to reimburse for hospital treatment of a primary diagnosis of deep vein thrombosis or pulmonary embolism following recent (within 30 days) hip or knee replacement surgery.
- Mechanical methods of thromboprophylaxis are not effective unless used for at least 18 to 20 hours a day.
- The latest ACCP guidelines recommend extended pharmacologic VTE prophylaxis for up to 28 days in select high-risk patients undergoing general or gynecologic surgery. Extended prophylaxis of varying duration is recommended for patients undergoing major orthopedic procedures.
- Aspirin alone is not recommended for perioperative VTE prophylaxis in any patient group by the ACCP or the International Union of Angiology.
- Patients with renal impairment have fewer anticoagulant options and may require dose adjustment. Weight-based dosing appears to be safe and effective for obese surgical patients.
- New selective and orally administered direct thrombin inhibitors and oral direct factor Xa inhibitors may soon be available for perioperative VTE prophylaxis.