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

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

Regularizing the approach to the irregularly irregular

Article Type
Changed
Tue, 10/03/2017 - 15:12
Display Headline
Regularizing the approach to the irregularly irregular

Atrial fibrillation is the most common chronic rapid arrhythmia requiring the attention of internists and cardiologists. Patients with this arrhythmia have higher rates of morbidity and death than similar patients with normal sinus rhythm, and they do so for a number of reasons.

Patients with atrial fibrillation have a slew of comorbidities, including hypertensive and ischemic heart disease. Patients undergoing cardiac surgery have a dramatically higher risk of a postoperative bout of atrial fibrillation. The main concerns are the risk of stroke and the symptoms of heart failure and fatigue (often with exercise intolerance).

Information from registries of patients with atrial fibrillation has permitted the development of prognosticators of stroke risk. The CHADS2 score (congestive heart failure, hypertension, age > 75, diabetes, and prior stroke or transient ischemic attack) is an amazingly simple way to identify patients with atrial fibrillation who are at highest risk of stroke. This in turn has allowed stratification of patients for entrance into various anticoagulation studies. And perhaps surprisingly, when many factors are considered, nothing turns out to be dramatically better than warfarin (Coumadin)—if the international normalized ratio (INR) can be appropriately controlled.

Not many options are available to prevent atrial fibrillation. Postoperative atrial fibrillation may be prevented with high-dose steroids or colchicine (Colcrys), but this is often a self-limited, situational event. Chronic or recurrent intermittent atrial fibrillation is not readily prevented in most patients, and many symptomatic patients, as discussed by Dr. Bruce Lindsay in this issue, may benefit from drug therapy or radiofrequency ablation.

Studies suggest that trying to convert atrial fibrillation to normal sinus rhythm (vs controlling the rate) may not be worth the effort and the risk in many patients with asymptomatic atrial fibrillation. Furthermore, in patients with symptomatic atrial fibrillation, determining the cause of symptoms is difficult. For example, it may not always be easily determined if fatigue in an elderly patient with chronic atrial fibrillation is due to mild rate-related congestive heart failure, decreased left ventricular output due to the loss of the atrial “kick,” chronic ischemia, or the sedating effect of a beta-blocker given in an effort to control the tachycardia.

Despite many large, well-done studies comparing antiarrhythmic drugs, ablation techniques, and anticoagulants, patients will still benefit most from an experienced clinician’s reflective, individualized assessment before embarking on algorithm-driven long-term therapy. We have more choices, more data, and more management algorithms, but there is still no panacea for patients with atrial fibrillation.

Article PDF
Author and Disclosure Information

Brian F. Mandell, MD, PhD
Editor in Chief

Issue
Cleveland Clinic Journal of Medicine - 79(8)
Publications
Topics
Page Number
522
Sections
Author and Disclosure Information

Brian F. Mandell, MD, PhD
Editor in Chief

Author and Disclosure Information

Brian F. Mandell, MD, PhD
Editor in Chief

Article PDF
Article PDF

Atrial fibrillation is the most common chronic rapid arrhythmia requiring the attention of internists and cardiologists. Patients with this arrhythmia have higher rates of morbidity and death than similar patients with normal sinus rhythm, and they do so for a number of reasons.

Patients with atrial fibrillation have a slew of comorbidities, including hypertensive and ischemic heart disease. Patients undergoing cardiac surgery have a dramatically higher risk of a postoperative bout of atrial fibrillation. The main concerns are the risk of stroke and the symptoms of heart failure and fatigue (often with exercise intolerance).

Information from registries of patients with atrial fibrillation has permitted the development of prognosticators of stroke risk. The CHADS2 score (congestive heart failure, hypertension, age > 75, diabetes, and prior stroke or transient ischemic attack) is an amazingly simple way to identify patients with atrial fibrillation who are at highest risk of stroke. This in turn has allowed stratification of patients for entrance into various anticoagulation studies. And perhaps surprisingly, when many factors are considered, nothing turns out to be dramatically better than warfarin (Coumadin)—if the international normalized ratio (INR) can be appropriately controlled.

Not many options are available to prevent atrial fibrillation. Postoperative atrial fibrillation may be prevented with high-dose steroids or colchicine (Colcrys), but this is often a self-limited, situational event. Chronic or recurrent intermittent atrial fibrillation is not readily prevented in most patients, and many symptomatic patients, as discussed by Dr. Bruce Lindsay in this issue, may benefit from drug therapy or radiofrequency ablation.

Studies suggest that trying to convert atrial fibrillation to normal sinus rhythm (vs controlling the rate) may not be worth the effort and the risk in many patients with asymptomatic atrial fibrillation. Furthermore, in patients with symptomatic atrial fibrillation, determining the cause of symptoms is difficult. For example, it may not always be easily determined if fatigue in an elderly patient with chronic atrial fibrillation is due to mild rate-related congestive heart failure, decreased left ventricular output due to the loss of the atrial “kick,” chronic ischemia, or the sedating effect of a beta-blocker given in an effort to control the tachycardia.

Despite many large, well-done studies comparing antiarrhythmic drugs, ablation techniques, and anticoagulants, patients will still benefit most from an experienced clinician’s reflective, individualized assessment before embarking on algorithm-driven long-term therapy. We have more choices, more data, and more management algorithms, but there is still no panacea for patients with atrial fibrillation.

Atrial fibrillation is the most common chronic rapid arrhythmia requiring the attention of internists and cardiologists. Patients with this arrhythmia have higher rates of morbidity and death than similar patients with normal sinus rhythm, and they do so for a number of reasons.

Patients with atrial fibrillation have a slew of comorbidities, including hypertensive and ischemic heart disease. Patients undergoing cardiac surgery have a dramatically higher risk of a postoperative bout of atrial fibrillation. The main concerns are the risk of stroke and the symptoms of heart failure and fatigue (often with exercise intolerance).

Information from registries of patients with atrial fibrillation has permitted the development of prognosticators of stroke risk. The CHADS2 score (congestive heart failure, hypertension, age > 75, diabetes, and prior stroke or transient ischemic attack) is an amazingly simple way to identify patients with atrial fibrillation who are at highest risk of stroke. This in turn has allowed stratification of patients for entrance into various anticoagulation studies. And perhaps surprisingly, when many factors are considered, nothing turns out to be dramatically better than warfarin (Coumadin)—if the international normalized ratio (INR) can be appropriately controlled.

Not many options are available to prevent atrial fibrillation. Postoperative atrial fibrillation may be prevented with high-dose steroids or colchicine (Colcrys), but this is often a self-limited, situational event. Chronic or recurrent intermittent atrial fibrillation is not readily prevented in most patients, and many symptomatic patients, as discussed by Dr. Bruce Lindsay in this issue, may benefit from drug therapy or radiofrequency ablation.

Studies suggest that trying to convert atrial fibrillation to normal sinus rhythm (vs controlling the rate) may not be worth the effort and the risk in many patients with asymptomatic atrial fibrillation. Furthermore, in patients with symptomatic atrial fibrillation, determining the cause of symptoms is difficult. For example, it may not always be easily determined if fatigue in an elderly patient with chronic atrial fibrillation is due to mild rate-related congestive heart failure, decreased left ventricular output due to the loss of the atrial “kick,” chronic ischemia, or the sedating effect of a beta-blocker given in an effort to control the tachycardia.

Despite many large, well-done studies comparing antiarrhythmic drugs, ablation techniques, and anticoagulants, patients will still benefit most from an experienced clinician’s reflective, individualized assessment before embarking on algorithm-driven long-term therapy. We have more choices, more data, and more management algorithms, but there is still no panacea for patients with atrial fibrillation.

Issue
Cleveland Clinic Journal of Medicine - 79(8)
Issue
Cleveland Clinic Journal of Medicine - 79(8)
Page Number
522
Page Number
522
Publications
Publications
Topics
Article Type
Display Headline
Regularizing the approach to the irregularly irregular
Display Headline
Regularizing the approach to the irregularly irregular
Sections
Disallow All Ads
Alternative CME
Article PDF Media

Atrial fibrillation: New drugs, devices, and procedures

Article Type
Changed
Tue, 10/03/2017 - 15:21
Display Headline
Atrial fibrillation: New drugs, devices, and procedures

Although many developments have occurred in the last decade for managing atrial fibrillation, challenges remain. New and emerging alternatives to warfarin (Coumadin) for anticoagulation therapy prevent stroke marginally better and pose slightly less risk of hemorrhage, but they have important drawbacks.

The antiarrhythmic drug dronedarone (Multaq) has been found to offer only temporary benefit for persistent atrial fibrillation, and significant risks have emerged.

Radiofrequency ablation is gaining prominence, but repeat procedures are sometimes necessary.

An investigational device can be implanted via percutaneous catheter in the left atrial appendage to prevent embolization. It is too soon to know its eventual role in clinical practice.

This article reviews the results of clinical trials of these new treatments and discusses their role in clinical practice.

CHALLENGES OF ANTICOAGULATION

The main focus of managing atrial fibrillation is on alleviating symptoms, by either rate control or rhythm control. The other focus is on preventing stroke—a devastating outcome—with anticoagulation therapy.

For deciding whether to give warfarin to patients with atrial fibrillation, the six-point CHADS2 score is a crude but effective way of assessing the risk of stroke based on the following risk factors: congestive heart failure, hypertension, age 75 years or older, and diabetes (1 point each); or a history of stroke or transient ischemic attack (2 points).1 Warfarin is given if patients have a score of at least 2 points.

Warfarin has a narrow therapeutic window, with a higher risk of ischemic stroke if the international normalized ratio (INR) is less than 2.0,2 and a higher risk of intracranial hemorrhage if the INR is more than 3.0.3 Keeping the INR in the therapeutic range is difficult because of variations in diet, concurrent medications, and other factors.

The percent of time that the INR is within the therapeutic range predicts the risk of adverse events. Connolly et al4 showed that the cumulative risk of stroke, myocardial infarction, systemic embolism, or vascular death was no better with warfarin than with clopidogrel (Plavix) plus aspirin if the INR was in the therapeutic range less than 65% of the time, but the risk was significantly less if the INR was in the therapeutic range more than 65% of the time.

Also, comparing warfarin with the combination of aspirin and clopidogrel, Verheugt5 found that the rates of stroke of any kind, of disabling and fatal stroke, and of stroke per major bleed were lower in patients taking warfarin. Although many physicians prefer aspirin plus clopidogrel because of concerns about bleeding with warfarin, the rates of major bleeding were about the same in the two groups.

In a trial in patients for whom warfarin was “unsuitable,”6 the combination of aspirin plus clopidogrel was associated with a lower rate of stroke than aspirin alone (2.4% per year vs 3.3% per year, relative risk 0.762) but a higher rate of major bleeding events (2.0% per year vs 1.3% per year, relative risk 1.57).

NEW ALTERNATIVES TO WARFARIN

Because of the problems with warfarin, alternatives have been sought for many years. Several new oral anticoagulants are available or are being developed,7 including the factor Xa inhibitors rivaroxaban (Xarelto) and apixaban (Eliquis) and the direct factor II (thrombin) inhibitor dabigatran (Pradaxa) (Table 1).

Dabigatran’s advantages and drawbacks

Dabigatran has been on the market for more than a year and has gained rapid acceptance. The dosage is 150 mg twice a day, or 75 mg twice a day if renal function is impaired. Cleared by the kidneys, it has a half-life of 12 to 17 hours; 75% is cleared within 24 hours. For a patient who needs surgery that poses a low risk of bleeding, the general recommendation is to stop dabigatran the night before the surgical procedure. For operations with a greater risk of bleeding, many surgeons recommend stopping the drug 3 or 4 days before.

Advantages of dabigatran include that it is not influenced by diet and that the onset of therapeutic benefit is within 1 hour. Although some drugs affect dabigatran, drug interactions are more troublesome with warfarin.

A serious concern about dabigatran and the other new agents is that if a bleeding problem arises, the effects of these drugs are not reversible by administration of fresh frozen plasma. Dabigatran is reversible by dialysis; however, if a patient is also hypotensive, dialysis is not an option, and simply waiting for the drug to clear is the only choice.

Another drawback is that therapeutic levels cannot be monitored. If a patient taking warfarin requires cardioversion, the INR is carefully monitored for several weeks beforehand to reduce the risk of stroke. With dabigatran, there is no way to know if a patient is actually taking the drug as prescribed.

 

 

Clinical trials show that alternatives are marginally better than warfarin

In the Randomized Evaluation of Long-Term Anticoagulation Therapy (RE-LY) trial,8 dabigatran was associated with a significantly lower incidence of intracranial hemorrhage, combined strokes, and systemic embolization than warfarin. The incidence of major bleeds was slightly lower with dabigatran. Although dabigatran performed better, the differences were small and would not require patients to change from warfarin if they are already doing well.

Apixaban and rivaroxaban are other alternatives to warfarin, with different mechanisms of action and metabolism. Although rivaroxaban’s half-life is similar to that of apixaban and dabigatran, it is being marketed as allowing once-daily dosing instead of twice-daily.

Recent randomized controlled clinical trials of the new drugs include:

  • The Apixaban Versus Acetylsalicylic Acid (ASA) to Prevent Stroke in Atrial Fibrillation Patients Who Have Failed or Are Unsuitable for Vitamin K Antagonist Treatment (AVERROES) trial,9 which compared apixaban and aspirin
  • The Apixaban for Reduction in Stroke and Other Thromboembolic Events in Atrial Fibrillation (ARISTOTLE) trial comparing apixaban and warfarin10
  • The Rivaroxaban Once Daily Oral Direct Factor Xa Inhibition Compared With Vitamin K Antagonism for Prevention of Stroke and Embolism Trial in Atrial Fibrillation (ROCKET AF),11 comparing rivaroxaban and warfarin
  • RE-LY,8 comparing dabigatran and warfarin.

In the ARISTOTLE,10 ROCKET AF,11 and RE-LY trials,8 the time that the warfarin patients’ INRs were in the therapeutic range varied from 55% to 68%. This seems low and is a problem when trying to compare therapies, but is probably about as high as one can expect in the real world.

In AVERROES,9 the combined rate of stroke and embolism was higher with aspirin than with apixaban. In the other trials, the rates were slightly better with the new drugs than with warfarin, and the rates of major hemorrhage and hemorrhagic stroke were only slightly higher with warfarin than with the new drugs. Because the differences in benefits and risks are so small, the main advantage of the newer drugs will probably be for patients who have difficulty staying in the therapeutic INR range on warfarin.

RATE CONTROL VS RESTORATION OF SINUS RHYTHM

Evidence is insufficient to determine the risk of very-long-term asymptomatic atrial fibrillation in patients on appropriate anticoagulation. Rate control is an option for asymptomatic patients but provides no change in quality of life and no definitive reduction in the risk of stroke. The main argument for restoring normal sinus rhythm in patients with mild to moderate symptoms is that it improves exercise capacity. The need for anticoagulation persists when patients are converted to sinus rhythm because the risk of recurrent atrial fibrillation remains high.

For patients with symptomatic atrial fibrillation, rate control is sometimes achieved with beta-blockers or calcium channel blockers. Rate control may be augmented with the addition of digoxin, but when used alone digoxin generally does not control the rate of atrial fibrillation. However, in many cases of atrial fibrillation, symptoms are not rate-related, and cardioversion to normal sinus rhythm should be attempted. In such cases, the symptoms may be attributable to a loss of atrial transport function.

Patients with the following risk factors should be admitted to the hospital to start antiarrhythmic drugs:

  • Borderline or a long QTc interval at baseline (> 450 msec)
  • Treatment with dofetilide (Tikosyn) because of its effects on the QT interval
  • Heart failure or poor left-ventricular function
  • Sinus node dysfunction
  • Significant atrioventricular conduction disease.

Selecting an antiarrhythmic drug

Any of the antiarrhythmic drugs listed in Table 2 can be used for a patient with lone atrial fibrillation (ie, not caused by underlying heart disease). The choice of drug should be determined by whether coronary artery disease or renal failure is present as well. Liver disease or chronic obstructive pulmonary disease also may affect this decision.

Benefits of dronedarone are mixed

In a randomized trial of dronedarone vs placebo in patients with atrial fibrillation, the rate of death and the rate of first hospitalization due to a cardiovascular event at 21 months were significantly lower with dronedarone.12 No difference was found between the two groups in the rate of death from all causes, but fewer people died of cardiovascular causes in the dronedarone group. More patients taking dronedarone developed bradycardia, QT-interval prolongation, nausea, diarrhea, rash, or a higher serum creatinine level. Gastrointestinal side effects are often a problem with dronedarone: 20% to 30% of patients cannot tolerate the drug.

Dronedarone may cause a small rise in creatinine, and although this effect should be monitored, by itself it should not be interpreted as impairment of renal function. In a study in healthy people,13 dronedarone caused a 10% to 15% increase in serum creatinine, but the glomerular filtration rate was unchanged, as were renal plasma flow and anion secretion.

Another trial, in patients with severe heart failure, found that patients taking dronedarone had higher rates of hospitalization and overall mortality, raising serious concern about the safety of this drug in patients with advanced heart failure.14

Singh et al15 pooled the data from two multicenter, randomized trials that compared dronedarone with placebo for maintaining sinus rhythm in patients with atrial fibrillation or flutter. The mean time to the recurrence of atrial fibrillation was 116 days with dronedarone and 53 days with placebo. Other trials also showed longer times to recurrence and lower recurrence rates with dronedarone. Although the differences were statistically significant, they may not be clinically meaningful for patients.

Dronedarone is structurally similar to amiodarone (Cordarone), but the two drugs work differently. A meta-analysis of clinical trials16 found that amiodarone recipients had a lower rate of recurrence of atrial fibrillation than did those receiving dronedarone.

Two safety warnings for dronedarone

In January 2011, the US Food and Drug Administration (FDA) issued an alert about cases of rare but severe liver injury in patients treated with dronedarone, including two cases of acute liver failure leading to liver transplantation.17

The Permanent Atrial Fibrillation Outcome Study Using Dronedarone on Top of Standard Therapy (PALLAS)18 compared dronedarone and placebo in patients with permanent atrial fibrillation. More people died or had serious cardiovascular adverse events in the dronedarone group. The study was stopped early after data monitoring showed that rates of death, stroke, and hospitalization for heart failure were two times higher in patients receiving dronedarone. This prompted the FDA to issue another safety alert in July 2011.

Interestingly, the PALLAS study did not set out to determine whether dronedarone controls atrial fibrillation, as the study patients had long-standing, persistent atrial fibrillation. The study was designed only to determine if the drug reduces the rate of adverse events; it clearly does not, and the study shows that dronedarone should not be used to control the heart rate in patients with persistent atrial fibrillation. Instead, its use is best restricted to patients with paroxysmal atrial fibrillation without significant cardiovascular disease.

 

 

ABLATION OF ATRIAL FIBRILLATION

Another way to try to restore sinus rhythm is to destroy or isolate the area that is generating the abnormal beats via a catheter-based procedure.

Radiofrequency ablation is generally tried in patients in whom one or two drugs have failed to control atrial fibrillation. Direct comparisons show that ablation is superior to drug therapy and is effective in about 75% of patients with paroxysmal atrial fibrillation vs 20% to 40% of patients on drug therapy. Ablation plus drug therapy is often more effective than either treatment alone.

Mechanisms of atrial fibrillation and ablation

In many cases, atrial fibrillation is stimulated by vagal and sympathetic inputs to the atrium that enter around the pulmonary veins and trigger electrical activations in the area, generating spiraling, reentering circuits. Focal atrial fibrillation also originates predominantly in the pulmonary veins. Ablation of tissue widely circumscribing the mouth of the pulmonary veins prevents the electrical signal from exiting into the atrium.

In about 11% to 37% of cases, atrial fibrillation originates elsewhere, eg, in the left atrium, in the superior vena cava, or in the vein of Marshall. Techniques have evolved to also ablate these regions.

Anticoagulation therapy is recommended before the procedure, and patients at low risk should continue it for a minimum of 2 months afterward. Patients with a higher CHADS2 score should receive anticoagulation therapy for at least 1 year. The consensus statement by the Heart Rhythm Society19 recommends that patients remain on warfarin or one of the newer anticoagulants if their CHADS2 score is 2 or higher. This is because patients have a significant risk of recurrence of atrial fibrillation after radiofrequency ablation, so if their stroke risk is high they should remain on anticoagulant therapy.

Ablation is usually effective, but it carries rare but serious risks

The efficacy of a single radiofrequency ablation procedure is in the range of 60% to 80% for paroxysmal atrial fibrillation and 40% to 60% for persistent atrial fibrillation. The Second International Ablation Registry20 shows a success rate of about 75% in patients with paroxysmal atrial fibrillation and about 65% in patients with persistent and permanent atrial fibrillation. Registry data are often more favorable because reporting is optional, but these results are consistent with those from experienced medical centers. Rates of suppression of atrial fibrillation are higher in patients who also take antiarrhythmic drugs, making a “hybrid” approach useful when ablation alone fails.

According to a worldwide survey, the risk of serious complications is 4.5%. These include stroke (0.23%), tamponade (1.3%), and pulmonary vein stenosis (< 0.29%). The esophagus lies just behind the right atrium, and burning through and creating a fistula between them occurs in about 0.04% of cases and is almost uniformly fatal.20

A second ablation procedure is sometimes indicated for the recurrence of atrial fibrillation, which is almost always caused by recovery of the pulmonary veins. Bhargava et al21 found that the success rate at Cleveland Clinic for a single procedure for paroxysmal atrial fibrillation was 77%, and that it was 92% after a repeat procedure. For persistent atrial fibrillation, success rates were 76% after the first procedure and 90% after the second. Even for long-standing persistent atrial fibrillation (ie, lasting more than 1 year), 80% success was achieved after two procedures. Patients who are less likely to have a successful ablation procedure are those with long-standing atrial fibrillation and coexisting heart disease, including severe valvular disease, although mitral regurgitation sometimes improves if sinus rhythm can be maintained.

The need for a second procedure

After ablation, patients should be closely monitored for a recurrence of atrial fibrillation. Continuous monitoring with implantable cardiac monitor loop recorders can detect unrecognized episodes of arrhythmia. Long-term follow-up is also required to track outcomes and quality of life.

According to the Heart Rhythm Society Task Force on Catheter and Surgical Ablation of Atrial Fibrillation,19 atrial fibrillation recurs after ablation in about 35% to 60% of patients in the first 3 months, with recurrence rates after 1 year ranging from 5% to 16%. The rate of success is determined by the skill of the surgeon, underlying heart disease, attention to follow-up, and how success is defined.

Freedom from recurrence early on is a good predictor that late recurrence is unlikely. Patients who only have a very early recurrence (within the first 4 weeks) are more likely to have long-term freedom from atrial fibrillation tha those who have recurrences after that time.22

In a study of 831 patients, Hussein et al23 found recurrence rates of 24% between months 3 to 13 following ablation and 9% after 12 months. At 55 months, 79% were free from atrial fibrillation without drugs, 11% were free of atrial fibrillation with medications, and 5% had refractory atrial fibrillation.

Recurrence—whether early or late—was more likely to occur in people with persistent vs paroxysmal atrial fibrillation. Other risk factors for late recurrence included older age and larger left atrial size (which is also a risk factor for recurrence on drug therapy). Although recurrent arrhythmia was most often atrial fibrillation, atrial flutter also occurred frequently (in 27% of patients with late recurrence). Three patients (4% of patients with late recurrence) developed atrial tachycardia.23

In patients with early recurrence, 81% underwent repeat ablation, all of whom had recovery of one or more pulmonary veins. After the second ablation, 21% had recurrence, 65% of whom were controlled by medications.23

Whether a patient should undergo subsequent ablation procedures depends on the severity of symptoms, the likelihood of success (based on an educated guess), and the patient’s willingness to undergo another procedure.

ATRIAL APPENDAGE OCCLUSION DEVICE UNDER INVESTIGATION

New devices are being investigated that occlude the left atrial appendage to try to prevent embolization.

The Watchman device, resembling an umbrella, is implanted via a percutaneous catheter in the left atrial appendage, closing it off to preclude a thrombus from forming in the appendage and embolizing to the body. Clinical trials showed that patients who received a device had a slightly lower risk of stroke than otherwise seen in clinical practice.24 Safety and efficacy are still being determined.

The device cannot be deployed in a patient with an existing thrombus because of the danger of dislodging the thrombus, allowing it to embolize.

References
  1. Gage BF, Waterman AD, Shannon W, Boechler M, Rich MW, Radford MJ. Validation of clinical classification schemes for predicting stroke: results from the national Registry of Atrial Fibrillation. JAMA 2001; 285:28642870.
  2. Hylek EM, Skates SJ, Sheehan MA, Singer DE. An analysis of the lowest effective intensity of prophylactic anticoagulation for patients with nonrheumatic atrial fibrillation. N Engl J Med 1996; 335:540546.
  3. Hylek EM, Singer DE. Risk factors for intracranial hemorrhage in outpatients taking warfarin. Ann Intern Med 1994; 120:897902.
  4. Connolly SJ, Pogue J, Eikelboom J, et al; ACTIVE W Investigators. Benefit of oral anticoagulant over antiplatelet therapy in atrial fibrillation depends on the quality of international normalized ratio control achieved by centers and countries as measured by time in therapeutic range. Circulation 2008; 118:20292037.
  5. Verheugt FWA. Who is ineligible for warfarin in atrial fibrillation? Lancet 2009; 374:510511.
  6. ACTIVE Investigators; Connolly SJ, Pogue J, Hart RG. Effect of clopidogrel added to aspirin in patients with atrial fibrillation. N Engl J Med 2009; 360:20662078.
  7. Harenberg J. New anticoagulants in atrial fibrillation. Semin Thromb Hemost 2009; 35:574585.
  8. Connolly SJ, Ezekowitz MD, Yusuf S, et al; RE-LY Steering Committee and Investigators. Dabigatran versus warfarin patients with atrial fibrillation. N Engl J Med 2009; 361:11391151.
  9. Connolly SJ, Eikelboom J, Joyner C, et al; AVERROES Steering Committee and Investigators. Apixaban in patients with atrial fibrillation. N Engl J Med 2011; 364:806817.
  10. Granger CB, Alexander JH, McMurray JJV, et al; for the ARISTOTLE Committees and Investigators. Apixaban versus warfarin in patients with atrial fibrillation. N Engl J Med August 28, 2011; 10.1056/nejmoa1107039.
  11. Patel MR, Mahaffey KW, Garg J, et al; the ROCKET AF Steering Committee. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation. N Engl J Med 2011; 365:883891.
  12. Hohnloser SH, Crijns HJ, van Eickels M, et al; ATHENA Investigators. Effect of dronedarone on cardiovascular events in atrial fibrillation. N Engl J Med 2009; 360:688678.
  13. Tschuppert Y, Buclin T, Rothuizen LE, et al. Effect of dronedarone on renal function in healthy subjects. Br J Clin Pharmacol 2007; 64:785791.
  14. Kóber L, Torp-Pederson C, McMurray JJ, et al; Dronedarone Study Group. Increased mortality after dronedarone therapy for severe heart failure. N Engl J Med 2008; 358:26782687.
  15. Singh BN, Connolly SJ, Crijns HJ, et al; EURDIS and ADONIS Investigators. Dronedarone for maintenance of sinus rhythm in atrial fibrillation or flutter. N Engl J Med 2007; 357:987999.
  16. Piccini JP, Hasselblad V, Peterson ED, Washam JB, Califf RM. Comparative efficacy of dronedarone and amiodarone for the maintenance of sinus rhythm in patients with atrial fibrillation. J Am Coll Cardiol 2009; 54:10891095.
  17. US Food and Drug Administration. FDA drug safety communication: severe liver injury associated with the use of dronedarone (marketed as Multaq). http://www.fda.gov/drugs/drugsafety/ucm240011.htm. Accessed July 5, 2012.
  18. Connolly SJ, Camm AJ, Halperin JL, et al; for the PALLAS Investigators. Dronedarone in high-risk permanent atrial fibrillation. N Engl J Med 2011; 365:22682276.
  19. HRS/EHRA/ECAS expert consensus statement on catheter and surgical ablation of atrial fibrillation: recommendations for personnel, policy, procedures and follow-up. Heart Rhythm 2007; 4:146.
  20. Cappato R, Calkins H, Chen SA, et al. Updated worldwide survey on the methods, efficacy, and safety of catheter ablation for human atrial fibrillation. Circ Arrhythm Electrophysiol 2010; 3:3238.
  21. Bhargava M, Di Biase L, Mohanty P, et al. Impact of type of atrial fibrillation and repeat catheter ablation on long-term freedom from atrial fibrillation: results from a multicenter study. Heart Rhythm 2009; 6:14031412.
  22. Themistoclakis S, Schweikert RA, Sliba WI, et al. Clinical predictors and relationship between early and late atrial tachyarrhythmias after pulmonary vein antrum isolation. Heart Rhythm 2008; 5:679685.
  23. Hussein AA, Saliba WI, Martin DO, et al. Natural history and long-term outcomes of ablated atrial fibrillation. Circ Arrhythm Electrophysiol 2011; 4:271278.
  24. Holmes DR, Reddy VY, Turi ZG, et al; for the PROTECT AF Investigators. Percutaneous closure of the left atrial appendage versus warfarin therapy for prevention of stroke in patients with atrial fibrillation: a randomised non-inferiority trial. Lancet 2009; 374:534542.
Article PDF
Author and Disclosure Information

Bruce D. Lindsay, MD
Head, Section of Clinical Cardiac Electrophysiology, Department of Cardiovascular Medicine, Cleveland Clinic

Address: Bruce D. Lindsay, MD, Department of Cardiovascular Medicine, J2-2, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail lindsab@ccf.org

Medical Grand Rounds articles are based on edited transcripts for Medicine Grand Rounds presentations at Cleveland Clinic. They are approved by the author but are not peer-reviewed.

Issue
Cleveland Clinic Journal of Medicine - 79(8)
Publications
Topics
Page Number
553-559
Sections
Author and Disclosure Information

Bruce D. Lindsay, MD
Head, Section of Clinical Cardiac Electrophysiology, Department of Cardiovascular Medicine, Cleveland Clinic

Address: Bruce D. Lindsay, MD, Department of Cardiovascular Medicine, J2-2, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail lindsab@ccf.org

Medical Grand Rounds articles are based on edited transcripts for Medicine Grand Rounds presentations at Cleveland Clinic. They are approved by the author but are not peer-reviewed.

Author and Disclosure Information

Bruce D. Lindsay, MD
Head, Section of Clinical Cardiac Electrophysiology, Department of Cardiovascular Medicine, Cleveland Clinic

Address: Bruce D. Lindsay, MD, Department of Cardiovascular Medicine, J2-2, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail lindsab@ccf.org

Medical Grand Rounds articles are based on edited transcripts for Medicine Grand Rounds presentations at Cleveland Clinic. They are approved by the author but are not peer-reviewed.

Article PDF
Article PDF

Although many developments have occurred in the last decade for managing atrial fibrillation, challenges remain. New and emerging alternatives to warfarin (Coumadin) for anticoagulation therapy prevent stroke marginally better and pose slightly less risk of hemorrhage, but they have important drawbacks.

The antiarrhythmic drug dronedarone (Multaq) has been found to offer only temporary benefit for persistent atrial fibrillation, and significant risks have emerged.

Radiofrequency ablation is gaining prominence, but repeat procedures are sometimes necessary.

An investigational device can be implanted via percutaneous catheter in the left atrial appendage to prevent embolization. It is too soon to know its eventual role in clinical practice.

This article reviews the results of clinical trials of these new treatments and discusses their role in clinical practice.

CHALLENGES OF ANTICOAGULATION

The main focus of managing atrial fibrillation is on alleviating symptoms, by either rate control or rhythm control. The other focus is on preventing stroke—a devastating outcome—with anticoagulation therapy.

For deciding whether to give warfarin to patients with atrial fibrillation, the six-point CHADS2 score is a crude but effective way of assessing the risk of stroke based on the following risk factors: congestive heart failure, hypertension, age 75 years or older, and diabetes (1 point each); or a history of stroke or transient ischemic attack (2 points).1 Warfarin is given if patients have a score of at least 2 points.

Warfarin has a narrow therapeutic window, with a higher risk of ischemic stroke if the international normalized ratio (INR) is less than 2.0,2 and a higher risk of intracranial hemorrhage if the INR is more than 3.0.3 Keeping the INR in the therapeutic range is difficult because of variations in diet, concurrent medications, and other factors.

The percent of time that the INR is within the therapeutic range predicts the risk of adverse events. Connolly et al4 showed that the cumulative risk of stroke, myocardial infarction, systemic embolism, or vascular death was no better with warfarin than with clopidogrel (Plavix) plus aspirin if the INR was in the therapeutic range less than 65% of the time, but the risk was significantly less if the INR was in the therapeutic range more than 65% of the time.

Also, comparing warfarin with the combination of aspirin and clopidogrel, Verheugt5 found that the rates of stroke of any kind, of disabling and fatal stroke, and of stroke per major bleed were lower in patients taking warfarin. Although many physicians prefer aspirin plus clopidogrel because of concerns about bleeding with warfarin, the rates of major bleeding were about the same in the two groups.

In a trial in patients for whom warfarin was “unsuitable,”6 the combination of aspirin plus clopidogrel was associated with a lower rate of stroke than aspirin alone (2.4% per year vs 3.3% per year, relative risk 0.762) but a higher rate of major bleeding events (2.0% per year vs 1.3% per year, relative risk 1.57).

NEW ALTERNATIVES TO WARFARIN

Because of the problems with warfarin, alternatives have been sought for many years. Several new oral anticoagulants are available or are being developed,7 including the factor Xa inhibitors rivaroxaban (Xarelto) and apixaban (Eliquis) and the direct factor II (thrombin) inhibitor dabigatran (Pradaxa) (Table 1).

Dabigatran’s advantages and drawbacks

Dabigatran has been on the market for more than a year and has gained rapid acceptance. The dosage is 150 mg twice a day, or 75 mg twice a day if renal function is impaired. Cleared by the kidneys, it has a half-life of 12 to 17 hours; 75% is cleared within 24 hours. For a patient who needs surgery that poses a low risk of bleeding, the general recommendation is to stop dabigatran the night before the surgical procedure. For operations with a greater risk of bleeding, many surgeons recommend stopping the drug 3 or 4 days before.

Advantages of dabigatran include that it is not influenced by diet and that the onset of therapeutic benefit is within 1 hour. Although some drugs affect dabigatran, drug interactions are more troublesome with warfarin.

A serious concern about dabigatran and the other new agents is that if a bleeding problem arises, the effects of these drugs are not reversible by administration of fresh frozen plasma. Dabigatran is reversible by dialysis; however, if a patient is also hypotensive, dialysis is not an option, and simply waiting for the drug to clear is the only choice.

Another drawback is that therapeutic levels cannot be monitored. If a patient taking warfarin requires cardioversion, the INR is carefully monitored for several weeks beforehand to reduce the risk of stroke. With dabigatran, there is no way to know if a patient is actually taking the drug as prescribed.

 

 

Clinical trials show that alternatives are marginally better than warfarin

In the Randomized Evaluation of Long-Term Anticoagulation Therapy (RE-LY) trial,8 dabigatran was associated with a significantly lower incidence of intracranial hemorrhage, combined strokes, and systemic embolization than warfarin. The incidence of major bleeds was slightly lower with dabigatran. Although dabigatran performed better, the differences were small and would not require patients to change from warfarin if they are already doing well.

Apixaban and rivaroxaban are other alternatives to warfarin, with different mechanisms of action and metabolism. Although rivaroxaban’s half-life is similar to that of apixaban and dabigatran, it is being marketed as allowing once-daily dosing instead of twice-daily.

Recent randomized controlled clinical trials of the new drugs include:

  • The Apixaban Versus Acetylsalicylic Acid (ASA) to Prevent Stroke in Atrial Fibrillation Patients Who Have Failed or Are Unsuitable for Vitamin K Antagonist Treatment (AVERROES) trial,9 which compared apixaban and aspirin
  • The Apixaban for Reduction in Stroke and Other Thromboembolic Events in Atrial Fibrillation (ARISTOTLE) trial comparing apixaban and warfarin10
  • The Rivaroxaban Once Daily Oral Direct Factor Xa Inhibition Compared With Vitamin K Antagonism for Prevention of Stroke and Embolism Trial in Atrial Fibrillation (ROCKET AF),11 comparing rivaroxaban and warfarin
  • RE-LY,8 comparing dabigatran and warfarin.

In the ARISTOTLE,10 ROCKET AF,11 and RE-LY trials,8 the time that the warfarin patients’ INRs were in the therapeutic range varied from 55% to 68%. This seems low and is a problem when trying to compare therapies, but is probably about as high as one can expect in the real world.

In AVERROES,9 the combined rate of stroke and embolism was higher with aspirin than with apixaban. In the other trials, the rates were slightly better with the new drugs than with warfarin, and the rates of major hemorrhage and hemorrhagic stroke were only slightly higher with warfarin than with the new drugs. Because the differences in benefits and risks are so small, the main advantage of the newer drugs will probably be for patients who have difficulty staying in the therapeutic INR range on warfarin.

RATE CONTROL VS RESTORATION OF SINUS RHYTHM

Evidence is insufficient to determine the risk of very-long-term asymptomatic atrial fibrillation in patients on appropriate anticoagulation. Rate control is an option for asymptomatic patients but provides no change in quality of life and no definitive reduction in the risk of stroke. The main argument for restoring normal sinus rhythm in patients with mild to moderate symptoms is that it improves exercise capacity. The need for anticoagulation persists when patients are converted to sinus rhythm because the risk of recurrent atrial fibrillation remains high.

For patients with symptomatic atrial fibrillation, rate control is sometimes achieved with beta-blockers or calcium channel blockers. Rate control may be augmented with the addition of digoxin, but when used alone digoxin generally does not control the rate of atrial fibrillation. However, in many cases of atrial fibrillation, symptoms are not rate-related, and cardioversion to normal sinus rhythm should be attempted. In such cases, the symptoms may be attributable to a loss of atrial transport function.

Patients with the following risk factors should be admitted to the hospital to start antiarrhythmic drugs:

  • Borderline or a long QTc interval at baseline (> 450 msec)
  • Treatment with dofetilide (Tikosyn) because of its effects on the QT interval
  • Heart failure or poor left-ventricular function
  • Sinus node dysfunction
  • Significant atrioventricular conduction disease.

Selecting an antiarrhythmic drug

Any of the antiarrhythmic drugs listed in Table 2 can be used for a patient with lone atrial fibrillation (ie, not caused by underlying heart disease). The choice of drug should be determined by whether coronary artery disease or renal failure is present as well. Liver disease or chronic obstructive pulmonary disease also may affect this decision.

Benefits of dronedarone are mixed

In a randomized trial of dronedarone vs placebo in patients with atrial fibrillation, the rate of death and the rate of first hospitalization due to a cardiovascular event at 21 months were significantly lower with dronedarone.12 No difference was found between the two groups in the rate of death from all causes, but fewer people died of cardiovascular causes in the dronedarone group. More patients taking dronedarone developed bradycardia, QT-interval prolongation, nausea, diarrhea, rash, or a higher serum creatinine level. Gastrointestinal side effects are often a problem with dronedarone: 20% to 30% of patients cannot tolerate the drug.

Dronedarone may cause a small rise in creatinine, and although this effect should be monitored, by itself it should not be interpreted as impairment of renal function. In a study in healthy people,13 dronedarone caused a 10% to 15% increase in serum creatinine, but the glomerular filtration rate was unchanged, as were renal plasma flow and anion secretion.

Another trial, in patients with severe heart failure, found that patients taking dronedarone had higher rates of hospitalization and overall mortality, raising serious concern about the safety of this drug in patients with advanced heart failure.14

Singh et al15 pooled the data from two multicenter, randomized trials that compared dronedarone with placebo for maintaining sinus rhythm in patients with atrial fibrillation or flutter. The mean time to the recurrence of atrial fibrillation was 116 days with dronedarone and 53 days with placebo. Other trials also showed longer times to recurrence and lower recurrence rates with dronedarone. Although the differences were statistically significant, they may not be clinically meaningful for patients.

Dronedarone is structurally similar to amiodarone (Cordarone), but the two drugs work differently. A meta-analysis of clinical trials16 found that amiodarone recipients had a lower rate of recurrence of atrial fibrillation than did those receiving dronedarone.

Two safety warnings for dronedarone

In January 2011, the US Food and Drug Administration (FDA) issued an alert about cases of rare but severe liver injury in patients treated with dronedarone, including two cases of acute liver failure leading to liver transplantation.17

The Permanent Atrial Fibrillation Outcome Study Using Dronedarone on Top of Standard Therapy (PALLAS)18 compared dronedarone and placebo in patients with permanent atrial fibrillation. More people died or had serious cardiovascular adverse events in the dronedarone group. The study was stopped early after data monitoring showed that rates of death, stroke, and hospitalization for heart failure were two times higher in patients receiving dronedarone. This prompted the FDA to issue another safety alert in July 2011.

Interestingly, the PALLAS study did not set out to determine whether dronedarone controls atrial fibrillation, as the study patients had long-standing, persistent atrial fibrillation. The study was designed only to determine if the drug reduces the rate of adverse events; it clearly does not, and the study shows that dronedarone should not be used to control the heart rate in patients with persistent atrial fibrillation. Instead, its use is best restricted to patients with paroxysmal atrial fibrillation without significant cardiovascular disease.

 

 

ABLATION OF ATRIAL FIBRILLATION

Another way to try to restore sinus rhythm is to destroy or isolate the area that is generating the abnormal beats via a catheter-based procedure.

Radiofrequency ablation is generally tried in patients in whom one or two drugs have failed to control atrial fibrillation. Direct comparisons show that ablation is superior to drug therapy and is effective in about 75% of patients with paroxysmal atrial fibrillation vs 20% to 40% of patients on drug therapy. Ablation plus drug therapy is often more effective than either treatment alone.

Mechanisms of atrial fibrillation and ablation

In many cases, atrial fibrillation is stimulated by vagal and sympathetic inputs to the atrium that enter around the pulmonary veins and trigger electrical activations in the area, generating spiraling, reentering circuits. Focal atrial fibrillation also originates predominantly in the pulmonary veins. Ablation of tissue widely circumscribing the mouth of the pulmonary veins prevents the electrical signal from exiting into the atrium.

In about 11% to 37% of cases, atrial fibrillation originates elsewhere, eg, in the left atrium, in the superior vena cava, or in the vein of Marshall. Techniques have evolved to also ablate these regions.

Anticoagulation therapy is recommended before the procedure, and patients at low risk should continue it for a minimum of 2 months afterward. Patients with a higher CHADS2 score should receive anticoagulation therapy for at least 1 year. The consensus statement by the Heart Rhythm Society19 recommends that patients remain on warfarin or one of the newer anticoagulants if their CHADS2 score is 2 or higher. This is because patients have a significant risk of recurrence of atrial fibrillation after radiofrequency ablation, so if their stroke risk is high they should remain on anticoagulant therapy.

Ablation is usually effective, but it carries rare but serious risks

The efficacy of a single radiofrequency ablation procedure is in the range of 60% to 80% for paroxysmal atrial fibrillation and 40% to 60% for persistent atrial fibrillation. The Second International Ablation Registry20 shows a success rate of about 75% in patients with paroxysmal atrial fibrillation and about 65% in patients with persistent and permanent atrial fibrillation. Registry data are often more favorable because reporting is optional, but these results are consistent with those from experienced medical centers. Rates of suppression of atrial fibrillation are higher in patients who also take antiarrhythmic drugs, making a “hybrid” approach useful when ablation alone fails.

According to a worldwide survey, the risk of serious complications is 4.5%. These include stroke (0.23%), tamponade (1.3%), and pulmonary vein stenosis (< 0.29%). The esophagus lies just behind the right atrium, and burning through and creating a fistula between them occurs in about 0.04% of cases and is almost uniformly fatal.20

A second ablation procedure is sometimes indicated for the recurrence of atrial fibrillation, which is almost always caused by recovery of the pulmonary veins. Bhargava et al21 found that the success rate at Cleveland Clinic for a single procedure for paroxysmal atrial fibrillation was 77%, and that it was 92% after a repeat procedure. For persistent atrial fibrillation, success rates were 76% after the first procedure and 90% after the second. Even for long-standing persistent atrial fibrillation (ie, lasting more than 1 year), 80% success was achieved after two procedures. Patients who are less likely to have a successful ablation procedure are those with long-standing atrial fibrillation and coexisting heart disease, including severe valvular disease, although mitral regurgitation sometimes improves if sinus rhythm can be maintained.

The need for a second procedure

After ablation, patients should be closely monitored for a recurrence of atrial fibrillation. Continuous monitoring with implantable cardiac monitor loop recorders can detect unrecognized episodes of arrhythmia. Long-term follow-up is also required to track outcomes and quality of life.

According to the Heart Rhythm Society Task Force on Catheter and Surgical Ablation of Atrial Fibrillation,19 atrial fibrillation recurs after ablation in about 35% to 60% of patients in the first 3 months, with recurrence rates after 1 year ranging from 5% to 16%. The rate of success is determined by the skill of the surgeon, underlying heart disease, attention to follow-up, and how success is defined.

Freedom from recurrence early on is a good predictor that late recurrence is unlikely. Patients who only have a very early recurrence (within the first 4 weeks) are more likely to have long-term freedom from atrial fibrillation tha those who have recurrences after that time.22

In a study of 831 patients, Hussein et al23 found recurrence rates of 24% between months 3 to 13 following ablation and 9% after 12 months. At 55 months, 79% were free from atrial fibrillation without drugs, 11% were free of atrial fibrillation with medications, and 5% had refractory atrial fibrillation.

Recurrence—whether early or late—was more likely to occur in people with persistent vs paroxysmal atrial fibrillation. Other risk factors for late recurrence included older age and larger left atrial size (which is also a risk factor for recurrence on drug therapy). Although recurrent arrhythmia was most often atrial fibrillation, atrial flutter also occurred frequently (in 27% of patients with late recurrence). Three patients (4% of patients with late recurrence) developed atrial tachycardia.23

In patients with early recurrence, 81% underwent repeat ablation, all of whom had recovery of one or more pulmonary veins. After the second ablation, 21% had recurrence, 65% of whom were controlled by medications.23

Whether a patient should undergo subsequent ablation procedures depends on the severity of symptoms, the likelihood of success (based on an educated guess), and the patient’s willingness to undergo another procedure.

ATRIAL APPENDAGE OCCLUSION DEVICE UNDER INVESTIGATION

New devices are being investigated that occlude the left atrial appendage to try to prevent embolization.

The Watchman device, resembling an umbrella, is implanted via a percutaneous catheter in the left atrial appendage, closing it off to preclude a thrombus from forming in the appendage and embolizing to the body. Clinical trials showed that patients who received a device had a slightly lower risk of stroke than otherwise seen in clinical practice.24 Safety and efficacy are still being determined.

The device cannot be deployed in a patient with an existing thrombus because of the danger of dislodging the thrombus, allowing it to embolize.

Although many developments have occurred in the last decade for managing atrial fibrillation, challenges remain. New and emerging alternatives to warfarin (Coumadin) for anticoagulation therapy prevent stroke marginally better and pose slightly less risk of hemorrhage, but they have important drawbacks.

The antiarrhythmic drug dronedarone (Multaq) has been found to offer only temporary benefit for persistent atrial fibrillation, and significant risks have emerged.

Radiofrequency ablation is gaining prominence, but repeat procedures are sometimes necessary.

An investigational device can be implanted via percutaneous catheter in the left atrial appendage to prevent embolization. It is too soon to know its eventual role in clinical practice.

This article reviews the results of clinical trials of these new treatments and discusses their role in clinical practice.

CHALLENGES OF ANTICOAGULATION

The main focus of managing atrial fibrillation is on alleviating symptoms, by either rate control or rhythm control. The other focus is on preventing stroke—a devastating outcome—with anticoagulation therapy.

For deciding whether to give warfarin to patients with atrial fibrillation, the six-point CHADS2 score is a crude but effective way of assessing the risk of stroke based on the following risk factors: congestive heart failure, hypertension, age 75 years or older, and diabetes (1 point each); or a history of stroke or transient ischemic attack (2 points).1 Warfarin is given if patients have a score of at least 2 points.

Warfarin has a narrow therapeutic window, with a higher risk of ischemic stroke if the international normalized ratio (INR) is less than 2.0,2 and a higher risk of intracranial hemorrhage if the INR is more than 3.0.3 Keeping the INR in the therapeutic range is difficult because of variations in diet, concurrent medications, and other factors.

The percent of time that the INR is within the therapeutic range predicts the risk of adverse events. Connolly et al4 showed that the cumulative risk of stroke, myocardial infarction, systemic embolism, or vascular death was no better with warfarin than with clopidogrel (Plavix) plus aspirin if the INR was in the therapeutic range less than 65% of the time, but the risk was significantly less if the INR was in the therapeutic range more than 65% of the time.

Also, comparing warfarin with the combination of aspirin and clopidogrel, Verheugt5 found that the rates of stroke of any kind, of disabling and fatal stroke, and of stroke per major bleed were lower in patients taking warfarin. Although many physicians prefer aspirin plus clopidogrel because of concerns about bleeding with warfarin, the rates of major bleeding were about the same in the two groups.

In a trial in patients for whom warfarin was “unsuitable,”6 the combination of aspirin plus clopidogrel was associated with a lower rate of stroke than aspirin alone (2.4% per year vs 3.3% per year, relative risk 0.762) but a higher rate of major bleeding events (2.0% per year vs 1.3% per year, relative risk 1.57).

NEW ALTERNATIVES TO WARFARIN

Because of the problems with warfarin, alternatives have been sought for many years. Several new oral anticoagulants are available or are being developed,7 including the factor Xa inhibitors rivaroxaban (Xarelto) and apixaban (Eliquis) and the direct factor II (thrombin) inhibitor dabigatran (Pradaxa) (Table 1).

Dabigatran’s advantages and drawbacks

Dabigatran has been on the market for more than a year and has gained rapid acceptance. The dosage is 150 mg twice a day, or 75 mg twice a day if renal function is impaired. Cleared by the kidneys, it has a half-life of 12 to 17 hours; 75% is cleared within 24 hours. For a patient who needs surgery that poses a low risk of bleeding, the general recommendation is to stop dabigatran the night before the surgical procedure. For operations with a greater risk of bleeding, many surgeons recommend stopping the drug 3 or 4 days before.

Advantages of dabigatran include that it is not influenced by diet and that the onset of therapeutic benefit is within 1 hour. Although some drugs affect dabigatran, drug interactions are more troublesome with warfarin.

A serious concern about dabigatran and the other new agents is that if a bleeding problem arises, the effects of these drugs are not reversible by administration of fresh frozen plasma. Dabigatran is reversible by dialysis; however, if a patient is also hypotensive, dialysis is not an option, and simply waiting for the drug to clear is the only choice.

Another drawback is that therapeutic levels cannot be monitored. If a patient taking warfarin requires cardioversion, the INR is carefully monitored for several weeks beforehand to reduce the risk of stroke. With dabigatran, there is no way to know if a patient is actually taking the drug as prescribed.

 

 

Clinical trials show that alternatives are marginally better than warfarin

In the Randomized Evaluation of Long-Term Anticoagulation Therapy (RE-LY) trial,8 dabigatran was associated with a significantly lower incidence of intracranial hemorrhage, combined strokes, and systemic embolization than warfarin. The incidence of major bleeds was slightly lower with dabigatran. Although dabigatran performed better, the differences were small and would not require patients to change from warfarin if they are already doing well.

Apixaban and rivaroxaban are other alternatives to warfarin, with different mechanisms of action and metabolism. Although rivaroxaban’s half-life is similar to that of apixaban and dabigatran, it is being marketed as allowing once-daily dosing instead of twice-daily.

Recent randomized controlled clinical trials of the new drugs include:

  • The Apixaban Versus Acetylsalicylic Acid (ASA) to Prevent Stroke in Atrial Fibrillation Patients Who Have Failed or Are Unsuitable for Vitamin K Antagonist Treatment (AVERROES) trial,9 which compared apixaban and aspirin
  • The Apixaban for Reduction in Stroke and Other Thromboembolic Events in Atrial Fibrillation (ARISTOTLE) trial comparing apixaban and warfarin10
  • The Rivaroxaban Once Daily Oral Direct Factor Xa Inhibition Compared With Vitamin K Antagonism for Prevention of Stroke and Embolism Trial in Atrial Fibrillation (ROCKET AF),11 comparing rivaroxaban and warfarin
  • RE-LY,8 comparing dabigatran and warfarin.

In the ARISTOTLE,10 ROCKET AF,11 and RE-LY trials,8 the time that the warfarin patients’ INRs were in the therapeutic range varied from 55% to 68%. This seems low and is a problem when trying to compare therapies, but is probably about as high as one can expect in the real world.

In AVERROES,9 the combined rate of stroke and embolism was higher with aspirin than with apixaban. In the other trials, the rates were slightly better with the new drugs than with warfarin, and the rates of major hemorrhage and hemorrhagic stroke were only slightly higher with warfarin than with the new drugs. Because the differences in benefits and risks are so small, the main advantage of the newer drugs will probably be for patients who have difficulty staying in the therapeutic INR range on warfarin.

RATE CONTROL VS RESTORATION OF SINUS RHYTHM

Evidence is insufficient to determine the risk of very-long-term asymptomatic atrial fibrillation in patients on appropriate anticoagulation. Rate control is an option for asymptomatic patients but provides no change in quality of life and no definitive reduction in the risk of stroke. The main argument for restoring normal sinus rhythm in patients with mild to moderate symptoms is that it improves exercise capacity. The need for anticoagulation persists when patients are converted to sinus rhythm because the risk of recurrent atrial fibrillation remains high.

For patients with symptomatic atrial fibrillation, rate control is sometimes achieved with beta-blockers or calcium channel blockers. Rate control may be augmented with the addition of digoxin, but when used alone digoxin generally does not control the rate of atrial fibrillation. However, in many cases of atrial fibrillation, symptoms are not rate-related, and cardioversion to normal sinus rhythm should be attempted. In such cases, the symptoms may be attributable to a loss of atrial transport function.

Patients with the following risk factors should be admitted to the hospital to start antiarrhythmic drugs:

  • Borderline or a long QTc interval at baseline (> 450 msec)
  • Treatment with dofetilide (Tikosyn) because of its effects on the QT interval
  • Heart failure or poor left-ventricular function
  • Sinus node dysfunction
  • Significant atrioventricular conduction disease.

Selecting an antiarrhythmic drug

Any of the antiarrhythmic drugs listed in Table 2 can be used for a patient with lone atrial fibrillation (ie, not caused by underlying heart disease). The choice of drug should be determined by whether coronary artery disease or renal failure is present as well. Liver disease or chronic obstructive pulmonary disease also may affect this decision.

Benefits of dronedarone are mixed

In a randomized trial of dronedarone vs placebo in patients with atrial fibrillation, the rate of death and the rate of first hospitalization due to a cardiovascular event at 21 months were significantly lower with dronedarone.12 No difference was found between the two groups in the rate of death from all causes, but fewer people died of cardiovascular causes in the dronedarone group. More patients taking dronedarone developed bradycardia, QT-interval prolongation, nausea, diarrhea, rash, or a higher serum creatinine level. Gastrointestinal side effects are often a problem with dronedarone: 20% to 30% of patients cannot tolerate the drug.

Dronedarone may cause a small rise in creatinine, and although this effect should be monitored, by itself it should not be interpreted as impairment of renal function. In a study in healthy people,13 dronedarone caused a 10% to 15% increase in serum creatinine, but the glomerular filtration rate was unchanged, as were renal plasma flow and anion secretion.

Another trial, in patients with severe heart failure, found that patients taking dronedarone had higher rates of hospitalization and overall mortality, raising serious concern about the safety of this drug in patients with advanced heart failure.14

Singh et al15 pooled the data from two multicenter, randomized trials that compared dronedarone with placebo for maintaining sinus rhythm in patients with atrial fibrillation or flutter. The mean time to the recurrence of atrial fibrillation was 116 days with dronedarone and 53 days with placebo. Other trials also showed longer times to recurrence and lower recurrence rates with dronedarone. Although the differences were statistically significant, they may not be clinically meaningful for patients.

Dronedarone is structurally similar to amiodarone (Cordarone), but the two drugs work differently. A meta-analysis of clinical trials16 found that amiodarone recipients had a lower rate of recurrence of atrial fibrillation than did those receiving dronedarone.

Two safety warnings for dronedarone

In January 2011, the US Food and Drug Administration (FDA) issued an alert about cases of rare but severe liver injury in patients treated with dronedarone, including two cases of acute liver failure leading to liver transplantation.17

The Permanent Atrial Fibrillation Outcome Study Using Dronedarone on Top of Standard Therapy (PALLAS)18 compared dronedarone and placebo in patients with permanent atrial fibrillation. More people died or had serious cardiovascular adverse events in the dronedarone group. The study was stopped early after data monitoring showed that rates of death, stroke, and hospitalization for heart failure were two times higher in patients receiving dronedarone. This prompted the FDA to issue another safety alert in July 2011.

Interestingly, the PALLAS study did not set out to determine whether dronedarone controls atrial fibrillation, as the study patients had long-standing, persistent atrial fibrillation. The study was designed only to determine if the drug reduces the rate of adverse events; it clearly does not, and the study shows that dronedarone should not be used to control the heart rate in patients with persistent atrial fibrillation. Instead, its use is best restricted to patients with paroxysmal atrial fibrillation without significant cardiovascular disease.

 

 

ABLATION OF ATRIAL FIBRILLATION

Another way to try to restore sinus rhythm is to destroy or isolate the area that is generating the abnormal beats via a catheter-based procedure.

Radiofrequency ablation is generally tried in patients in whom one or two drugs have failed to control atrial fibrillation. Direct comparisons show that ablation is superior to drug therapy and is effective in about 75% of patients with paroxysmal atrial fibrillation vs 20% to 40% of patients on drug therapy. Ablation plus drug therapy is often more effective than either treatment alone.

Mechanisms of atrial fibrillation and ablation

In many cases, atrial fibrillation is stimulated by vagal and sympathetic inputs to the atrium that enter around the pulmonary veins and trigger electrical activations in the area, generating spiraling, reentering circuits. Focal atrial fibrillation also originates predominantly in the pulmonary veins. Ablation of tissue widely circumscribing the mouth of the pulmonary veins prevents the electrical signal from exiting into the atrium.

In about 11% to 37% of cases, atrial fibrillation originates elsewhere, eg, in the left atrium, in the superior vena cava, or in the vein of Marshall. Techniques have evolved to also ablate these regions.

Anticoagulation therapy is recommended before the procedure, and patients at low risk should continue it for a minimum of 2 months afterward. Patients with a higher CHADS2 score should receive anticoagulation therapy for at least 1 year. The consensus statement by the Heart Rhythm Society19 recommends that patients remain on warfarin or one of the newer anticoagulants if their CHADS2 score is 2 or higher. This is because patients have a significant risk of recurrence of atrial fibrillation after radiofrequency ablation, so if their stroke risk is high they should remain on anticoagulant therapy.

Ablation is usually effective, but it carries rare but serious risks

The efficacy of a single radiofrequency ablation procedure is in the range of 60% to 80% for paroxysmal atrial fibrillation and 40% to 60% for persistent atrial fibrillation. The Second International Ablation Registry20 shows a success rate of about 75% in patients with paroxysmal atrial fibrillation and about 65% in patients with persistent and permanent atrial fibrillation. Registry data are often more favorable because reporting is optional, but these results are consistent with those from experienced medical centers. Rates of suppression of atrial fibrillation are higher in patients who also take antiarrhythmic drugs, making a “hybrid” approach useful when ablation alone fails.

According to a worldwide survey, the risk of serious complications is 4.5%. These include stroke (0.23%), tamponade (1.3%), and pulmonary vein stenosis (< 0.29%). The esophagus lies just behind the right atrium, and burning through and creating a fistula between them occurs in about 0.04% of cases and is almost uniformly fatal.20

A second ablation procedure is sometimes indicated for the recurrence of atrial fibrillation, which is almost always caused by recovery of the pulmonary veins. Bhargava et al21 found that the success rate at Cleveland Clinic for a single procedure for paroxysmal atrial fibrillation was 77%, and that it was 92% after a repeat procedure. For persistent atrial fibrillation, success rates were 76% after the first procedure and 90% after the second. Even for long-standing persistent atrial fibrillation (ie, lasting more than 1 year), 80% success was achieved after two procedures. Patients who are less likely to have a successful ablation procedure are those with long-standing atrial fibrillation and coexisting heart disease, including severe valvular disease, although mitral regurgitation sometimes improves if sinus rhythm can be maintained.

The need for a second procedure

After ablation, patients should be closely monitored for a recurrence of atrial fibrillation. Continuous monitoring with implantable cardiac monitor loop recorders can detect unrecognized episodes of arrhythmia. Long-term follow-up is also required to track outcomes and quality of life.

According to the Heart Rhythm Society Task Force on Catheter and Surgical Ablation of Atrial Fibrillation,19 atrial fibrillation recurs after ablation in about 35% to 60% of patients in the first 3 months, with recurrence rates after 1 year ranging from 5% to 16%. The rate of success is determined by the skill of the surgeon, underlying heart disease, attention to follow-up, and how success is defined.

Freedom from recurrence early on is a good predictor that late recurrence is unlikely. Patients who only have a very early recurrence (within the first 4 weeks) are more likely to have long-term freedom from atrial fibrillation tha those who have recurrences after that time.22

In a study of 831 patients, Hussein et al23 found recurrence rates of 24% between months 3 to 13 following ablation and 9% after 12 months. At 55 months, 79% were free from atrial fibrillation without drugs, 11% were free of atrial fibrillation with medications, and 5% had refractory atrial fibrillation.

Recurrence—whether early or late—was more likely to occur in people with persistent vs paroxysmal atrial fibrillation. Other risk factors for late recurrence included older age and larger left atrial size (which is also a risk factor for recurrence on drug therapy). Although recurrent arrhythmia was most often atrial fibrillation, atrial flutter also occurred frequently (in 27% of patients with late recurrence). Three patients (4% of patients with late recurrence) developed atrial tachycardia.23

In patients with early recurrence, 81% underwent repeat ablation, all of whom had recovery of one or more pulmonary veins. After the second ablation, 21% had recurrence, 65% of whom were controlled by medications.23

Whether a patient should undergo subsequent ablation procedures depends on the severity of symptoms, the likelihood of success (based on an educated guess), and the patient’s willingness to undergo another procedure.

ATRIAL APPENDAGE OCCLUSION DEVICE UNDER INVESTIGATION

New devices are being investigated that occlude the left atrial appendage to try to prevent embolization.

The Watchman device, resembling an umbrella, is implanted via a percutaneous catheter in the left atrial appendage, closing it off to preclude a thrombus from forming in the appendage and embolizing to the body. Clinical trials showed that patients who received a device had a slightly lower risk of stroke than otherwise seen in clinical practice.24 Safety and efficacy are still being determined.

The device cannot be deployed in a patient with an existing thrombus because of the danger of dislodging the thrombus, allowing it to embolize.

References
  1. Gage BF, Waterman AD, Shannon W, Boechler M, Rich MW, Radford MJ. Validation of clinical classification schemes for predicting stroke: results from the national Registry of Atrial Fibrillation. JAMA 2001; 285:28642870.
  2. Hylek EM, Skates SJ, Sheehan MA, Singer DE. An analysis of the lowest effective intensity of prophylactic anticoagulation for patients with nonrheumatic atrial fibrillation. N Engl J Med 1996; 335:540546.
  3. Hylek EM, Singer DE. Risk factors for intracranial hemorrhage in outpatients taking warfarin. Ann Intern Med 1994; 120:897902.
  4. Connolly SJ, Pogue J, Eikelboom J, et al; ACTIVE W Investigators. Benefit of oral anticoagulant over antiplatelet therapy in atrial fibrillation depends on the quality of international normalized ratio control achieved by centers and countries as measured by time in therapeutic range. Circulation 2008; 118:20292037.
  5. Verheugt FWA. Who is ineligible for warfarin in atrial fibrillation? Lancet 2009; 374:510511.
  6. ACTIVE Investigators; Connolly SJ, Pogue J, Hart RG. Effect of clopidogrel added to aspirin in patients with atrial fibrillation. N Engl J Med 2009; 360:20662078.
  7. Harenberg J. New anticoagulants in atrial fibrillation. Semin Thromb Hemost 2009; 35:574585.
  8. Connolly SJ, Ezekowitz MD, Yusuf S, et al; RE-LY Steering Committee and Investigators. Dabigatran versus warfarin patients with atrial fibrillation. N Engl J Med 2009; 361:11391151.
  9. Connolly SJ, Eikelboom J, Joyner C, et al; AVERROES Steering Committee and Investigators. Apixaban in patients with atrial fibrillation. N Engl J Med 2011; 364:806817.
  10. Granger CB, Alexander JH, McMurray JJV, et al; for the ARISTOTLE Committees and Investigators. Apixaban versus warfarin in patients with atrial fibrillation. N Engl J Med August 28, 2011; 10.1056/nejmoa1107039.
  11. Patel MR, Mahaffey KW, Garg J, et al; the ROCKET AF Steering Committee. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation. N Engl J Med 2011; 365:883891.
  12. Hohnloser SH, Crijns HJ, van Eickels M, et al; ATHENA Investigators. Effect of dronedarone on cardiovascular events in atrial fibrillation. N Engl J Med 2009; 360:688678.
  13. Tschuppert Y, Buclin T, Rothuizen LE, et al. Effect of dronedarone on renal function in healthy subjects. Br J Clin Pharmacol 2007; 64:785791.
  14. Kóber L, Torp-Pederson C, McMurray JJ, et al; Dronedarone Study Group. Increased mortality after dronedarone therapy for severe heart failure. N Engl J Med 2008; 358:26782687.
  15. Singh BN, Connolly SJ, Crijns HJ, et al; EURDIS and ADONIS Investigators. Dronedarone for maintenance of sinus rhythm in atrial fibrillation or flutter. N Engl J Med 2007; 357:987999.
  16. Piccini JP, Hasselblad V, Peterson ED, Washam JB, Califf RM. Comparative efficacy of dronedarone and amiodarone for the maintenance of sinus rhythm in patients with atrial fibrillation. J Am Coll Cardiol 2009; 54:10891095.
  17. US Food and Drug Administration. FDA drug safety communication: severe liver injury associated with the use of dronedarone (marketed as Multaq). http://www.fda.gov/drugs/drugsafety/ucm240011.htm. Accessed July 5, 2012.
  18. Connolly SJ, Camm AJ, Halperin JL, et al; for the PALLAS Investigators. Dronedarone in high-risk permanent atrial fibrillation. N Engl J Med 2011; 365:22682276.
  19. HRS/EHRA/ECAS expert consensus statement on catheter and surgical ablation of atrial fibrillation: recommendations for personnel, policy, procedures and follow-up. Heart Rhythm 2007; 4:146.
  20. Cappato R, Calkins H, Chen SA, et al. Updated worldwide survey on the methods, efficacy, and safety of catheter ablation for human atrial fibrillation. Circ Arrhythm Electrophysiol 2010; 3:3238.
  21. Bhargava M, Di Biase L, Mohanty P, et al. Impact of type of atrial fibrillation and repeat catheter ablation on long-term freedom from atrial fibrillation: results from a multicenter study. Heart Rhythm 2009; 6:14031412.
  22. Themistoclakis S, Schweikert RA, Sliba WI, et al. Clinical predictors and relationship between early and late atrial tachyarrhythmias after pulmonary vein antrum isolation. Heart Rhythm 2008; 5:679685.
  23. Hussein AA, Saliba WI, Martin DO, et al. Natural history and long-term outcomes of ablated atrial fibrillation. Circ Arrhythm Electrophysiol 2011; 4:271278.
  24. Holmes DR, Reddy VY, Turi ZG, et al; for the PROTECT AF Investigators. Percutaneous closure of the left atrial appendage versus warfarin therapy for prevention of stroke in patients with atrial fibrillation: a randomised non-inferiority trial. Lancet 2009; 374:534542.
References
  1. Gage BF, Waterman AD, Shannon W, Boechler M, Rich MW, Radford MJ. Validation of clinical classification schemes for predicting stroke: results from the national Registry of Atrial Fibrillation. JAMA 2001; 285:28642870.
  2. Hylek EM, Skates SJ, Sheehan MA, Singer DE. An analysis of the lowest effective intensity of prophylactic anticoagulation for patients with nonrheumatic atrial fibrillation. N Engl J Med 1996; 335:540546.
  3. Hylek EM, Singer DE. Risk factors for intracranial hemorrhage in outpatients taking warfarin. Ann Intern Med 1994; 120:897902.
  4. Connolly SJ, Pogue J, Eikelboom J, et al; ACTIVE W Investigators. Benefit of oral anticoagulant over antiplatelet therapy in atrial fibrillation depends on the quality of international normalized ratio control achieved by centers and countries as measured by time in therapeutic range. Circulation 2008; 118:20292037.
  5. Verheugt FWA. Who is ineligible for warfarin in atrial fibrillation? Lancet 2009; 374:510511.
  6. ACTIVE Investigators; Connolly SJ, Pogue J, Hart RG. Effect of clopidogrel added to aspirin in patients with atrial fibrillation. N Engl J Med 2009; 360:20662078.
  7. Harenberg J. New anticoagulants in atrial fibrillation. Semin Thromb Hemost 2009; 35:574585.
  8. Connolly SJ, Ezekowitz MD, Yusuf S, et al; RE-LY Steering Committee and Investigators. Dabigatran versus warfarin patients with atrial fibrillation. N Engl J Med 2009; 361:11391151.
  9. Connolly SJ, Eikelboom J, Joyner C, et al; AVERROES Steering Committee and Investigators. Apixaban in patients with atrial fibrillation. N Engl J Med 2011; 364:806817.
  10. Granger CB, Alexander JH, McMurray JJV, et al; for the ARISTOTLE Committees and Investigators. Apixaban versus warfarin in patients with atrial fibrillation. N Engl J Med August 28, 2011; 10.1056/nejmoa1107039.
  11. Patel MR, Mahaffey KW, Garg J, et al; the ROCKET AF Steering Committee. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation. N Engl J Med 2011; 365:883891.
  12. Hohnloser SH, Crijns HJ, van Eickels M, et al; ATHENA Investigators. Effect of dronedarone on cardiovascular events in atrial fibrillation. N Engl J Med 2009; 360:688678.
  13. Tschuppert Y, Buclin T, Rothuizen LE, et al. Effect of dronedarone on renal function in healthy subjects. Br J Clin Pharmacol 2007; 64:785791.
  14. Kóber L, Torp-Pederson C, McMurray JJ, et al; Dronedarone Study Group. Increased mortality after dronedarone therapy for severe heart failure. N Engl J Med 2008; 358:26782687.
  15. Singh BN, Connolly SJ, Crijns HJ, et al; EURDIS and ADONIS Investigators. Dronedarone for maintenance of sinus rhythm in atrial fibrillation or flutter. N Engl J Med 2007; 357:987999.
  16. Piccini JP, Hasselblad V, Peterson ED, Washam JB, Califf RM. Comparative efficacy of dronedarone and amiodarone for the maintenance of sinus rhythm in patients with atrial fibrillation. J Am Coll Cardiol 2009; 54:10891095.
  17. US Food and Drug Administration. FDA drug safety communication: severe liver injury associated with the use of dronedarone (marketed as Multaq). http://www.fda.gov/drugs/drugsafety/ucm240011.htm. Accessed July 5, 2012.
  18. Connolly SJ, Camm AJ, Halperin JL, et al; for the PALLAS Investigators. Dronedarone in high-risk permanent atrial fibrillation. N Engl J Med 2011; 365:22682276.
  19. HRS/EHRA/ECAS expert consensus statement on catheter and surgical ablation of atrial fibrillation: recommendations for personnel, policy, procedures and follow-up. Heart Rhythm 2007; 4:146.
  20. Cappato R, Calkins H, Chen SA, et al. Updated worldwide survey on the methods, efficacy, and safety of catheter ablation for human atrial fibrillation. Circ Arrhythm Electrophysiol 2010; 3:3238.
  21. Bhargava M, Di Biase L, Mohanty P, et al. Impact of type of atrial fibrillation and repeat catheter ablation on long-term freedom from atrial fibrillation: results from a multicenter study. Heart Rhythm 2009; 6:14031412.
  22. Themistoclakis S, Schweikert RA, Sliba WI, et al. Clinical predictors and relationship between early and late atrial tachyarrhythmias after pulmonary vein antrum isolation. Heart Rhythm 2008; 5:679685.
  23. Hussein AA, Saliba WI, Martin DO, et al. Natural history and long-term outcomes of ablated atrial fibrillation. Circ Arrhythm Electrophysiol 2011; 4:271278.
  24. Holmes DR, Reddy VY, Turi ZG, et al; for the PROTECT AF Investigators. Percutaneous closure of the left atrial appendage versus warfarin therapy for prevention of stroke in patients with atrial fibrillation: a randomised non-inferiority trial. Lancet 2009; 374:534542.
Issue
Cleveland Clinic Journal of Medicine - 79(8)
Issue
Cleveland Clinic Journal of Medicine - 79(8)
Page Number
553-559
Page Number
553-559
Publications
Publications
Topics
Article Type
Display Headline
Atrial fibrillation: New drugs, devices, and procedures
Display Headline
Atrial fibrillation: New drugs, devices, and procedures
Sections
Inside the Article

KEY POINTS

  • Warfarin is as safe as—and more effective than—the combination of aspirin and clopidogrel (Plavix) if the international normalized ratio is in the therapeutic range 65% of the time or more.
  • New anticoagulants are promising alternatives to warfarin, but they also pose risks. Patients who are doing well on warfarin need not change.
  • Several antiarrhythmic drugs are available to control symptomatic atrial fibrillation. Dronedarone (Multaq) should only be considered for patients with paroxysmal atrial fibrillation without significant cardiovascular disease.
  • Ablation is often effective in controlling atrial fibrillation, but recurrence is common. Early recurrence often subsides, but late recurrence often requires a repeat procedure.
Disallow All Ads
Alternative CME
Article PDF Media

Movement Disorders: Therapy Update for the Modern Clinician

Article Type
Changed
Wed, 04/10/2019 - 11:25
Display Headline
Movement Disorders: Therapy Update for the Modern Clinician
Proceedings of a Symposium

Supplement Editors:
Hubert H. Fernandez, MD, and Nestor Galvez-Jimenez, MD

Contents

Managing the patient with newly diagnosed Parkinson disease
Carlos Singer, MD

Off spells and dyskinesias: Pharmacologic management of motor complications
Tarannum S. Khan, MD

Nonmotor complications of Parkinson disease
Hubert H. Fernandez, MD

Deep brain stimulation for movement disorders: Patient selection and technical options
Andre G. Machado, MD, PhD; Milind Deogaonkar, MD; and Scott Cooper, MD, PhD

Use of chemodenervation in dystonic conditions
Maurice Hanson, MD

Comprehensive treatment of Huntington disease and other choreic disorders
Carlos Singer, MD

Tics and Tourette syndrome: An adult perspective
Nestor Galvez-Jimenez, MD, MSc, MS(HSA), FACP

Surgical considerations for tremor and dystonia
Scott Cooper, MD, PhD, and Mark Bowes, PhD

 

Article PDF
Issue
Cleveland Clinic Journal of Medicine - 79(7)
Publications
Topics
Page Number
S1-S43
Sections
Article PDF
Article PDF
Proceedings of a Symposium
Proceedings of a Symposium

Supplement Editors:
Hubert H. Fernandez, MD, and Nestor Galvez-Jimenez, MD

Contents

Managing the patient with newly diagnosed Parkinson disease
Carlos Singer, MD

Off spells and dyskinesias: Pharmacologic management of motor complications
Tarannum S. Khan, MD

Nonmotor complications of Parkinson disease
Hubert H. Fernandez, MD

Deep brain stimulation for movement disorders: Patient selection and technical options
Andre G. Machado, MD, PhD; Milind Deogaonkar, MD; and Scott Cooper, MD, PhD

Use of chemodenervation in dystonic conditions
Maurice Hanson, MD

Comprehensive treatment of Huntington disease and other choreic disorders
Carlos Singer, MD

Tics and Tourette syndrome: An adult perspective
Nestor Galvez-Jimenez, MD, MSc, MS(HSA), FACP

Surgical considerations for tremor and dystonia
Scott Cooper, MD, PhD, and Mark Bowes, PhD

 

Supplement Editors:
Hubert H. Fernandez, MD, and Nestor Galvez-Jimenez, MD

Contents

Managing the patient with newly diagnosed Parkinson disease
Carlos Singer, MD

Off spells and dyskinesias: Pharmacologic management of motor complications
Tarannum S. Khan, MD

Nonmotor complications of Parkinson disease
Hubert H. Fernandez, MD

Deep brain stimulation for movement disorders: Patient selection and technical options
Andre G. Machado, MD, PhD; Milind Deogaonkar, MD; and Scott Cooper, MD, PhD

Use of chemodenervation in dystonic conditions
Maurice Hanson, MD

Comprehensive treatment of Huntington disease and other choreic disorders
Carlos Singer, MD

Tics and Tourette syndrome: An adult perspective
Nestor Galvez-Jimenez, MD, MSc, MS(HSA), FACP

Surgical considerations for tremor and dystonia
Scott Cooper, MD, PhD, and Mark Bowes, PhD

 

Issue
Cleveland Clinic Journal of Medicine - 79(7)
Issue
Cleveland Clinic Journal of Medicine - 79(7)
Page Number
S1-S43
Page Number
S1-S43
Publications
Publications
Topics
Article Type
Display Headline
Movement Disorders: Therapy Update for the Modern Clinician
Display Headline
Movement Disorders: Therapy Update for the Modern Clinician
Sections
Citation Override
Cleveland Clinic Journal of Medicine 2012 July;79(7 suppl 2):S1-S43
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Managing the patient with newly diagnosed Parkinson disease

Article Type
Changed
Mon, 10/01/2018 - 15:32
Display Headline
Managing the patient with newly diagnosed Parkinson disease

Parkinson disease (PD) is a slowly progressive neurodegenerative disorder. Early PD, or stage 1 or 2 on the Unified Parkinson’s Disease Rating Scale (UPDRS), is characterized by mild symptoms, minimal to mild disability, and lack of postural instability or cognitive decline. The goal of therapy in PD is to help patients retain functional independence for as long as possible. Therapeutic choices in early PD are guided by the effect of symptoms on function and quality of life, consideration of complications associated with long-term levodopa, the likelihood of response fluctuations to levodopa, and the potential for a neuroprotective effect.

SYMPTOMATIC THERAPIES IN EARLY PD

Dopaminergic replacement therapy with levodopa is a legitimate choice for the treatment of early PD. Use of carbidopa-levodopa has been shown to slow the progression of PD in a dose-dependent manner as evidenced by a decrease in total score on the UPDRS in patients with early PD who were randomly assigned to receive carbidopa-levodopa compared with those who received a placebo.1

Alternatives to levodopa

There are several reasons to choose an alternative to levodopa for the treatment of early PD. The first is to postpone the development of levodopa-induced dyskinesias, which are linked to duration of levodopa treatment and total exposure to levodopa. The second is postponement of the “wearing-off” effect; that is, the reemergence of symptoms that occurs in some patients before their next scheduled dose of levodopa. Such reasoning applies to early PD patients with minimal or no disability and—in particular—to young-onset PD patients who tend to develop vigorous dyskinesias and dramatic wearing-off phenomena. Pharmacologic alternatives to levodopa in early PD include monoamine oxidase (MAO)-B inhibitors, amantadine, and dopamine agonists.

Reprinted with permission from The New England Journal of Medicine (Olanow CW, et al. A double-blind, delayed-start trial of rasagiline in Parkinson’s disease. N Engl J Med 2009; 361:168–178). © 2009 Massachusetts Medical Society.
Figure 1. Rasagiline at doses of 1 mg/day (A) and mg/day (B) slowed the rate of worsening of the Unified Parkinson’s Disease Rating Scale (UPDRS) score compared with placebo in patients with untreated Parkinson disease.2
MAO-B inhibitors. Two MAO-B inhibitors are approved by the US Food and Drug Administration for the treatment of PD: rasagiline and selegiline. These agents have a mildly symptomatic effect. In the Attenuation of Disease Progression with Azilect Given Once-daily (ADAGIO) trial, use of rasagiline at doses of 1 and 2 mg/d slowed the rate of worsening of the UPDRS score compared with placebo in patients with untreated PD (Figure 1).2 Patients were randomly assigned to an early-start group (rasagiline, 1 or 2 mg/d, or placebo for 72 weeks) or a late-start group (placebo for 36 weeks followed by rasagiline, 1 or 2 mg/d, or placebo for 36 weeks). The rate of change in the UPDRS score was slowed significantly with early treatment with rasagiline at a dosage of 1 mg/d, but not at 2 mg/d. Because the hierarchical primary end points for the ADAGIO trial were met only for the cohort receiving 1 mg of rasagiline early, it remains inconclusive whether rasagiline has a neuroprotective effect.

In a placebo-controlled study of selegiline in de novo early-phase PD, Pålhagen et al showed that selegiline monotherapy delayed the need for levodopa. When used in combination with levodopa, selegiline was able to slow the progression of PD as measured by the change in UPDRS total score.3

Amantadine. In an early study of 54 patients with PD, functional disability scores improved significantly with administration of amantadine 200 to 300 mg/d compared with placebo.4 A small subset of patients, perhaps 20% or less, who are treated with amantadine experience robust symptom improvement. Side effects of amantadine include hallucinations, edema, livedo reticularis, and anticholinergic effects. A more recently discovered potential side effect is corneal edema.

Dopamine agonists. Pramipexole (immediate-release [IR] and extended-release [ER]), and ropinirole IR and ER are dopamine agonists that have demonstrated disease-modifying effects and efficacy in improving PD symptoms.

Pramipexole ER administered once daily in early PD was shown to be superior to placebo on the mean UPDRS total score.5 Ropinirole ER produced mean plasma concentrations over 24 hours similar to those achieved with ropinirole IR, and showed noninferiority to ropinirole IR on efficacy measures in patients with de novo PD.5

The effective dosage range of pramipexole ER in early PD is 0.375 to 4.5 mg/d. Side effects include hallucinations, edema, excessive diurnal somnolence, and impulse control disorders (ie, pathologic gambling, hypersexuality, excessive craving for sweets). Compared with pramipexole IR, compliance is enhanced with the ER formulation because of ease of administration, but this formulation also is more expensive.

In early PD, the effective dosage range of ropinirole ER is 8 to 12 mg/d.6 The side effects are the same as with pramipexole ER with the same compliance advantage and cost disadvantage compared with the IR formulation.

Research indicates that dopamine agonists may have a neuroprotective effect. In two large clinical trials in which patients with PD were followed with an imaging marker of dopamine neuronal degeneration (using single-photon emission computed tomography or positron emission tomography), recipients of pramipexole7 or ropinirole8 showed slower neuronal deterioration compared with levodopa recipients. A counterargument to the neuroprotective theory is that these differences between the dopamine agonists and levodopa reflect neurotoxicity of levodopa rather than neuroprotection by dopamine agonists. The absence of a placebo comparison in both trials adds to the difficulty in drawing a conclusion, as some critics ascribed the differences between groups to downregulation of tracer binding with levodopa.

Nonergoline dopamine agonist. Transdermal rotigotine is a nonergot D1/D2/D3 agonist. Higher doses produce higher plasma levels of rotigotine, which remain steady over the 24-hour dosing interval.9 Transdermal rotigotine has demonstrated effectiveness in early PD in several clinical trials.10,11 The patch, applied once daily, provides a constant release of medication. Removing the patch immediately interrupts drug administration.

Rotigotine patches must be refrigerated to prevent crystallization, a requirement that has delayed the product’s arrival on the market. The patch is reputed to be difficult to peel from its backing and apply. Skin reactions are a side effect, and nonergot side effects are possible. Despite these drawbacks, transdermal rotigotine represents a convenient option for perioperative management of PD and in patients with dysphagia.

Exercise. Exercise has symptomatic and possibly neuroprotective benefits in PD, supporting its use as an additional medical measure. Evidence supports the value of treadmill walking and high-impact exercise in improving stride length, quality of life, and motor response to levodopa.

 

 

SYMPTOMATIC THERAPIES: THE FUTURE

Partial dopamine agonists

Pardoprunox is a partial dopamine agonist with full 5-HT1A–agonist activity. A partial dopamine agonist acts in two ways: (1) It stimulates dopamine production in brain regions with low dopamine tone, and (2) it has dopamine antagonist activity under circumstances of high dopamine sensitivity, theoretically avoiding overstimulation of dopamine receptors. Because it inhibits excessive dopamine effect, pardoprunox may prevent dyskinesia. In addition, because pardoprunox has serotonin agonist activity, it may also act as an antidepressant.

In a phase 2 study, significantly more patients randomized to pardoprunox had a 30% or greater reduction in UPDRS motor score compared with placebo at end-of-dose titration (35.8% for pardoprunox vs 15.7% for placebo; P = .0065) and at end point (50.7% for pardoprunox vs 15.7% for placebo; P < .0001).12

Adenosine A2A-receptor antagonists

Adenosine A2A receptors are located in the basal ganglia, primarily on gamma aminobutyric acid (GABA)–mediated enkephalin-expressing medium spiny neurons in the striatum. These receptors modulate dopamine transmission by opposing D2-receptor activity. The D2 pathway is an indirect pathway that promotes suppression of unnecessary movement.

Two A2A-receptor antagonists have demonstrated efficacy in clinical trials. Vipadenant has been proven effective as monotherapy in phase 2 clinical trials. Preladenant has been shown to improve “off time” as an adjunct to levodopa without increasing dyskinesia.

Safinamide

Safinamide, currently in phase 3 clinical trials, has three mechanisms of action. It is an inhibitor of dopamine reuptake, a reversible inhibitor of MAO-B, and an inhibitor of excessive glutamate release. The addition of safinamide to a stable dose of a single dopamine agonist in patients with early PD resulted in improvement of motor symptoms and cognitive function.13,14

NEUROPROTECTIVE STRATEGIES UNDER INVESTIGATION

Four neuroprotective strategies are under study: enhanced mitochondrial function, antiinflammatory mechanisms, calcium channel blockade, and uric acid elevation.

Enhanced mitochondrial function

Creatine has generated interest as a disease-modifying agent in response to preclinical data showing that it could enhance mitochondrial function and prevent mitochondrial loss in the brain in models of PD. Creatine is now the subject of a large phase 3 National Institutes of Health–sponsored clinical trial in patients with early-stage PD.15

Coenzyme Q10 (CoQ 10) exhibited a trend for neuroprotection at 1,200 mg/d, lowering the total mean UPDRS score compared with placebo in a 16-month study.16 Current efforts are directed at determining whether 1,200 or 2,400 mg/d of CoQ10 are neuroprotective. A nanoparticulate form of CoQ10, 100 mg three times a day, has been shown to produce plasma levels of CoQ10 equivalent to those produced by 1,200-mg doses of the standard form.17 CoQ10 is free of symptomatic effects.

Antiinflammatory mechanisms

Parkinson disease may have an important inflammatory component. A meta-analysis of seven studies showed an overall hazard ratio of 0.85 for development of PD in users of nonaspirin nonsteroidal antiinflammatory drugs (NSAIDs), with each of the seven studies demonstrating a hazard ratio less than 1.18 A similar meta-analysis showed no such association.19 Further study is warranted.

The antidiabetic agent pioglitazone, shown in mice to prevent dopaminergic nigral cell loss, has been entered into a phase 2 clinical trial to assess its antiinflammatory properties in PD.

Calcium channel blockade

A sustained-release formulation of isradipine, an L-type calcium channel blocker, is being studied in a phase 2 clinical trial for the treatment of early PD; experimental evidence in animals suggests that it may be neuroprotective against PD.

Uric acid elevation

Urate concentration in the cerebrospinal fluid predicts progression of PD, with higher levels associated with slower progression of disease.20 Urate may delay oxidative destruction of dopaminergic neurons that occurs with progression of PD. Pharmacologic elevation of uric acid is being explored as a treatment option in PD.

ELECTRODES, VECTORS, AND STEM CELLS

Deep brain stimulation

Reprinted with permission from Movement Disorders (Espay AJ, et al. Early versus delayed bilateral subthalamic deep brain stimulation for Parkinson’s disease: a decision analysis. Mov Disord 2010; 25:1456–1463). Copyright © 2010 Movement Disorder Society.
Figure 2. In a comparison of early versus late subthalamic nucleus deep brain stimulation (STN DBS), the annual rate of progression of both cognitive (A) and motor (B) decline was slower when STN DBS was administered earlier in the course of Parkinson disease (PD). Late STN DBS is favored if the annual rate of motor progression is greater than 25%, but this is an unrealistic scenario for PD.21
Deep brain stimulation (DBS) is currently used as a treatment for advanced PD (patients suffering from levodopainduced motor complications), but it might also slow the progression of cognitive and motor decline in earlier stages of PD. The annual rate of progression of both cognitive and motor decline was slower when DBS was administered earlier in the course of PD (off time on levodopa of about 2 hours) versus in a later stage of PD (off time on levodopa of about 4 hours) (Figure 2).21 The strategy is being tested further in clinical trials of early PD.

Stem cell therapy

Stem cells obtained from blastocytes, fibroblasts, bone marrow, or the adult, embryonic, or fetal central nervous system through “molecular alchemy” can form dopaminergic neuroblasts. Given the high cost and potential risks of stem cell therapy, it must be proven superior to DBS to be considered an option for early PD. Several practical problems act as hurdles to successful stem cell therapy. Efficient generation of dopamine-producing neurons and successful grafting are required. Tumor growth is a risk. Involuntary movements have been observed in some patients who received fetal implants. A limitation of stem cell therapy is that it will only affect those aspects of PD that are dependent on dopamine.

Gene therapy

Gene delivery of the growth factor analogue adeno-associated type-2 vector (AAV2)-neurturin has been investigated in patients with advanced PD. When surgically placed inside a neuron, neurturin enhances neuron vitality, enabling it to better fight oxidative stress and other attacks. It fared no better than sham surgery on changes in UPDRS motor score at 12 months in a randomized trial.22 A few patients enrolled in this trial have been followed for longer than 12 months, at which time the mean change in motor scores appears to favor the group assigned to gene delivery of AAV2-neurturin. A phase 1/2 trial is investigating the safety and efficacy of bilateral intraputaminal and intranigral administration of neurturin.

SUMMARY

Levodopa is a legitimate choice for the treatment of early PD. Two MAO-B inhibitors, rasagiline and selegiline, have a symptomatic effect.

Long-acting oral and transdermal dopamine agonists are effective symptomatic therapies, but they also have an interesting array of side effects, making levodopa a reasonable alternative treatment sooner or later despite its dyskinetic effect. Potential neuroprotective effects remain to be identified.

Amantadine is sometimes overlooked as an option for treating early PD, but it has some special side effects including leg edema, livedo reticularis, and corneal edema. Amantadine does not cause orthostatic hypotension and is free of the side effects of excessive diurnal somnolence and impulse control disorders that are prevalent with dopamine agonists.

In the future, partial dopamine agonists and adenosine antagonists may provide us with additional symptomatic therapies. CoQ10, creatine, calcium channel blockers, and inosine, as well as NSAIDs, are being actively studied as potential disease-modifying agents. Further studies are likely to come from the use of NSAIDs.

Early DBS is a new avenue of investigation as a potential disease modifier. Stem cells are still being studied and limitations of sufficient production and potential tumor growth, among others, have delayed the institution of clinical trials. Gene therapy is an interesting additional treatment modality in active research.

References
  1. Fahn S, Oakes D, Shoulson I, et al. Levodopa and the progression of Parkinson’s disease. N Engl J Med 2004; 351:24982508.
  2. Olanow CW, Rascol O, Hauser R, et al. A double-blind, delayed-start trial of rasagiline in Parkinson’s disease. N Engl J Med 2009; 361:12681278.
  3. Pålhagen S, Heinonen E, Hägglund J, et al. Selegiline slows the progression of the symptoms of Parkinson disease. Neurology 2006; 66:12001206.
  4. Barbeau A, Mars H, Botez MI, Joubert M. Amantadine-HCl (Symmetrel) in the management of Parkinson’s disease: a double-blind cross-over study. Can Med Assoc J 1971; 105:4246.
  5. Hauser RA, Schapira AH, Rascol O, et al. Randomized, double-blind, multicenter evaluation of pramipexole extended release once daily in early Parkinson’s disease. Mov Disord 2010; 25:25422549.
  6. Onofrj M, Bonanni L, De Angelis MV, Anzellotti F, Ciccocioppo F, Thomas A. Long half-life and prolonged-release dopamine receptor antagonists: a review of ropinirole prolonged-release studies. Parkinsonism Relat Disord 2009; 15 (suppl 4):S85S92.
  7. Parkinson Study Group. Dopamine transporter brain imaging to assess the effects of pramipexole vs levodopa on Parkinson disease progression. JAMA 2002; 287:16531661.
  8. Whone AL, Watts RL, Stoessl AJ, et al. Slower progression of Parkinson’s disease with ropinirole versus levodopa: the REAL-PET study. Ann Neurol 2003; 54:93101.
  9. Reichmann H. Transdermal delivery of dopamine receptor agonists. Parkinsonism Relat Disord 2009; 15 (suppl 4):S93S96.
  10. Parkinson Study Group. A controlled trial of rotigotine monotherapy in early Parkinson’s disease. Arch Neurol 2003; 60:17211728.
  11. Watts RL, Jankovic J, Waters C, et al. Randomized, blind, controlled trial of transdermal rotigotine in early Parkinson disease. Neurology 2007; 68:272276.
  12. Bronzova J, Sampaio C, Hauser RA, et al. Double-blind study of pardoprunox, a new partial dopamine agonist, in early Parkinson’s disease. Mov Disord 2010; 25:738746.
  13. Stocchi F, Arnold G, Onofrj M, et al; Safinamide Parkinson’s Study Group. Improvement of motor function in early Parkinson disease by safinamide. Neurology 2004; 63:746748.
  14. Schapira AHV. Safinamide in the treatment of Parkinson’s disease. Expert Opin Pharmacother 2010; 11:22612268.
  15. NINDS NET-PD Investigators. A randomized, double-blind, futility clinical trial of creatine and minocycline in early Parkinson disease. Neurology 2006; 66:664671.
  16. Shults CW, Oakes D, Kieburtz K; the Parkinson Study Group. Effects of coenzyme q10 in early Parkinson disease: evidence of slowing of the functional decline. Arch Neurol 2002; 59:15411550.
  17. Storch A, Jost W, Vieregge P, et al. Randomized, double-blind, placebo-controlled trial on symptomatic effects of coenzyme q(10) in Parkinson disease. Arch Neurol 2007; 64:938944.
  18. Gagne JJ, Power MC. Anti-inflammatory drugs and risk of Parkinson disease: a meta-analysis. Neurology 2010; 74:9951002.
  19. Samii A, Etminan M, Wiens MO, Jafari S. NSAID use and the risk of Parkinson’s disease: systematic review and meta-analysis of observational studies. Drugs Aging 2009; 26:769779.
  20. Ascherio A, LeWitt PA, Xu K, et al; Parkinson Study Group DATATOP Investigators. Urate as a predictor of the rate of clinical decline in Parkinson disease. Arch Neurol 2009; 66:14601468.
  21. Espay AJ, Vaughan JE, Marras C, Fowler R, Eckman MH. Early versus delayed bilateral subthalamic deep brain stimulation for Parkinson’s disease: a decision analysis. Mov Disord 2010; 25:14561463.
  22. Marks WJ, Bartus RT, Siffert J, et al. Gene delivery of AAV2-neurturin for Parkinson’s disease: a double-blind, randomised, controlled trial. Lancet Neur 2010; 9:11641172.
Article PDF
Author and Disclosure Information

Carlos Singer, MD
Professor of Neurology, Chief, Center for Parkinson’s Disease and Movement Disorders, Leonard M. Miller School of Medicine, University of Miami, Miami, FL

Correspondence: Carlos Singer, MD, Center for Parkinson’s Disease and Movement Disorders, Clinical Research Building, Leonard M. Miller School of Medicine, University of Miami, 1120 NW 14th Street, 13th Floor, Miami, FL 33136; CSinger@med.miami.edu

Dr. Singer reported receiving grant support from Boehringer Ingelheim and Teva Pharmaceuticals, Inc.; and advisory committee or review panel membership for Lundbeck.

This article is based on Dr. Singer’s presentation at "The Annual Therapy Symposium on Movement Disorders for the Modern Clinician" held in Fort Lauderdale, Florida, on January 29, 2011. The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Singer.

Publications
Page Number
S3-S7
Author and Disclosure Information

Carlos Singer, MD
Professor of Neurology, Chief, Center for Parkinson’s Disease and Movement Disorders, Leonard M. Miller School of Medicine, University of Miami, Miami, FL

Correspondence: Carlos Singer, MD, Center for Parkinson’s Disease and Movement Disorders, Clinical Research Building, Leonard M. Miller School of Medicine, University of Miami, 1120 NW 14th Street, 13th Floor, Miami, FL 33136; CSinger@med.miami.edu

Dr. Singer reported receiving grant support from Boehringer Ingelheim and Teva Pharmaceuticals, Inc.; and advisory committee or review panel membership for Lundbeck.

This article is based on Dr. Singer’s presentation at "The Annual Therapy Symposium on Movement Disorders for the Modern Clinician" held in Fort Lauderdale, Florida, on January 29, 2011. The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Singer.

Author and Disclosure Information

Carlos Singer, MD
Professor of Neurology, Chief, Center for Parkinson’s Disease and Movement Disorders, Leonard M. Miller School of Medicine, University of Miami, Miami, FL

Correspondence: Carlos Singer, MD, Center for Parkinson’s Disease and Movement Disorders, Clinical Research Building, Leonard M. Miller School of Medicine, University of Miami, 1120 NW 14th Street, 13th Floor, Miami, FL 33136; CSinger@med.miami.edu

Dr. Singer reported receiving grant support from Boehringer Ingelheim and Teva Pharmaceuticals, Inc.; and advisory committee or review panel membership for Lundbeck.

This article is based on Dr. Singer’s presentation at "The Annual Therapy Symposium on Movement Disorders for the Modern Clinician" held in Fort Lauderdale, Florida, on January 29, 2011. The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Singer.

Article PDF
Article PDF

Parkinson disease (PD) is a slowly progressive neurodegenerative disorder. Early PD, or stage 1 or 2 on the Unified Parkinson’s Disease Rating Scale (UPDRS), is characterized by mild symptoms, minimal to mild disability, and lack of postural instability or cognitive decline. The goal of therapy in PD is to help patients retain functional independence for as long as possible. Therapeutic choices in early PD are guided by the effect of symptoms on function and quality of life, consideration of complications associated with long-term levodopa, the likelihood of response fluctuations to levodopa, and the potential for a neuroprotective effect.

SYMPTOMATIC THERAPIES IN EARLY PD

Dopaminergic replacement therapy with levodopa is a legitimate choice for the treatment of early PD. Use of carbidopa-levodopa has been shown to slow the progression of PD in a dose-dependent manner as evidenced by a decrease in total score on the UPDRS in patients with early PD who were randomly assigned to receive carbidopa-levodopa compared with those who received a placebo.1

Alternatives to levodopa

There are several reasons to choose an alternative to levodopa for the treatment of early PD. The first is to postpone the development of levodopa-induced dyskinesias, which are linked to duration of levodopa treatment and total exposure to levodopa. The second is postponement of the “wearing-off” effect; that is, the reemergence of symptoms that occurs in some patients before their next scheduled dose of levodopa. Such reasoning applies to early PD patients with minimal or no disability and—in particular—to young-onset PD patients who tend to develop vigorous dyskinesias and dramatic wearing-off phenomena. Pharmacologic alternatives to levodopa in early PD include monoamine oxidase (MAO)-B inhibitors, amantadine, and dopamine agonists.

Reprinted with permission from The New England Journal of Medicine (Olanow CW, et al. A double-blind, delayed-start trial of rasagiline in Parkinson’s disease. N Engl J Med 2009; 361:168–178). © 2009 Massachusetts Medical Society.
Figure 1. Rasagiline at doses of 1 mg/day (A) and mg/day (B) slowed the rate of worsening of the Unified Parkinson’s Disease Rating Scale (UPDRS) score compared with placebo in patients with untreated Parkinson disease.2
MAO-B inhibitors. Two MAO-B inhibitors are approved by the US Food and Drug Administration for the treatment of PD: rasagiline and selegiline. These agents have a mildly symptomatic effect. In the Attenuation of Disease Progression with Azilect Given Once-daily (ADAGIO) trial, use of rasagiline at doses of 1 and 2 mg/d slowed the rate of worsening of the UPDRS score compared with placebo in patients with untreated PD (Figure 1).2 Patients were randomly assigned to an early-start group (rasagiline, 1 or 2 mg/d, or placebo for 72 weeks) or a late-start group (placebo for 36 weeks followed by rasagiline, 1 or 2 mg/d, or placebo for 36 weeks). The rate of change in the UPDRS score was slowed significantly with early treatment with rasagiline at a dosage of 1 mg/d, but not at 2 mg/d. Because the hierarchical primary end points for the ADAGIO trial were met only for the cohort receiving 1 mg of rasagiline early, it remains inconclusive whether rasagiline has a neuroprotective effect.

In a placebo-controlled study of selegiline in de novo early-phase PD, Pålhagen et al showed that selegiline monotherapy delayed the need for levodopa. When used in combination with levodopa, selegiline was able to slow the progression of PD as measured by the change in UPDRS total score.3

Amantadine. In an early study of 54 patients with PD, functional disability scores improved significantly with administration of amantadine 200 to 300 mg/d compared with placebo.4 A small subset of patients, perhaps 20% or less, who are treated with amantadine experience robust symptom improvement. Side effects of amantadine include hallucinations, edema, livedo reticularis, and anticholinergic effects. A more recently discovered potential side effect is corneal edema.

Dopamine agonists. Pramipexole (immediate-release [IR] and extended-release [ER]), and ropinirole IR and ER are dopamine agonists that have demonstrated disease-modifying effects and efficacy in improving PD symptoms.

Pramipexole ER administered once daily in early PD was shown to be superior to placebo on the mean UPDRS total score.5 Ropinirole ER produced mean plasma concentrations over 24 hours similar to those achieved with ropinirole IR, and showed noninferiority to ropinirole IR on efficacy measures in patients with de novo PD.5

The effective dosage range of pramipexole ER in early PD is 0.375 to 4.5 mg/d. Side effects include hallucinations, edema, excessive diurnal somnolence, and impulse control disorders (ie, pathologic gambling, hypersexuality, excessive craving for sweets). Compared with pramipexole IR, compliance is enhanced with the ER formulation because of ease of administration, but this formulation also is more expensive.

In early PD, the effective dosage range of ropinirole ER is 8 to 12 mg/d.6 The side effects are the same as with pramipexole ER with the same compliance advantage and cost disadvantage compared with the IR formulation.

Research indicates that dopamine agonists may have a neuroprotective effect. In two large clinical trials in which patients with PD were followed with an imaging marker of dopamine neuronal degeneration (using single-photon emission computed tomography or positron emission tomography), recipients of pramipexole7 or ropinirole8 showed slower neuronal deterioration compared with levodopa recipients. A counterargument to the neuroprotective theory is that these differences between the dopamine agonists and levodopa reflect neurotoxicity of levodopa rather than neuroprotection by dopamine agonists. The absence of a placebo comparison in both trials adds to the difficulty in drawing a conclusion, as some critics ascribed the differences between groups to downregulation of tracer binding with levodopa.

Nonergoline dopamine agonist. Transdermal rotigotine is a nonergot D1/D2/D3 agonist. Higher doses produce higher plasma levels of rotigotine, which remain steady over the 24-hour dosing interval.9 Transdermal rotigotine has demonstrated effectiveness in early PD in several clinical trials.10,11 The patch, applied once daily, provides a constant release of medication. Removing the patch immediately interrupts drug administration.

Rotigotine patches must be refrigerated to prevent crystallization, a requirement that has delayed the product’s arrival on the market. The patch is reputed to be difficult to peel from its backing and apply. Skin reactions are a side effect, and nonergot side effects are possible. Despite these drawbacks, transdermal rotigotine represents a convenient option for perioperative management of PD and in patients with dysphagia.

Exercise. Exercise has symptomatic and possibly neuroprotective benefits in PD, supporting its use as an additional medical measure. Evidence supports the value of treadmill walking and high-impact exercise in improving stride length, quality of life, and motor response to levodopa.

 

 

SYMPTOMATIC THERAPIES: THE FUTURE

Partial dopamine agonists

Pardoprunox is a partial dopamine agonist with full 5-HT1A–agonist activity. A partial dopamine agonist acts in two ways: (1) It stimulates dopamine production in brain regions with low dopamine tone, and (2) it has dopamine antagonist activity under circumstances of high dopamine sensitivity, theoretically avoiding overstimulation of dopamine receptors. Because it inhibits excessive dopamine effect, pardoprunox may prevent dyskinesia. In addition, because pardoprunox has serotonin agonist activity, it may also act as an antidepressant.

In a phase 2 study, significantly more patients randomized to pardoprunox had a 30% or greater reduction in UPDRS motor score compared with placebo at end-of-dose titration (35.8% for pardoprunox vs 15.7% for placebo; P = .0065) and at end point (50.7% for pardoprunox vs 15.7% for placebo; P < .0001).12

Adenosine A2A-receptor antagonists

Adenosine A2A receptors are located in the basal ganglia, primarily on gamma aminobutyric acid (GABA)–mediated enkephalin-expressing medium spiny neurons in the striatum. These receptors modulate dopamine transmission by opposing D2-receptor activity. The D2 pathway is an indirect pathway that promotes suppression of unnecessary movement.

Two A2A-receptor antagonists have demonstrated efficacy in clinical trials. Vipadenant has been proven effective as monotherapy in phase 2 clinical trials. Preladenant has been shown to improve “off time” as an adjunct to levodopa without increasing dyskinesia.

Safinamide

Safinamide, currently in phase 3 clinical trials, has three mechanisms of action. It is an inhibitor of dopamine reuptake, a reversible inhibitor of MAO-B, and an inhibitor of excessive glutamate release. The addition of safinamide to a stable dose of a single dopamine agonist in patients with early PD resulted in improvement of motor symptoms and cognitive function.13,14

NEUROPROTECTIVE STRATEGIES UNDER INVESTIGATION

Four neuroprotective strategies are under study: enhanced mitochondrial function, antiinflammatory mechanisms, calcium channel blockade, and uric acid elevation.

Enhanced mitochondrial function

Creatine has generated interest as a disease-modifying agent in response to preclinical data showing that it could enhance mitochondrial function and prevent mitochondrial loss in the brain in models of PD. Creatine is now the subject of a large phase 3 National Institutes of Health–sponsored clinical trial in patients with early-stage PD.15

Coenzyme Q10 (CoQ 10) exhibited a trend for neuroprotection at 1,200 mg/d, lowering the total mean UPDRS score compared with placebo in a 16-month study.16 Current efforts are directed at determining whether 1,200 or 2,400 mg/d of CoQ10 are neuroprotective. A nanoparticulate form of CoQ10, 100 mg three times a day, has been shown to produce plasma levels of CoQ10 equivalent to those produced by 1,200-mg doses of the standard form.17 CoQ10 is free of symptomatic effects.

Antiinflammatory mechanisms

Parkinson disease may have an important inflammatory component. A meta-analysis of seven studies showed an overall hazard ratio of 0.85 for development of PD in users of nonaspirin nonsteroidal antiinflammatory drugs (NSAIDs), with each of the seven studies demonstrating a hazard ratio less than 1.18 A similar meta-analysis showed no such association.19 Further study is warranted.

The antidiabetic agent pioglitazone, shown in mice to prevent dopaminergic nigral cell loss, has been entered into a phase 2 clinical trial to assess its antiinflammatory properties in PD.

Calcium channel blockade

A sustained-release formulation of isradipine, an L-type calcium channel blocker, is being studied in a phase 2 clinical trial for the treatment of early PD; experimental evidence in animals suggests that it may be neuroprotective against PD.

Uric acid elevation

Urate concentration in the cerebrospinal fluid predicts progression of PD, with higher levels associated with slower progression of disease.20 Urate may delay oxidative destruction of dopaminergic neurons that occurs with progression of PD. Pharmacologic elevation of uric acid is being explored as a treatment option in PD.

ELECTRODES, VECTORS, AND STEM CELLS

Deep brain stimulation

Reprinted with permission from Movement Disorders (Espay AJ, et al. Early versus delayed bilateral subthalamic deep brain stimulation for Parkinson’s disease: a decision analysis. Mov Disord 2010; 25:1456–1463). Copyright © 2010 Movement Disorder Society.
Figure 2. In a comparison of early versus late subthalamic nucleus deep brain stimulation (STN DBS), the annual rate of progression of both cognitive (A) and motor (B) decline was slower when STN DBS was administered earlier in the course of Parkinson disease (PD). Late STN DBS is favored if the annual rate of motor progression is greater than 25%, but this is an unrealistic scenario for PD.21
Deep brain stimulation (DBS) is currently used as a treatment for advanced PD (patients suffering from levodopainduced motor complications), but it might also slow the progression of cognitive and motor decline in earlier stages of PD. The annual rate of progression of both cognitive and motor decline was slower when DBS was administered earlier in the course of PD (off time on levodopa of about 2 hours) versus in a later stage of PD (off time on levodopa of about 4 hours) (Figure 2).21 The strategy is being tested further in clinical trials of early PD.

Stem cell therapy

Stem cells obtained from blastocytes, fibroblasts, bone marrow, or the adult, embryonic, or fetal central nervous system through “molecular alchemy” can form dopaminergic neuroblasts. Given the high cost and potential risks of stem cell therapy, it must be proven superior to DBS to be considered an option for early PD. Several practical problems act as hurdles to successful stem cell therapy. Efficient generation of dopamine-producing neurons and successful grafting are required. Tumor growth is a risk. Involuntary movements have been observed in some patients who received fetal implants. A limitation of stem cell therapy is that it will only affect those aspects of PD that are dependent on dopamine.

Gene therapy

Gene delivery of the growth factor analogue adeno-associated type-2 vector (AAV2)-neurturin has been investigated in patients with advanced PD. When surgically placed inside a neuron, neurturin enhances neuron vitality, enabling it to better fight oxidative stress and other attacks. It fared no better than sham surgery on changes in UPDRS motor score at 12 months in a randomized trial.22 A few patients enrolled in this trial have been followed for longer than 12 months, at which time the mean change in motor scores appears to favor the group assigned to gene delivery of AAV2-neurturin. A phase 1/2 trial is investigating the safety and efficacy of bilateral intraputaminal and intranigral administration of neurturin.

SUMMARY

Levodopa is a legitimate choice for the treatment of early PD. Two MAO-B inhibitors, rasagiline and selegiline, have a symptomatic effect.

Long-acting oral and transdermal dopamine agonists are effective symptomatic therapies, but they also have an interesting array of side effects, making levodopa a reasonable alternative treatment sooner or later despite its dyskinetic effect. Potential neuroprotective effects remain to be identified.

Amantadine is sometimes overlooked as an option for treating early PD, but it has some special side effects including leg edema, livedo reticularis, and corneal edema. Amantadine does not cause orthostatic hypotension and is free of the side effects of excessive diurnal somnolence and impulse control disorders that are prevalent with dopamine agonists.

In the future, partial dopamine agonists and adenosine antagonists may provide us with additional symptomatic therapies. CoQ10, creatine, calcium channel blockers, and inosine, as well as NSAIDs, are being actively studied as potential disease-modifying agents. Further studies are likely to come from the use of NSAIDs.

Early DBS is a new avenue of investigation as a potential disease modifier. Stem cells are still being studied and limitations of sufficient production and potential tumor growth, among others, have delayed the institution of clinical trials. Gene therapy is an interesting additional treatment modality in active research.

Parkinson disease (PD) is a slowly progressive neurodegenerative disorder. Early PD, or stage 1 or 2 on the Unified Parkinson’s Disease Rating Scale (UPDRS), is characterized by mild symptoms, minimal to mild disability, and lack of postural instability or cognitive decline. The goal of therapy in PD is to help patients retain functional independence for as long as possible. Therapeutic choices in early PD are guided by the effect of symptoms on function and quality of life, consideration of complications associated with long-term levodopa, the likelihood of response fluctuations to levodopa, and the potential for a neuroprotective effect.

SYMPTOMATIC THERAPIES IN EARLY PD

Dopaminergic replacement therapy with levodopa is a legitimate choice for the treatment of early PD. Use of carbidopa-levodopa has been shown to slow the progression of PD in a dose-dependent manner as evidenced by a decrease in total score on the UPDRS in patients with early PD who were randomly assigned to receive carbidopa-levodopa compared with those who received a placebo.1

Alternatives to levodopa

There are several reasons to choose an alternative to levodopa for the treatment of early PD. The first is to postpone the development of levodopa-induced dyskinesias, which are linked to duration of levodopa treatment and total exposure to levodopa. The second is postponement of the “wearing-off” effect; that is, the reemergence of symptoms that occurs in some patients before their next scheduled dose of levodopa. Such reasoning applies to early PD patients with minimal or no disability and—in particular—to young-onset PD patients who tend to develop vigorous dyskinesias and dramatic wearing-off phenomena. Pharmacologic alternatives to levodopa in early PD include monoamine oxidase (MAO)-B inhibitors, amantadine, and dopamine agonists.

Reprinted with permission from The New England Journal of Medicine (Olanow CW, et al. A double-blind, delayed-start trial of rasagiline in Parkinson’s disease. N Engl J Med 2009; 361:168–178). © 2009 Massachusetts Medical Society.
Figure 1. Rasagiline at doses of 1 mg/day (A) and mg/day (B) slowed the rate of worsening of the Unified Parkinson’s Disease Rating Scale (UPDRS) score compared with placebo in patients with untreated Parkinson disease.2
MAO-B inhibitors. Two MAO-B inhibitors are approved by the US Food and Drug Administration for the treatment of PD: rasagiline and selegiline. These agents have a mildly symptomatic effect. In the Attenuation of Disease Progression with Azilect Given Once-daily (ADAGIO) trial, use of rasagiline at doses of 1 and 2 mg/d slowed the rate of worsening of the UPDRS score compared with placebo in patients with untreated PD (Figure 1).2 Patients were randomly assigned to an early-start group (rasagiline, 1 or 2 mg/d, or placebo for 72 weeks) or a late-start group (placebo for 36 weeks followed by rasagiline, 1 or 2 mg/d, or placebo for 36 weeks). The rate of change in the UPDRS score was slowed significantly with early treatment with rasagiline at a dosage of 1 mg/d, but not at 2 mg/d. Because the hierarchical primary end points for the ADAGIO trial were met only for the cohort receiving 1 mg of rasagiline early, it remains inconclusive whether rasagiline has a neuroprotective effect.

In a placebo-controlled study of selegiline in de novo early-phase PD, Pålhagen et al showed that selegiline monotherapy delayed the need for levodopa. When used in combination with levodopa, selegiline was able to slow the progression of PD as measured by the change in UPDRS total score.3

Amantadine. In an early study of 54 patients with PD, functional disability scores improved significantly with administration of amantadine 200 to 300 mg/d compared with placebo.4 A small subset of patients, perhaps 20% or less, who are treated with amantadine experience robust symptom improvement. Side effects of amantadine include hallucinations, edema, livedo reticularis, and anticholinergic effects. A more recently discovered potential side effect is corneal edema.

Dopamine agonists. Pramipexole (immediate-release [IR] and extended-release [ER]), and ropinirole IR and ER are dopamine agonists that have demonstrated disease-modifying effects and efficacy in improving PD symptoms.

Pramipexole ER administered once daily in early PD was shown to be superior to placebo on the mean UPDRS total score.5 Ropinirole ER produced mean plasma concentrations over 24 hours similar to those achieved with ropinirole IR, and showed noninferiority to ropinirole IR on efficacy measures in patients with de novo PD.5

The effective dosage range of pramipexole ER in early PD is 0.375 to 4.5 mg/d. Side effects include hallucinations, edema, excessive diurnal somnolence, and impulse control disorders (ie, pathologic gambling, hypersexuality, excessive craving for sweets). Compared with pramipexole IR, compliance is enhanced with the ER formulation because of ease of administration, but this formulation also is more expensive.

In early PD, the effective dosage range of ropinirole ER is 8 to 12 mg/d.6 The side effects are the same as with pramipexole ER with the same compliance advantage and cost disadvantage compared with the IR formulation.

Research indicates that dopamine agonists may have a neuroprotective effect. In two large clinical trials in which patients with PD were followed with an imaging marker of dopamine neuronal degeneration (using single-photon emission computed tomography or positron emission tomography), recipients of pramipexole7 or ropinirole8 showed slower neuronal deterioration compared with levodopa recipients. A counterargument to the neuroprotective theory is that these differences between the dopamine agonists and levodopa reflect neurotoxicity of levodopa rather than neuroprotection by dopamine agonists. The absence of a placebo comparison in both trials adds to the difficulty in drawing a conclusion, as some critics ascribed the differences between groups to downregulation of tracer binding with levodopa.

Nonergoline dopamine agonist. Transdermal rotigotine is a nonergot D1/D2/D3 agonist. Higher doses produce higher plasma levels of rotigotine, which remain steady over the 24-hour dosing interval.9 Transdermal rotigotine has demonstrated effectiveness in early PD in several clinical trials.10,11 The patch, applied once daily, provides a constant release of medication. Removing the patch immediately interrupts drug administration.

Rotigotine patches must be refrigerated to prevent crystallization, a requirement that has delayed the product’s arrival on the market. The patch is reputed to be difficult to peel from its backing and apply. Skin reactions are a side effect, and nonergot side effects are possible. Despite these drawbacks, transdermal rotigotine represents a convenient option for perioperative management of PD and in patients with dysphagia.

Exercise. Exercise has symptomatic and possibly neuroprotective benefits in PD, supporting its use as an additional medical measure. Evidence supports the value of treadmill walking and high-impact exercise in improving stride length, quality of life, and motor response to levodopa.

 

 

SYMPTOMATIC THERAPIES: THE FUTURE

Partial dopamine agonists

Pardoprunox is a partial dopamine agonist with full 5-HT1A–agonist activity. A partial dopamine agonist acts in two ways: (1) It stimulates dopamine production in brain regions with low dopamine tone, and (2) it has dopamine antagonist activity under circumstances of high dopamine sensitivity, theoretically avoiding overstimulation of dopamine receptors. Because it inhibits excessive dopamine effect, pardoprunox may prevent dyskinesia. In addition, because pardoprunox has serotonin agonist activity, it may also act as an antidepressant.

In a phase 2 study, significantly more patients randomized to pardoprunox had a 30% or greater reduction in UPDRS motor score compared with placebo at end-of-dose titration (35.8% for pardoprunox vs 15.7% for placebo; P = .0065) and at end point (50.7% for pardoprunox vs 15.7% for placebo; P < .0001).12

Adenosine A2A-receptor antagonists

Adenosine A2A receptors are located in the basal ganglia, primarily on gamma aminobutyric acid (GABA)–mediated enkephalin-expressing medium spiny neurons in the striatum. These receptors modulate dopamine transmission by opposing D2-receptor activity. The D2 pathway is an indirect pathway that promotes suppression of unnecessary movement.

Two A2A-receptor antagonists have demonstrated efficacy in clinical trials. Vipadenant has been proven effective as monotherapy in phase 2 clinical trials. Preladenant has been shown to improve “off time” as an adjunct to levodopa without increasing dyskinesia.

Safinamide

Safinamide, currently in phase 3 clinical trials, has three mechanisms of action. It is an inhibitor of dopamine reuptake, a reversible inhibitor of MAO-B, and an inhibitor of excessive glutamate release. The addition of safinamide to a stable dose of a single dopamine agonist in patients with early PD resulted in improvement of motor symptoms and cognitive function.13,14

NEUROPROTECTIVE STRATEGIES UNDER INVESTIGATION

Four neuroprotective strategies are under study: enhanced mitochondrial function, antiinflammatory mechanisms, calcium channel blockade, and uric acid elevation.

Enhanced mitochondrial function

Creatine has generated interest as a disease-modifying agent in response to preclinical data showing that it could enhance mitochondrial function and prevent mitochondrial loss in the brain in models of PD. Creatine is now the subject of a large phase 3 National Institutes of Health–sponsored clinical trial in patients with early-stage PD.15

Coenzyme Q10 (CoQ 10) exhibited a trend for neuroprotection at 1,200 mg/d, lowering the total mean UPDRS score compared with placebo in a 16-month study.16 Current efforts are directed at determining whether 1,200 or 2,400 mg/d of CoQ10 are neuroprotective. A nanoparticulate form of CoQ10, 100 mg three times a day, has been shown to produce plasma levels of CoQ10 equivalent to those produced by 1,200-mg doses of the standard form.17 CoQ10 is free of symptomatic effects.

Antiinflammatory mechanisms

Parkinson disease may have an important inflammatory component. A meta-analysis of seven studies showed an overall hazard ratio of 0.85 for development of PD in users of nonaspirin nonsteroidal antiinflammatory drugs (NSAIDs), with each of the seven studies demonstrating a hazard ratio less than 1.18 A similar meta-analysis showed no such association.19 Further study is warranted.

The antidiabetic agent pioglitazone, shown in mice to prevent dopaminergic nigral cell loss, has been entered into a phase 2 clinical trial to assess its antiinflammatory properties in PD.

Calcium channel blockade

A sustained-release formulation of isradipine, an L-type calcium channel blocker, is being studied in a phase 2 clinical trial for the treatment of early PD; experimental evidence in animals suggests that it may be neuroprotective against PD.

Uric acid elevation

Urate concentration in the cerebrospinal fluid predicts progression of PD, with higher levels associated with slower progression of disease.20 Urate may delay oxidative destruction of dopaminergic neurons that occurs with progression of PD. Pharmacologic elevation of uric acid is being explored as a treatment option in PD.

ELECTRODES, VECTORS, AND STEM CELLS

Deep brain stimulation

Reprinted with permission from Movement Disorders (Espay AJ, et al. Early versus delayed bilateral subthalamic deep brain stimulation for Parkinson’s disease: a decision analysis. Mov Disord 2010; 25:1456–1463). Copyright © 2010 Movement Disorder Society.
Figure 2. In a comparison of early versus late subthalamic nucleus deep brain stimulation (STN DBS), the annual rate of progression of both cognitive (A) and motor (B) decline was slower when STN DBS was administered earlier in the course of Parkinson disease (PD). Late STN DBS is favored if the annual rate of motor progression is greater than 25%, but this is an unrealistic scenario for PD.21
Deep brain stimulation (DBS) is currently used as a treatment for advanced PD (patients suffering from levodopainduced motor complications), but it might also slow the progression of cognitive and motor decline in earlier stages of PD. The annual rate of progression of both cognitive and motor decline was slower when DBS was administered earlier in the course of PD (off time on levodopa of about 2 hours) versus in a later stage of PD (off time on levodopa of about 4 hours) (Figure 2).21 The strategy is being tested further in clinical trials of early PD.

Stem cell therapy

Stem cells obtained from blastocytes, fibroblasts, bone marrow, or the adult, embryonic, or fetal central nervous system through “molecular alchemy” can form dopaminergic neuroblasts. Given the high cost and potential risks of stem cell therapy, it must be proven superior to DBS to be considered an option for early PD. Several practical problems act as hurdles to successful stem cell therapy. Efficient generation of dopamine-producing neurons and successful grafting are required. Tumor growth is a risk. Involuntary movements have been observed in some patients who received fetal implants. A limitation of stem cell therapy is that it will only affect those aspects of PD that are dependent on dopamine.

Gene therapy

Gene delivery of the growth factor analogue adeno-associated type-2 vector (AAV2)-neurturin has been investigated in patients with advanced PD. When surgically placed inside a neuron, neurturin enhances neuron vitality, enabling it to better fight oxidative stress and other attacks. It fared no better than sham surgery on changes in UPDRS motor score at 12 months in a randomized trial.22 A few patients enrolled in this trial have been followed for longer than 12 months, at which time the mean change in motor scores appears to favor the group assigned to gene delivery of AAV2-neurturin. A phase 1/2 trial is investigating the safety and efficacy of bilateral intraputaminal and intranigral administration of neurturin.

SUMMARY

Levodopa is a legitimate choice for the treatment of early PD. Two MAO-B inhibitors, rasagiline and selegiline, have a symptomatic effect.

Long-acting oral and transdermal dopamine agonists are effective symptomatic therapies, but they also have an interesting array of side effects, making levodopa a reasonable alternative treatment sooner or later despite its dyskinetic effect. Potential neuroprotective effects remain to be identified.

Amantadine is sometimes overlooked as an option for treating early PD, but it has some special side effects including leg edema, livedo reticularis, and corneal edema. Amantadine does not cause orthostatic hypotension and is free of the side effects of excessive diurnal somnolence and impulse control disorders that are prevalent with dopamine agonists.

In the future, partial dopamine agonists and adenosine antagonists may provide us with additional symptomatic therapies. CoQ10, creatine, calcium channel blockers, and inosine, as well as NSAIDs, are being actively studied as potential disease-modifying agents. Further studies are likely to come from the use of NSAIDs.

Early DBS is a new avenue of investigation as a potential disease modifier. Stem cells are still being studied and limitations of sufficient production and potential tumor growth, among others, have delayed the institution of clinical trials. Gene therapy is an interesting additional treatment modality in active research.

References
  1. Fahn S, Oakes D, Shoulson I, et al. Levodopa and the progression of Parkinson’s disease. N Engl J Med 2004; 351:24982508.
  2. Olanow CW, Rascol O, Hauser R, et al. A double-blind, delayed-start trial of rasagiline in Parkinson’s disease. N Engl J Med 2009; 361:12681278.
  3. Pålhagen S, Heinonen E, Hägglund J, et al. Selegiline slows the progression of the symptoms of Parkinson disease. Neurology 2006; 66:12001206.
  4. Barbeau A, Mars H, Botez MI, Joubert M. Amantadine-HCl (Symmetrel) in the management of Parkinson’s disease: a double-blind cross-over study. Can Med Assoc J 1971; 105:4246.
  5. Hauser RA, Schapira AH, Rascol O, et al. Randomized, double-blind, multicenter evaluation of pramipexole extended release once daily in early Parkinson’s disease. Mov Disord 2010; 25:25422549.
  6. Onofrj M, Bonanni L, De Angelis MV, Anzellotti F, Ciccocioppo F, Thomas A. Long half-life and prolonged-release dopamine receptor antagonists: a review of ropinirole prolonged-release studies. Parkinsonism Relat Disord 2009; 15 (suppl 4):S85S92.
  7. Parkinson Study Group. Dopamine transporter brain imaging to assess the effects of pramipexole vs levodopa on Parkinson disease progression. JAMA 2002; 287:16531661.
  8. Whone AL, Watts RL, Stoessl AJ, et al. Slower progression of Parkinson’s disease with ropinirole versus levodopa: the REAL-PET study. Ann Neurol 2003; 54:93101.
  9. Reichmann H. Transdermal delivery of dopamine receptor agonists. Parkinsonism Relat Disord 2009; 15 (suppl 4):S93S96.
  10. Parkinson Study Group. A controlled trial of rotigotine monotherapy in early Parkinson’s disease. Arch Neurol 2003; 60:17211728.
  11. Watts RL, Jankovic J, Waters C, et al. Randomized, blind, controlled trial of transdermal rotigotine in early Parkinson disease. Neurology 2007; 68:272276.
  12. Bronzova J, Sampaio C, Hauser RA, et al. Double-blind study of pardoprunox, a new partial dopamine agonist, in early Parkinson’s disease. Mov Disord 2010; 25:738746.
  13. Stocchi F, Arnold G, Onofrj M, et al; Safinamide Parkinson’s Study Group. Improvement of motor function in early Parkinson disease by safinamide. Neurology 2004; 63:746748.
  14. Schapira AHV. Safinamide in the treatment of Parkinson’s disease. Expert Opin Pharmacother 2010; 11:22612268.
  15. NINDS NET-PD Investigators. A randomized, double-blind, futility clinical trial of creatine and minocycline in early Parkinson disease. Neurology 2006; 66:664671.
  16. Shults CW, Oakes D, Kieburtz K; the Parkinson Study Group. Effects of coenzyme q10 in early Parkinson disease: evidence of slowing of the functional decline. Arch Neurol 2002; 59:15411550.
  17. Storch A, Jost W, Vieregge P, et al. Randomized, double-blind, placebo-controlled trial on symptomatic effects of coenzyme q(10) in Parkinson disease. Arch Neurol 2007; 64:938944.
  18. Gagne JJ, Power MC. Anti-inflammatory drugs and risk of Parkinson disease: a meta-analysis. Neurology 2010; 74:9951002.
  19. Samii A, Etminan M, Wiens MO, Jafari S. NSAID use and the risk of Parkinson’s disease: systematic review and meta-analysis of observational studies. Drugs Aging 2009; 26:769779.
  20. Ascherio A, LeWitt PA, Xu K, et al; Parkinson Study Group DATATOP Investigators. Urate as a predictor of the rate of clinical decline in Parkinson disease. Arch Neurol 2009; 66:14601468.
  21. Espay AJ, Vaughan JE, Marras C, Fowler R, Eckman MH. Early versus delayed bilateral subthalamic deep brain stimulation for Parkinson’s disease: a decision analysis. Mov Disord 2010; 25:14561463.
  22. Marks WJ, Bartus RT, Siffert J, et al. Gene delivery of AAV2-neurturin for Parkinson’s disease: a double-blind, randomised, controlled trial. Lancet Neur 2010; 9:11641172.
References
  1. Fahn S, Oakes D, Shoulson I, et al. Levodopa and the progression of Parkinson’s disease. N Engl J Med 2004; 351:24982508.
  2. Olanow CW, Rascol O, Hauser R, et al. A double-blind, delayed-start trial of rasagiline in Parkinson’s disease. N Engl J Med 2009; 361:12681278.
  3. Pålhagen S, Heinonen E, Hägglund J, et al. Selegiline slows the progression of the symptoms of Parkinson disease. Neurology 2006; 66:12001206.
  4. Barbeau A, Mars H, Botez MI, Joubert M. Amantadine-HCl (Symmetrel) in the management of Parkinson’s disease: a double-blind cross-over study. Can Med Assoc J 1971; 105:4246.
  5. Hauser RA, Schapira AH, Rascol O, et al. Randomized, double-blind, multicenter evaluation of pramipexole extended release once daily in early Parkinson’s disease. Mov Disord 2010; 25:25422549.
  6. Onofrj M, Bonanni L, De Angelis MV, Anzellotti F, Ciccocioppo F, Thomas A. Long half-life and prolonged-release dopamine receptor antagonists: a review of ropinirole prolonged-release studies. Parkinsonism Relat Disord 2009; 15 (suppl 4):S85S92.
  7. Parkinson Study Group. Dopamine transporter brain imaging to assess the effects of pramipexole vs levodopa on Parkinson disease progression. JAMA 2002; 287:16531661.
  8. Whone AL, Watts RL, Stoessl AJ, et al. Slower progression of Parkinson’s disease with ropinirole versus levodopa: the REAL-PET study. Ann Neurol 2003; 54:93101.
  9. Reichmann H. Transdermal delivery of dopamine receptor agonists. Parkinsonism Relat Disord 2009; 15 (suppl 4):S93S96.
  10. Parkinson Study Group. A controlled trial of rotigotine monotherapy in early Parkinson’s disease. Arch Neurol 2003; 60:17211728.
  11. Watts RL, Jankovic J, Waters C, et al. Randomized, blind, controlled trial of transdermal rotigotine in early Parkinson disease. Neurology 2007; 68:272276.
  12. Bronzova J, Sampaio C, Hauser RA, et al. Double-blind study of pardoprunox, a new partial dopamine agonist, in early Parkinson’s disease. Mov Disord 2010; 25:738746.
  13. Stocchi F, Arnold G, Onofrj M, et al; Safinamide Parkinson’s Study Group. Improvement of motor function in early Parkinson disease by safinamide. Neurology 2004; 63:746748.
  14. Schapira AHV. Safinamide in the treatment of Parkinson’s disease. Expert Opin Pharmacother 2010; 11:22612268.
  15. NINDS NET-PD Investigators. A randomized, double-blind, futility clinical trial of creatine and minocycline in early Parkinson disease. Neurology 2006; 66:664671.
  16. Shults CW, Oakes D, Kieburtz K; the Parkinson Study Group. Effects of coenzyme q10 in early Parkinson disease: evidence of slowing of the functional decline. Arch Neurol 2002; 59:15411550.
  17. Storch A, Jost W, Vieregge P, et al. Randomized, double-blind, placebo-controlled trial on symptomatic effects of coenzyme q(10) in Parkinson disease. Arch Neurol 2007; 64:938944.
  18. Gagne JJ, Power MC. Anti-inflammatory drugs and risk of Parkinson disease: a meta-analysis. Neurology 2010; 74:9951002.
  19. Samii A, Etminan M, Wiens MO, Jafari S. NSAID use and the risk of Parkinson’s disease: systematic review and meta-analysis of observational studies. Drugs Aging 2009; 26:769779.
  20. Ascherio A, LeWitt PA, Xu K, et al; Parkinson Study Group DATATOP Investigators. Urate as a predictor of the rate of clinical decline in Parkinson disease. Arch Neurol 2009; 66:14601468.
  21. Espay AJ, Vaughan JE, Marras C, Fowler R, Eckman MH. Early versus delayed bilateral subthalamic deep brain stimulation for Parkinson’s disease: a decision analysis. Mov Disord 2010; 25:14561463.
  22. Marks WJ, Bartus RT, Siffert J, et al. Gene delivery of AAV2-neurturin for Parkinson’s disease: a double-blind, randomised, controlled trial. Lancet Neur 2010; 9:11641172.
Page Number
S3-S7
Page Number
S3-S7
Publications
Publications
Article Type
Display Headline
Managing the patient with newly diagnosed Parkinson disease
Display Headline
Managing the patient with newly diagnosed Parkinson disease
Citation Override
Cleveland Clinic Journal of Medicine 2012 July:79(suppl 2):S3-S7
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media

Off spells and dyskinesias: Pharmacologic management of motor complications

Article Type
Changed
Mon, 10/01/2018 - 15:33
Display Headline
Off spells and dyskinesias: Pharmacologic management of motor complications

Dopaminergic treatment is extremely beneficial in inducing symptom improvement in early Parkinson disease (PD). Patients typically experience a smooth and even response to the early stages of levodopa treatment. With disease progression, however, the effect of levodopa begins to weaken approximately 4 hours after each dose, leaving patients anticipating the need for their next dose and vulnerable to motor fluctuations and dyskinesias.

Motor fluctuations refer to the unanticipated loss of effect of a given dose of levodopa; instead of a smooth, predictable symptomatic benefit, the patient may lose benefit earlier than usual (termed “wearing off”) or may suddenly switch from “on” (symptoms controlled) to “off” (symptoms return). Dyskinesias, or involuntary movements, occur when dopamine levels are too high.

Motor complications are a major cause of disability in PD. They affect 60% to 90% of PD patients after 5 to 10 years of treatment. Moreover, in one study of 143 PD patients, motor complications diminished quality of life; the most strongly affected dimensions were mobility, activities of daily living, communication, and stigma.1

PATHOGENESIS OF MOTOR COMPLICATIONS

Under physiologic conditions, dopamine stimulation of the striatal dopamine receptors occurs in a sustained fashion. In early PD, the pool of remaining neurons of the substantia nigra is believed to be sufficiently active to smooth out changes in levodopa levels, providing a relatively constant amount of dopamine. Many PD patients therefore have several years of trouble-free treatment following diagnosis. In the advanced disease states, however, the number of presynaptic dopaminergic neurons progressively decreases. With fewer dopaminergic neurons, a constant dopaminergic concentration cannot be sustained. As PD advances, the progressive loss of dopaminergic neurons leads to impaired dopamine storage. Thus, the buffering capacity of dopaminergic neurons decreases and synaptic dopamine levels begin to reflect systemic or exogenous levodopa levels.

Figure. Pathogenesis of motor complications related to the Parkinson disease (PD) process and levodopa therapy. As PD advances (left), the progressive loss of dopaminergic neurons associated with nigrostriatal damage leads to impaired dopamine storage and clearance. This reduces the buffering capacity of dopaminergic neurons, causing early wearing off. During chronic levodopa treatment (right), pulsatile dopaminergic stimulation causes changes in postsynaptic receptors, referred to as “priming,” that increase the responsiveness of the receptors. The increased response results in severe levodopa-induced dyskinesias and on-off fluctuations. These postsynaptic changes are mediated through postsynaptic dopamine D1 receptors and N-methyl-d-aspartate glutamate receptors.
With disease progression, the “honeymoon” phase diminishes, and most PD patients begin to develop motor complications after 5 or more years. At this stage, medications frequently need to be adjusted, which can be a complex task for the physician and patient. The schema for the pathogenesis of motor complications related to the disease process and chronic levodopa treatment are depicted in the Figure.

According to current views, the total motor response to levodopa results from the combination of endogenous dopamine production along with the short-duration response (SDR) and the long-duration response (LDR) to exogenous levodopa.2 The SDR represents an improvement in parkinsonian symptoms and signs, lasting minutes to hours, that is closely related to the rise and fall of plasma levodopa concentrations. The SDR parallels the fluctuations in motor response and has received the most attention in the literature. The LDR is an improvement in parkinsonism that builds up over days and likewise decays over days. The LDR decays more rapidly in severely affected patients. Negative response, or “super off,” is a transient worsening of motor function to below the baseline level that may occur as the effects of the SDR dissipate.

The proportions of the SDR and LDR can vary according to disease progression. The LDR is more prominent in early stages, accounting for the stable response seen in the honeymoon period of treatment.

Peripheral factors

Additional peripheral factors such as changes in gastric motility and absorption contribute to motor complications. Levodopa is transported by a saturable active transporter system, the large neutral amino acid system, in the gut, and across the blood-brain barrier. Levodopa absorption is thus affected by food intake, especially protein. Levodopa and dietary amino acids compete with each other for absorption at the intestinal and blood-brain levels. Levodopa and other dopaminergic therapies further chronically reduce gastric emptying.

Pulsatile dopamine stimulation

The latency from the time of levodopa administration to the onset of motor improvement is typically 30 to 90 minutes. Latency is longer in late stages when the striatal buffer is weakened and the plasma concentration of levodopa fluctuates.

TYPES OF MOTOR FLUCTUATIONS

Fluctuating motor response in levodopa-treated patients refers to clinically apparent oscillations in motor function. Management of the fluctuating response may require frequent daily dosing of levodopa.

Motor fluctuations in PD take four forms: wearing off, off, delayed on/no on, and dyskinesias.

Wearing off

Wearing off refers to the premature loss of benefit from a given dose of levodopa, causing a predictable return of parkinsonian symptoms (bradykinesia, tremors, rigidity, and gait problems) in advance of the next scheduled dose. Observed in early and moderate PD, wearing off is the most common type of motor fluctuation. Its pathophysiology relates to disease progression and pharmacokinetics of levodopa. It can be sudden or gradual, predictable or unpredictable.

Off state

The off state is the unpredictable reappearance of parkinsonian symptoms at a time when central levels of antiparkinsonian drugs are expected to be within the target therapeutic range. Such symptoms include pain, stiffness, paresthesia, cognitive symptoms (depression, anxiety, difficulty with concentration, and mental slowing), inner restlessness, and inner tremulousness. The off state can be sudden or gradual, predictable or unpredictable.

Delayed on/no on

Delayed on is a prolongation of the time required for the central antiparkinsonian drug effect to appear. As the disease progresses, wearing off becomes more complicated and more unpredictable. The dosing responses vary, and patients sometimes report delayed on. The causes of delayed on or no on can be an insufficient dose, dosing with high-protein meals, or delayed gastric emptying. Metoclopramide or domperidone can help with gastric emptying. Metoclopramide can cross the blood-brain barrier and thus may cause adverse effects related to dopaminergic blockade; domperidone does not cross the blood-brain barrier.

Dyskinesias

Dyskinesias are hyperkinetic movements related to dopaminergic effects that are greater or less than the therapeutic threshold. They are common with long-term levodopa therapy and have three patterns:

Off dystonia occurs when levodopa concentrations are low and the SDR has dissipated. Dystonic states may be a manifestation of too little or too much levodopa; differentiating the two is important. Off dystonia occurs mostly in the early mornings, when plasma levodopa levels are low, and mostly involves the more affected side first.

Peak-dose dyskinesia, which occurs during the SDR, is the most common type of dykinesia and is related to peak plasma levodopa levels. It is characterized by stereotypic, choreic abnormal movements involving the head, neck, trunk, and limbs, and possibly hemidyskinesia in young-onset PD. Peak-dose dyskinesias are sometimes severe enough to be disabling.

Diphasic dyskinesias are stereotyped, dystonic, or choreic movements that occur at the beginning of the SDR and again as the SDR dissipates. They predominantly affect the legs and spare the trunk, neck, and arms.

 

 

TREATMENT OF OFF AND WEARING OFF

Increasing dopaminergic stimulation is the backbone of treatment of off periods or wearing off. The strategies to increase dopaminergic stimulation include addressing food and tolerance issues and adding a monoamine oxidase B (MAO-B) inhibitor or a catechol-O-methyltransferase (COMT) inhibitor such as entacapone or tolcapone to the regimen (Table). If the patient is already taking levodopa or a dopamine agonist, the dosage can be increased; or, levodopa can be added to a dopamine agonist regimen and vice versa.

Food and tolerance

The patient should not take levodopa with protein-containing meals, particularly if his or her PD is at an advanced stage. If excessive nausea, vomiting, or lightheadedness prevents the patient from taking an adequate dose, adding carbidopa (up to 75 mg) to the regimen will be helpful.

MAO-B inhibitors

By inhibiting one of the central dopamine catabolic pathways, MAO-B inhibitors (selegiline, rasagiline, and Zydis selegiline) prolong the half-life of dopamine in the brain and increase on time.

Improvement in off time with rasagiline is comparable to that seen with the COMT inhibitor entacapone. In an 18-week, double-blind trial of 687 patients randomized to receive once-daily rasagiline, entacapone, or placebo as an adjunct to levodopa, both rasagiline and entacapone reduced off time by 1.2 hours compared with placebo.3

In a 26-week placebo-controlled study, rasagiline decreased off time by 29% when added to levodopa in patients with PD and motor fluctuations, compared with a 15% reduction in the placebo group.4 This study confirmed the benefit of adding rasagiline to the regimens of patients who were already optimally treated with levodopa, dopamine agonists, amantadine, anticholinergics, and entacapone before enrolling in the study.

An orally disintegrating selegiline (Zydis selegiline) tablet is particularly useful for patients who have difficulty swallowing. The bioavailability of Zydis selegiline is 80% compared with 10% for selegiline, resulting in faster absorption. Pregastric absorption of Zydis selegiline avoids extensive first-pass metabolism in the liver and, therefore, the concentration of amphetamine-like metabolites is much lower.

In a 3-month, placebo-controlled study of patients with PD who were experiencing levodopa-related motor fluctuations, Zydis selegiline was associated with a 2.2-hour reduction in the total number of off hours compared with 0.6 hours in the placebo group, and dyskinesia-free on hours increased by 1.8 hours.5

The use of MAO-B inhibitors with tricyclic antidepressants or selective serotonin reuptake inhibitors has been reported to induce the serotonin syndrome by activation of 5HT1a and 5HT2 receptors. Serotonin syndrome is a potentially life-threatening accumulation of serotonin that can cause encephalopathy, severe rigidity of the legs, dysautonomia (diarrhea, mydriasis, and excessive lacrimation), myoclonus, hyperreflexia, and seizures.

COMT inhibitors

Catechol-O-methyltransferase inhibitors (entacapone and tolcapone) block peripheral degradation of levodopa. Tolcapone also blocks central degradation of levodopa and dopamine. These mechanisms increase central levodopa and dopamine levels and prolong levodopa half-life and bioavailability. Tolcapone has more powerful COMT inhibition than entacapone because tolcapone crosses the blood-brain barrier and inhibits the peripheral and central pathways of levodopa degradation. Use of COMT inhibitors can increase daily on time, but diarrhea is a common side effect and leads to withdrawal of these agents in about 3% of patients.

Tolcapone-treated patients show significant improvement in off time with improvement in motor fluctuations.6 Because tolcapone causes rare instances of fulminant hepatitis, liver function needs to be monitored every other week. For this reason, tolcapone should be reserved for patients in whom other treatments, including entacapone, have failed.

Controlled-release levodopa

Controlled-release levodopa was developed to provide more constant delivery of levodopa to the striatum. The benefit of controlled-release levodopa is only mild, however, as absorption of this formulation is variable. In advanced PD cases, the effects of controlled-release levodopa are more unpredictable than those with standard levodopa. Controlled-release levodopa is effective in patients with less severe wearing off, but it is not as effective in patients with a less predictable pattern of fluctuations.

Dopamine agonists

Dopamine agonists (pramipexole, ropinirole, apomorphine, and bromocriptine) have shown beneficial effects as adjunctive therapy to reduce wearing off. Side effects of dopamine agonists include ankle edema, hallucinations, somnolence, and impulse control disorders. These side effects should be discussed with patients before instituting therapy, and therapy should be discontinued if any of them occur.

In patients with advanced PD, pramipexole was shown to improve motor function during on and off periods, decrease the total off time, and decrease the severity of off time. Further, a larger reduction in the dosage of levodopa was possible in the pramipexole-treated patients than in the placebo-treated patients.7

In a comparison of pramipexole with levodopa on the end point of motor complications of PD in 300 patients, the incidences of wearing off and dyskinesia were significantly lower in the patients randomized to pramipexole with follow-up over 4 years.8 Only 25% of patients initially treated with pramipexole exhibited dyskinesia compared with 54% of patients initially treated with levodopa. Forty-seven percent of patients in the pramipexole group experienced wearing off compared with 63% initially treated with levodopa. Pramipexole is available as tablets ranging from 0.125 mg to 1.5 mg in size. It is given in three divided daily doses with gradual increments of 0.25 mg three times a day every week. Pramipexole is now also available in an extended-release formulation for once-a-day dosing in tablets ranging in size from 0.375 mg to 4.5 mg.

Ropinirole adds clinical benefit in PD patients with motor fluctuations and also permits a reduction in the dosage of levodopa.9

In one study, ropinirole monotherapy was compared with levodopa therapy in 268 patients with early PD. By the end of the 5-year study, 45% of the levodopa patients experienced dyskinesias versus 20% of the ropinirole patients.10

Ropinirole is available as tablets ranging in size from 0.25 mg to 5 mg. It is now also available in an extended-release (XL) formulation, with tablet sizes ranging from 2 mg to 12 mg. Ropinirole XL is taken once a day.

Bromocriptine is an old ergot-derived dopamine agonist that has also been studied for monotherapy and add-on treatment in PD. Due to the potential risks of pulmonary, retroperitoneal, and heart valve fibrosis, bromocriptine is not commonly used.

Apomorphine was approved by the US Food and Drug Administration in 2004 as an acute, intermittent, subcutaneous injection for the treatment of hypomobility off episodes (end-of-dose wearing off and unpredictable on-off episodes) associated with advanced PD. Apomorphine has been shown to be beneficial in patients with unpredictable off periods.11 Its onset of action is 10 to 15 minutes, and the effects of each dose last for 60 to 90 minutes. The best tolerated dose is 4 mg to 10 mg. Apomorphine appears to be most useful as as rescue medication in the refractory off periods with severe bradykinesia and unpredictable off periods.

 

 

TREATMENT OF DYSKINESIAS

Reduction of levodopa doses will reduce the frequency of dyskinesias, but at a cost of worsened parkinsonism and increased numbers of off periods. An alternative is to spread out the doses of levodopa (more frequent smaller doses), but this practice has not achieved good results. Replacing levodopa with dopamine agonists can also reduce the frequency of dyskinesias, but control of PD symptoms is less optimal than with levodopa. Amantadine and clozapine both have been shown to reduce dyskinesias.

Amantadine

Amantadine is an N-methyl-d-aspartate antagonist with antidyskinesia effects. Metman et al12 demonstrated that amantadine reduced dyskinesia severity by 60%, without exacerbation of motor function, in a randomized placebo-controlled crossover study. Dose-response studies with amantadine have not been conducted, but 100 mg two or three times daily is used in practice. In some studies, a short duration of benefit has been a concern. Side effects of amantadine include leg edema, hallucinations, confusion, and rash.

Clozapine

Clozapine is an atypical antipsychotic that has been shown in open-label trials and a randomized, double-blind, placebo-controlled trial to reduce the duration and severity of levodopa-induced dyskinesias without worsening of parkinsonian features and with no change in motor fluctuation.13 No benefit of clozapine was observed during activation dyskinesia, however. Cloza pine carries the inconvenience of weekly blood draws to monitor for the development of agranulocytosis, which occurs rarely.

GAIT FREEZING

Gait freezing, most commonly a manifestation of off states, causes substantial disability. It has been thought to occur as a result of a loss of noradrenaline due to locus ceruleus degeneration. Improvement in gait freezing has been shown with apomorphine and methylphenidate.

CONCEPT OF CONTINUOUS DOPAMINE STIMULATION

Short-acting dopaminergic drugs have the potential for nonphysiologic pulsatile stimulation of postsynaptic receptors, leading to motor complications. Continuous dopaminergic stimulation to prevent this pulsatile stimulation would theoretically avoid motor complications.14 Continuous dopaminergic stimulation can be achieved by using the extended-release formulation of ropinirole or pramipexole or by continuous delivery of levodopa or dopamine agonists. Several double-blind controlled trials have shown that treatment with long-acting dopamine agonists lowers the risk of motor complications compared with short-acting levodopa treatment.

In 2005, Stocchi concluded that in patients with advanced PD, a continuous infusion of levodopa was more effective in reducing motor complications than standard oral formulations.15 The reduction in motor complications was attributed to avoidance of low plasma levodopa trough levels; motor complications were not affected by relatively high plasma levodopa concentrations. The authors of this study speculated that if oral levodopa could be given “in a manner that mirrors the pharmacokinetic pattern of infusion,” it might be able to reduce motor complications.

This hypothesis led to an interest in treatment with levodopa plus entacapone. A regimen of levodopa-carbidopa-entacapone, four times daily at 3.5-hour intervals, was compared with levodopa-carbidopa in 747 patients with early PD over 134 weeks.16 Initiating levodopa therapy with levodopa-carbidopa in combination with entacapone did not delay the induction of dyskinesia compared with levodopa-carbidopa alone. In fact, levodopa-carbidopa-entacapone was associated with a shorter time to onset and an increased frequency of dyskinesia compared with levodopa-carbidopa.

Potential future treatment options

An intrajejunal pump system delivers a constant-rate infusion of levodopa. A double-blind study of this system is being conducted in the United States. Implantation of the system is an invasive procedure with the potential for infection, kinking dislocation, and occlusion and reposition of the catheter.

Miniature pumps for continuous subcutaneous delivery of apomorphine, currently available only in Europe, have been shown to reverse dyskinesias and motor fluctuations. Limitations of the minipumps are the development of red itchy nodules, ulcerations, and abscesses at infusion sites.

Extended-release dopamine agonists

Extended-release formulations of the dopamine agonists ropinirole and pramipexole are easy to administer, and they maintain therapeutic plasma levels for up to 24 hours. They are unlikely to replace stronger continuous dopamine stimulation with levodopa and apomorphine.

SUMMARY

Motor complications in PD result from progression of the disease and limitations of levodopa. Although the effects of levodopa on PD eventually wane, leaving patients vulnerable to motor complications, clinicians should not undertreat patients.

Effective options for the management of motor complications include prolonging the efficacy of levodopa through the use of selective MAO-B inhibitors and COMT inhibitors as adjuncts to levodopa or continuous dopaminergic stimulation achieved by the use of long-acting dopamine agonists or continuous intraduodenal levodopa.

Emerging therapies will be more efficient for continuous delivery of dopaminergic drugs. Pump delivery systems and extended-release formulations have shown promise.

References
  1. Chapuis S, Ouchchane L, Metz O, Gerbaud L, Durif F. Impact of the motor complications of Parkinson’s disease on the quality of life. Mov Disord 2005; 20:224230.
  2. Nutt JG, Holford NHG. The response to levodopa in Parkinson’s disease: imposing pharmacological law and order. Ann Neurol 1996; 39:561573.
  3. Rascol O, Brooks DJ, Oertel W, et al. Rasagiline as an adjunct to levodopa in patients with Parkinson’s disease and motor fluctuations (LARGO, Lasting effect in Adjunct therapy with Rasagiline Given Once daily, study): a randomised, double-blind, parallel-group trial. Lancet 2005; 365:947954.
  4. Parkinson Study Group. A randomized placebo-controlled trial of rasagiline in levodopa-treated patients with Parkinson disease and motor fluctuations: the PRESTO study. Arch Neurol 2005; 62:241248.
  5. Waters CH, Sethi KD, Hauser RA, et al. Zydis selegiline reduces off time in Parkinson’s disease patients with motor fluctuations: a 3-month, randomized, placebo-controlled study. Mov Disord 2004; 19:426432.
  6. Rajput AH, Martin W, Saint-Hilaire MH, Dorflinger E, Pedder S. Tolcapone improves motor function in parkinsonian patients with the “wearing-off” phenomenon: a double-blind, placebo-controlled, multicenter trial. Neurology 1997; 49:10661071.
  7. Lieberman A, Ranhosky A, Korts D. Clinical evaluation of pramipexole in advanced Parkinson’s disease: results of a double-blind, placebo-controlled, parallel-group study. Neurology 1997; 49:162168.
  8. Parkinson Study Group. Pramipexole vs levodopa as initial treatment for Parkinson disease: a 4-year randomized controlled trial. JAMA 2004; 61:10441053.
  9. Lieberman A, Olanow CW, Sethi K, et al. A multicenter trial of ropinirole as adjunct treatment for Parkinson’s disease. Neurology 1998; 51:10571062.
  10. Rascol O, Brooks DJ, Korczyn AD, et al. A five-year study of the incidence of dyskinesia in patients with early Parkinson’s disease who were treated with ropinirole or levodopa. N Engl J Med 2000; 342:14841491.
  11. Dewey RB, Hutton JT, LeWitt PA, Factor SA. A randomized, double-blind, placebo-controlled trial of subcutaneously injected apomorphine for parkinsonian off-state events. Arch Neurol 2001; 58:13851392.
  12. Metman LV, Del Dotto P, van den Munckhof P, Fang J, Mouradian MM, Chase TN. Amantadine as treatment for dyskinesias and motor fluctuations in Parkinson’s disease. Neurology 1998; 50:13231326.
  13. Durif F, Debilly B, Galitzky M, et al. Clozapine improves dyskinesias in Parkinson disease: a double-blind, placebo-controlled study. Neurology 2004; 62:381388.
  14. Olanow CW, Obeso JA, Stocchi F. Continuous dopamine-receptor treatment of Parkinson’s disease: scientific rationale and clinical implications. Lancet Neurol 2006; 5:677687.
  15. Stocchi F, Vacca L, Ruggieri S, Olanow CW. Intermittent vs continuous levodopa administration in patients with advanced Parkinson disease: a clinical and pharmacokinetic study. Arch Neurol 2005; 62:905910.
  16. Stocchi F, Rascol O, Kieburtz K, et al. Initiating levodopa/carbidopa therapy with and without entacapone in early Parkinson disease: The STRIDE-PD study. Ann Neurol 2010; 68:1827.
Article PDF
Author and Disclosure Information

Tarannum S. Khan, MD
Neurologist, Movement Disorders Program, Cleveland Clinic Florida, Weston, FL

Correspondence: Tarannum S. Khan, MD, Dept. of Neurology, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd., Weston, FL 33331; khant@ccf.org

Dr. Khan reported that she has no financial interests or relationships that pose a potential conflict of interest with this article.

This article is based on Dr. Khan’s presentation at "The Annual Therapy Symposium on Movement Disorders for the Modern Clinician" held in Fort Lauderdale, Florida, on January 29, 2011. The article was drafted by Cleveland Clinic Journal of Medicine and was then reviewed, revised, and approved by Dr. Khan.

Publications
Page Number
S8-S13
Author and Disclosure Information

Tarannum S. Khan, MD
Neurologist, Movement Disorders Program, Cleveland Clinic Florida, Weston, FL

Correspondence: Tarannum S. Khan, MD, Dept. of Neurology, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd., Weston, FL 33331; khant@ccf.org

Dr. Khan reported that she has no financial interests or relationships that pose a potential conflict of interest with this article.

This article is based on Dr. Khan’s presentation at "The Annual Therapy Symposium on Movement Disorders for the Modern Clinician" held in Fort Lauderdale, Florida, on January 29, 2011. The article was drafted by Cleveland Clinic Journal of Medicine and was then reviewed, revised, and approved by Dr. Khan.

Author and Disclosure Information

Tarannum S. Khan, MD
Neurologist, Movement Disorders Program, Cleveland Clinic Florida, Weston, FL

Correspondence: Tarannum S. Khan, MD, Dept. of Neurology, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd., Weston, FL 33331; khant@ccf.org

Dr. Khan reported that she has no financial interests or relationships that pose a potential conflict of interest with this article.

This article is based on Dr. Khan’s presentation at "The Annual Therapy Symposium on Movement Disorders for the Modern Clinician" held in Fort Lauderdale, Florida, on January 29, 2011. The article was drafted by Cleveland Clinic Journal of Medicine and was then reviewed, revised, and approved by Dr. Khan.

Article PDF
Article PDF

Dopaminergic treatment is extremely beneficial in inducing symptom improvement in early Parkinson disease (PD). Patients typically experience a smooth and even response to the early stages of levodopa treatment. With disease progression, however, the effect of levodopa begins to weaken approximately 4 hours after each dose, leaving patients anticipating the need for their next dose and vulnerable to motor fluctuations and dyskinesias.

Motor fluctuations refer to the unanticipated loss of effect of a given dose of levodopa; instead of a smooth, predictable symptomatic benefit, the patient may lose benefit earlier than usual (termed “wearing off”) or may suddenly switch from “on” (symptoms controlled) to “off” (symptoms return). Dyskinesias, or involuntary movements, occur when dopamine levels are too high.

Motor complications are a major cause of disability in PD. They affect 60% to 90% of PD patients after 5 to 10 years of treatment. Moreover, in one study of 143 PD patients, motor complications diminished quality of life; the most strongly affected dimensions were mobility, activities of daily living, communication, and stigma.1

PATHOGENESIS OF MOTOR COMPLICATIONS

Under physiologic conditions, dopamine stimulation of the striatal dopamine receptors occurs in a sustained fashion. In early PD, the pool of remaining neurons of the substantia nigra is believed to be sufficiently active to smooth out changes in levodopa levels, providing a relatively constant amount of dopamine. Many PD patients therefore have several years of trouble-free treatment following diagnosis. In the advanced disease states, however, the number of presynaptic dopaminergic neurons progressively decreases. With fewer dopaminergic neurons, a constant dopaminergic concentration cannot be sustained. As PD advances, the progressive loss of dopaminergic neurons leads to impaired dopamine storage. Thus, the buffering capacity of dopaminergic neurons decreases and synaptic dopamine levels begin to reflect systemic or exogenous levodopa levels.

Figure. Pathogenesis of motor complications related to the Parkinson disease (PD) process and levodopa therapy. As PD advances (left), the progressive loss of dopaminergic neurons associated with nigrostriatal damage leads to impaired dopamine storage and clearance. This reduces the buffering capacity of dopaminergic neurons, causing early wearing off. During chronic levodopa treatment (right), pulsatile dopaminergic stimulation causes changes in postsynaptic receptors, referred to as “priming,” that increase the responsiveness of the receptors. The increased response results in severe levodopa-induced dyskinesias and on-off fluctuations. These postsynaptic changes are mediated through postsynaptic dopamine D1 receptors and N-methyl-d-aspartate glutamate receptors.
With disease progression, the “honeymoon” phase diminishes, and most PD patients begin to develop motor complications after 5 or more years. At this stage, medications frequently need to be adjusted, which can be a complex task for the physician and patient. The schema for the pathogenesis of motor complications related to the disease process and chronic levodopa treatment are depicted in the Figure.

According to current views, the total motor response to levodopa results from the combination of endogenous dopamine production along with the short-duration response (SDR) and the long-duration response (LDR) to exogenous levodopa.2 The SDR represents an improvement in parkinsonian symptoms and signs, lasting minutes to hours, that is closely related to the rise and fall of plasma levodopa concentrations. The SDR parallels the fluctuations in motor response and has received the most attention in the literature. The LDR is an improvement in parkinsonism that builds up over days and likewise decays over days. The LDR decays more rapidly in severely affected patients. Negative response, or “super off,” is a transient worsening of motor function to below the baseline level that may occur as the effects of the SDR dissipate.

The proportions of the SDR and LDR can vary according to disease progression. The LDR is more prominent in early stages, accounting for the stable response seen in the honeymoon period of treatment.

Peripheral factors

Additional peripheral factors such as changes in gastric motility and absorption contribute to motor complications. Levodopa is transported by a saturable active transporter system, the large neutral amino acid system, in the gut, and across the blood-brain barrier. Levodopa absorption is thus affected by food intake, especially protein. Levodopa and dietary amino acids compete with each other for absorption at the intestinal and blood-brain levels. Levodopa and other dopaminergic therapies further chronically reduce gastric emptying.

Pulsatile dopamine stimulation

The latency from the time of levodopa administration to the onset of motor improvement is typically 30 to 90 minutes. Latency is longer in late stages when the striatal buffer is weakened and the plasma concentration of levodopa fluctuates.

TYPES OF MOTOR FLUCTUATIONS

Fluctuating motor response in levodopa-treated patients refers to clinically apparent oscillations in motor function. Management of the fluctuating response may require frequent daily dosing of levodopa.

Motor fluctuations in PD take four forms: wearing off, off, delayed on/no on, and dyskinesias.

Wearing off

Wearing off refers to the premature loss of benefit from a given dose of levodopa, causing a predictable return of parkinsonian symptoms (bradykinesia, tremors, rigidity, and gait problems) in advance of the next scheduled dose. Observed in early and moderate PD, wearing off is the most common type of motor fluctuation. Its pathophysiology relates to disease progression and pharmacokinetics of levodopa. It can be sudden or gradual, predictable or unpredictable.

Off state

The off state is the unpredictable reappearance of parkinsonian symptoms at a time when central levels of antiparkinsonian drugs are expected to be within the target therapeutic range. Such symptoms include pain, stiffness, paresthesia, cognitive symptoms (depression, anxiety, difficulty with concentration, and mental slowing), inner restlessness, and inner tremulousness. The off state can be sudden or gradual, predictable or unpredictable.

Delayed on/no on

Delayed on is a prolongation of the time required for the central antiparkinsonian drug effect to appear. As the disease progresses, wearing off becomes more complicated and more unpredictable. The dosing responses vary, and patients sometimes report delayed on. The causes of delayed on or no on can be an insufficient dose, dosing with high-protein meals, or delayed gastric emptying. Metoclopramide or domperidone can help with gastric emptying. Metoclopramide can cross the blood-brain barrier and thus may cause adverse effects related to dopaminergic blockade; domperidone does not cross the blood-brain barrier.

Dyskinesias

Dyskinesias are hyperkinetic movements related to dopaminergic effects that are greater or less than the therapeutic threshold. They are common with long-term levodopa therapy and have three patterns:

Off dystonia occurs when levodopa concentrations are low and the SDR has dissipated. Dystonic states may be a manifestation of too little or too much levodopa; differentiating the two is important. Off dystonia occurs mostly in the early mornings, when plasma levodopa levels are low, and mostly involves the more affected side first.

Peak-dose dyskinesia, which occurs during the SDR, is the most common type of dykinesia and is related to peak plasma levodopa levels. It is characterized by stereotypic, choreic abnormal movements involving the head, neck, trunk, and limbs, and possibly hemidyskinesia in young-onset PD. Peak-dose dyskinesias are sometimes severe enough to be disabling.

Diphasic dyskinesias are stereotyped, dystonic, or choreic movements that occur at the beginning of the SDR and again as the SDR dissipates. They predominantly affect the legs and spare the trunk, neck, and arms.

 

 

TREATMENT OF OFF AND WEARING OFF

Increasing dopaminergic stimulation is the backbone of treatment of off periods or wearing off. The strategies to increase dopaminergic stimulation include addressing food and tolerance issues and adding a monoamine oxidase B (MAO-B) inhibitor or a catechol-O-methyltransferase (COMT) inhibitor such as entacapone or tolcapone to the regimen (Table). If the patient is already taking levodopa or a dopamine agonist, the dosage can be increased; or, levodopa can be added to a dopamine agonist regimen and vice versa.

Food and tolerance

The patient should not take levodopa with protein-containing meals, particularly if his or her PD is at an advanced stage. If excessive nausea, vomiting, or lightheadedness prevents the patient from taking an adequate dose, adding carbidopa (up to 75 mg) to the regimen will be helpful.

MAO-B inhibitors

By inhibiting one of the central dopamine catabolic pathways, MAO-B inhibitors (selegiline, rasagiline, and Zydis selegiline) prolong the half-life of dopamine in the brain and increase on time.

Improvement in off time with rasagiline is comparable to that seen with the COMT inhibitor entacapone. In an 18-week, double-blind trial of 687 patients randomized to receive once-daily rasagiline, entacapone, or placebo as an adjunct to levodopa, both rasagiline and entacapone reduced off time by 1.2 hours compared with placebo.3

In a 26-week placebo-controlled study, rasagiline decreased off time by 29% when added to levodopa in patients with PD and motor fluctuations, compared with a 15% reduction in the placebo group.4 This study confirmed the benefit of adding rasagiline to the regimens of patients who were already optimally treated with levodopa, dopamine agonists, amantadine, anticholinergics, and entacapone before enrolling in the study.

An orally disintegrating selegiline (Zydis selegiline) tablet is particularly useful for patients who have difficulty swallowing. The bioavailability of Zydis selegiline is 80% compared with 10% for selegiline, resulting in faster absorption. Pregastric absorption of Zydis selegiline avoids extensive first-pass metabolism in the liver and, therefore, the concentration of amphetamine-like metabolites is much lower.

In a 3-month, placebo-controlled study of patients with PD who were experiencing levodopa-related motor fluctuations, Zydis selegiline was associated with a 2.2-hour reduction in the total number of off hours compared with 0.6 hours in the placebo group, and dyskinesia-free on hours increased by 1.8 hours.5

The use of MAO-B inhibitors with tricyclic antidepressants or selective serotonin reuptake inhibitors has been reported to induce the serotonin syndrome by activation of 5HT1a and 5HT2 receptors. Serotonin syndrome is a potentially life-threatening accumulation of serotonin that can cause encephalopathy, severe rigidity of the legs, dysautonomia (diarrhea, mydriasis, and excessive lacrimation), myoclonus, hyperreflexia, and seizures.

COMT inhibitors

Catechol-O-methyltransferase inhibitors (entacapone and tolcapone) block peripheral degradation of levodopa. Tolcapone also blocks central degradation of levodopa and dopamine. These mechanisms increase central levodopa and dopamine levels and prolong levodopa half-life and bioavailability. Tolcapone has more powerful COMT inhibition than entacapone because tolcapone crosses the blood-brain barrier and inhibits the peripheral and central pathways of levodopa degradation. Use of COMT inhibitors can increase daily on time, but diarrhea is a common side effect and leads to withdrawal of these agents in about 3% of patients.

Tolcapone-treated patients show significant improvement in off time with improvement in motor fluctuations.6 Because tolcapone causes rare instances of fulminant hepatitis, liver function needs to be monitored every other week. For this reason, tolcapone should be reserved for patients in whom other treatments, including entacapone, have failed.

Controlled-release levodopa

Controlled-release levodopa was developed to provide more constant delivery of levodopa to the striatum. The benefit of controlled-release levodopa is only mild, however, as absorption of this formulation is variable. In advanced PD cases, the effects of controlled-release levodopa are more unpredictable than those with standard levodopa. Controlled-release levodopa is effective in patients with less severe wearing off, but it is not as effective in patients with a less predictable pattern of fluctuations.

Dopamine agonists

Dopamine agonists (pramipexole, ropinirole, apomorphine, and bromocriptine) have shown beneficial effects as adjunctive therapy to reduce wearing off. Side effects of dopamine agonists include ankle edema, hallucinations, somnolence, and impulse control disorders. These side effects should be discussed with patients before instituting therapy, and therapy should be discontinued if any of them occur.

In patients with advanced PD, pramipexole was shown to improve motor function during on and off periods, decrease the total off time, and decrease the severity of off time. Further, a larger reduction in the dosage of levodopa was possible in the pramipexole-treated patients than in the placebo-treated patients.7

In a comparison of pramipexole with levodopa on the end point of motor complications of PD in 300 patients, the incidences of wearing off and dyskinesia were significantly lower in the patients randomized to pramipexole with follow-up over 4 years.8 Only 25% of patients initially treated with pramipexole exhibited dyskinesia compared with 54% of patients initially treated with levodopa. Forty-seven percent of patients in the pramipexole group experienced wearing off compared with 63% initially treated with levodopa. Pramipexole is available as tablets ranging from 0.125 mg to 1.5 mg in size. It is given in three divided daily doses with gradual increments of 0.25 mg three times a day every week. Pramipexole is now also available in an extended-release formulation for once-a-day dosing in tablets ranging in size from 0.375 mg to 4.5 mg.

Ropinirole adds clinical benefit in PD patients with motor fluctuations and also permits a reduction in the dosage of levodopa.9

In one study, ropinirole monotherapy was compared with levodopa therapy in 268 patients with early PD. By the end of the 5-year study, 45% of the levodopa patients experienced dyskinesias versus 20% of the ropinirole patients.10

Ropinirole is available as tablets ranging in size from 0.25 mg to 5 mg. It is now also available in an extended-release (XL) formulation, with tablet sizes ranging from 2 mg to 12 mg. Ropinirole XL is taken once a day.

Bromocriptine is an old ergot-derived dopamine agonist that has also been studied for monotherapy and add-on treatment in PD. Due to the potential risks of pulmonary, retroperitoneal, and heart valve fibrosis, bromocriptine is not commonly used.

Apomorphine was approved by the US Food and Drug Administration in 2004 as an acute, intermittent, subcutaneous injection for the treatment of hypomobility off episodes (end-of-dose wearing off and unpredictable on-off episodes) associated with advanced PD. Apomorphine has been shown to be beneficial in patients with unpredictable off periods.11 Its onset of action is 10 to 15 minutes, and the effects of each dose last for 60 to 90 minutes. The best tolerated dose is 4 mg to 10 mg. Apomorphine appears to be most useful as as rescue medication in the refractory off periods with severe bradykinesia and unpredictable off periods.

 

 

TREATMENT OF DYSKINESIAS

Reduction of levodopa doses will reduce the frequency of dyskinesias, but at a cost of worsened parkinsonism and increased numbers of off periods. An alternative is to spread out the doses of levodopa (more frequent smaller doses), but this practice has not achieved good results. Replacing levodopa with dopamine agonists can also reduce the frequency of dyskinesias, but control of PD symptoms is less optimal than with levodopa. Amantadine and clozapine both have been shown to reduce dyskinesias.

Amantadine

Amantadine is an N-methyl-d-aspartate antagonist with antidyskinesia effects. Metman et al12 demonstrated that amantadine reduced dyskinesia severity by 60%, without exacerbation of motor function, in a randomized placebo-controlled crossover study. Dose-response studies with amantadine have not been conducted, but 100 mg two or three times daily is used in practice. In some studies, a short duration of benefit has been a concern. Side effects of amantadine include leg edema, hallucinations, confusion, and rash.

Clozapine

Clozapine is an atypical antipsychotic that has been shown in open-label trials and a randomized, double-blind, placebo-controlled trial to reduce the duration and severity of levodopa-induced dyskinesias without worsening of parkinsonian features and with no change in motor fluctuation.13 No benefit of clozapine was observed during activation dyskinesia, however. Cloza pine carries the inconvenience of weekly blood draws to monitor for the development of agranulocytosis, which occurs rarely.

GAIT FREEZING

Gait freezing, most commonly a manifestation of off states, causes substantial disability. It has been thought to occur as a result of a loss of noradrenaline due to locus ceruleus degeneration. Improvement in gait freezing has been shown with apomorphine and methylphenidate.

CONCEPT OF CONTINUOUS DOPAMINE STIMULATION

Short-acting dopaminergic drugs have the potential for nonphysiologic pulsatile stimulation of postsynaptic receptors, leading to motor complications. Continuous dopaminergic stimulation to prevent this pulsatile stimulation would theoretically avoid motor complications.14 Continuous dopaminergic stimulation can be achieved by using the extended-release formulation of ropinirole or pramipexole or by continuous delivery of levodopa or dopamine agonists. Several double-blind controlled trials have shown that treatment with long-acting dopamine agonists lowers the risk of motor complications compared with short-acting levodopa treatment.

In 2005, Stocchi concluded that in patients with advanced PD, a continuous infusion of levodopa was more effective in reducing motor complications than standard oral formulations.15 The reduction in motor complications was attributed to avoidance of low plasma levodopa trough levels; motor complications were not affected by relatively high plasma levodopa concentrations. The authors of this study speculated that if oral levodopa could be given “in a manner that mirrors the pharmacokinetic pattern of infusion,” it might be able to reduce motor complications.

This hypothesis led to an interest in treatment with levodopa plus entacapone. A regimen of levodopa-carbidopa-entacapone, four times daily at 3.5-hour intervals, was compared with levodopa-carbidopa in 747 patients with early PD over 134 weeks.16 Initiating levodopa therapy with levodopa-carbidopa in combination with entacapone did not delay the induction of dyskinesia compared with levodopa-carbidopa alone. In fact, levodopa-carbidopa-entacapone was associated with a shorter time to onset and an increased frequency of dyskinesia compared with levodopa-carbidopa.

Potential future treatment options

An intrajejunal pump system delivers a constant-rate infusion of levodopa. A double-blind study of this system is being conducted in the United States. Implantation of the system is an invasive procedure with the potential for infection, kinking dislocation, and occlusion and reposition of the catheter.

Miniature pumps for continuous subcutaneous delivery of apomorphine, currently available only in Europe, have been shown to reverse dyskinesias and motor fluctuations. Limitations of the minipumps are the development of red itchy nodules, ulcerations, and abscesses at infusion sites.

Extended-release dopamine agonists

Extended-release formulations of the dopamine agonists ropinirole and pramipexole are easy to administer, and they maintain therapeutic plasma levels for up to 24 hours. They are unlikely to replace stronger continuous dopamine stimulation with levodopa and apomorphine.

SUMMARY

Motor complications in PD result from progression of the disease and limitations of levodopa. Although the effects of levodopa on PD eventually wane, leaving patients vulnerable to motor complications, clinicians should not undertreat patients.

Effective options for the management of motor complications include prolonging the efficacy of levodopa through the use of selective MAO-B inhibitors and COMT inhibitors as adjuncts to levodopa or continuous dopaminergic stimulation achieved by the use of long-acting dopamine agonists or continuous intraduodenal levodopa.

Emerging therapies will be more efficient for continuous delivery of dopaminergic drugs. Pump delivery systems and extended-release formulations have shown promise.

Dopaminergic treatment is extremely beneficial in inducing symptom improvement in early Parkinson disease (PD). Patients typically experience a smooth and even response to the early stages of levodopa treatment. With disease progression, however, the effect of levodopa begins to weaken approximately 4 hours after each dose, leaving patients anticipating the need for their next dose and vulnerable to motor fluctuations and dyskinesias.

Motor fluctuations refer to the unanticipated loss of effect of a given dose of levodopa; instead of a smooth, predictable symptomatic benefit, the patient may lose benefit earlier than usual (termed “wearing off”) or may suddenly switch from “on” (symptoms controlled) to “off” (symptoms return). Dyskinesias, or involuntary movements, occur when dopamine levels are too high.

Motor complications are a major cause of disability in PD. They affect 60% to 90% of PD patients after 5 to 10 years of treatment. Moreover, in one study of 143 PD patients, motor complications diminished quality of life; the most strongly affected dimensions were mobility, activities of daily living, communication, and stigma.1

PATHOGENESIS OF MOTOR COMPLICATIONS

Under physiologic conditions, dopamine stimulation of the striatal dopamine receptors occurs in a sustained fashion. In early PD, the pool of remaining neurons of the substantia nigra is believed to be sufficiently active to smooth out changes in levodopa levels, providing a relatively constant amount of dopamine. Many PD patients therefore have several years of trouble-free treatment following diagnosis. In the advanced disease states, however, the number of presynaptic dopaminergic neurons progressively decreases. With fewer dopaminergic neurons, a constant dopaminergic concentration cannot be sustained. As PD advances, the progressive loss of dopaminergic neurons leads to impaired dopamine storage. Thus, the buffering capacity of dopaminergic neurons decreases and synaptic dopamine levels begin to reflect systemic or exogenous levodopa levels.

Figure. Pathogenesis of motor complications related to the Parkinson disease (PD) process and levodopa therapy. As PD advances (left), the progressive loss of dopaminergic neurons associated with nigrostriatal damage leads to impaired dopamine storage and clearance. This reduces the buffering capacity of dopaminergic neurons, causing early wearing off. During chronic levodopa treatment (right), pulsatile dopaminergic stimulation causes changes in postsynaptic receptors, referred to as “priming,” that increase the responsiveness of the receptors. The increased response results in severe levodopa-induced dyskinesias and on-off fluctuations. These postsynaptic changes are mediated through postsynaptic dopamine D1 receptors and N-methyl-d-aspartate glutamate receptors.
With disease progression, the “honeymoon” phase diminishes, and most PD patients begin to develop motor complications after 5 or more years. At this stage, medications frequently need to be adjusted, which can be a complex task for the physician and patient. The schema for the pathogenesis of motor complications related to the disease process and chronic levodopa treatment are depicted in the Figure.

According to current views, the total motor response to levodopa results from the combination of endogenous dopamine production along with the short-duration response (SDR) and the long-duration response (LDR) to exogenous levodopa.2 The SDR represents an improvement in parkinsonian symptoms and signs, lasting minutes to hours, that is closely related to the rise and fall of plasma levodopa concentrations. The SDR parallels the fluctuations in motor response and has received the most attention in the literature. The LDR is an improvement in parkinsonism that builds up over days and likewise decays over days. The LDR decays more rapidly in severely affected patients. Negative response, or “super off,” is a transient worsening of motor function to below the baseline level that may occur as the effects of the SDR dissipate.

The proportions of the SDR and LDR can vary according to disease progression. The LDR is more prominent in early stages, accounting for the stable response seen in the honeymoon period of treatment.

Peripheral factors

Additional peripheral factors such as changes in gastric motility and absorption contribute to motor complications. Levodopa is transported by a saturable active transporter system, the large neutral amino acid system, in the gut, and across the blood-brain barrier. Levodopa absorption is thus affected by food intake, especially protein. Levodopa and dietary amino acids compete with each other for absorption at the intestinal and blood-brain levels. Levodopa and other dopaminergic therapies further chronically reduce gastric emptying.

Pulsatile dopamine stimulation

The latency from the time of levodopa administration to the onset of motor improvement is typically 30 to 90 minutes. Latency is longer in late stages when the striatal buffer is weakened and the plasma concentration of levodopa fluctuates.

TYPES OF MOTOR FLUCTUATIONS

Fluctuating motor response in levodopa-treated patients refers to clinically apparent oscillations in motor function. Management of the fluctuating response may require frequent daily dosing of levodopa.

Motor fluctuations in PD take four forms: wearing off, off, delayed on/no on, and dyskinesias.

Wearing off

Wearing off refers to the premature loss of benefit from a given dose of levodopa, causing a predictable return of parkinsonian symptoms (bradykinesia, tremors, rigidity, and gait problems) in advance of the next scheduled dose. Observed in early and moderate PD, wearing off is the most common type of motor fluctuation. Its pathophysiology relates to disease progression and pharmacokinetics of levodopa. It can be sudden or gradual, predictable or unpredictable.

Off state

The off state is the unpredictable reappearance of parkinsonian symptoms at a time when central levels of antiparkinsonian drugs are expected to be within the target therapeutic range. Such symptoms include pain, stiffness, paresthesia, cognitive symptoms (depression, anxiety, difficulty with concentration, and mental slowing), inner restlessness, and inner tremulousness. The off state can be sudden or gradual, predictable or unpredictable.

Delayed on/no on

Delayed on is a prolongation of the time required for the central antiparkinsonian drug effect to appear. As the disease progresses, wearing off becomes more complicated and more unpredictable. The dosing responses vary, and patients sometimes report delayed on. The causes of delayed on or no on can be an insufficient dose, dosing with high-protein meals, or delayed gastric emptying. Metoclopramide or domperidone can help with gastric emptying. Metoclopramide can cross the blood-brain barrier and thus may cause adverse effects related to dopaminergic blockade; domperidone does not cross the blood-brain barrier.

Dyskinesias

Dyskinesias are hyperkinetic movements related to dopaminergic effects that are greater or less than the therapeutic threshold. They are common with long-term levodopa therapy and have three patterns:

Off dystonia occurs when levodopa concentrations are low and the SDR has dissipated. Dystonic states may be a manifestation of too little or too much levodopa; differentiating the two is important. Off dystonia occurs mostly in the early mornings, when plasma levodopa levels are low, and mostly involves the more affected side first.

Peak-dose dyskinesia, which occurs during the SDR, is the most common type of dykinesia and is related to peak plasma levodopa levels. It is characterized by stereotypic, choreic abnormal movements involving the head, neck, trunk, and limbs, and possibly hemidyskinesia in young-onset PD. Peak-dose dyskinesias are sometimes severe enough to be disabling.

Diphasic dyskinesias are stereotyped, dystonic, or choreic movements that occur at the beginning of the SDR and again as the SDR dissipates. They predominantly affect the legs and spare the trunk, neck, and arms.

 

 

TREATMENT OF OFF AND WEARING OFF

Increasing dopaminergic stimulation is the backbone of treatment of off periods or wearing off. The strategies to increase dopaminergic stimulation include addressing food and tolerance issues and adding a monoamine oxidase B (MAO-B) inhibitor or a catechol-O-methyltransferase (COMT) inhibitor such as entacapone or tolcapone to the regimen (Table). If the patient is already taking levodopa or a dopamine agonist, the dosage can be increased; or, levodopa can be added to a dopamine agonist regimen and vice versa.

Food and tolerance

The patient should not take levodopa with protein-containing meals, particularly if his or her PD is at an advanced stage. If excessive nausea, vomiting, or lightheadedness prevents the patient from taking an adequate dose, adding carbidopa (up to 75 mg) to the regimen will be helpful.

MAO-B inhibitors

By inhibiting one of the central dopamine catabolic pathways, MAO-B inhibitors (selegiline, rasagiline, and Zydis selegiline) prolong the half-life of dopamine in the brain and increase on time.

Improvement in off time with rasagiline is comparable to that seen with the COMT inhibitor entacapone. In an 18-week, double-blind trial of 687 patients randomized to receive once-daily rasagiline, entacapone, or placebo as an adjunct to levodopa, both rasagiline and entacapone reduced off time by 1.2 hours compared with placebo.3

In a 26-week placebo-controlled study, rasagiline decreased off time by 29% when added to levodopa in patients with PD and motor fluctuations, compared with a 15% reduction in the placebo group.4 This study confirmed the benefit of adding rasagiline to the regimens of patients who were already optimally treated with levodopa, dopamine agonists, amantadine, anticholinergics, and entacapone before enrolling in the study.

An orally disintegrating selegiline (Zydis selegiline) tablet is particularly useful for patients who have difficulty swallowing. The bioavailability of Zydis selegiline is 80% compared with 10% for selegiline, resulting in faster absorption. Pregastric absorption of Zydis selegiline avoids extensive first-pass metabolism in the liver and, therefore, the concentration of amphetamine-like metabolites is much lower.

In a 3-month, placebo-controlled study of patients with PD who were experiencing levodopa-related motor fluctuations, Zydis selegiline was associated with a 2.2-hour reduction in the total number of off hours compared with 0.6 hours in the placebo group, and dyskinesia-free on hours increased by 1.8 hours.5

The use of MAO-B inhibitors with tricyclic antidepressants or selective serotonin reuptake inhibitors has been reported to induce the serotonin syndrome by activation of 5HT1a and 5HT2 receptors. Serotonin syndrome is a potentially life-threatening accumulation of serotonin that can cause encephalopathy, severe rigidity of the legs, dysautonomia (diarrhea, mydriasis, and excessive lacrimation), myoclonus, hyperreflexia, and seizures.

COMT inhibitors

Catechol-O-methyltransferase inhibitors (entacapone and tolcapone) block peripheral degradation of levodopa. Tolcapone also blocks central degradation of levodopa and dopamine. These mechanisms increase central levodopa and dopamine levels and prolong levodopa half-life and bioavailability. Tolcapone has more powerful COMT inhibition than entacapone because tolcapone crosses the blood-brain barrier and inhibits the peripheral and central pathways of levodopa degradation. Use of COMT inhibitors can increase daily on time, but diarrhea is a common side effect and leads to withdrawal of these agents in about 3% of patients.

Tolcapone-treated patients show significant improvement in off time with improvement in motor fluctuations.6 Because tolcapone causes rare instances of fulminant hepatitis, liver function needs to be monitored every other week. For this reason, tolcapone should be reserved for patients in whom other treatments, including entacapone, have failed.

Controlled-release levodopa

Controlled-release levodopa was developed to provide more constant delivery of levodopa to the striatum. The benefit of controlled-release levodopa is only mild, however, as absorption of this formulation is variable. In advanced PD cases, the effects of controlled-release levodopa are more unpredictable than those with standard levodopa. Controlled-release levodopa is effective in patients with less severe wearing off, but it is not as effective in patients with a less predictable pattern of fluctuations.

Dopamine agonists

Dopamine agonists (pramipexole, ropinirole, apomorphine, and bromocriptine) have shown beneficial effects as adjunctive therapy to reduce wearing off. Side effects of dopamine agonists include ankle edema, hallucinations, somnolence, and impulse control disorders. These side effects should be discussed with patients before instituting therapy, and therapy should be discontinued if any of them occur.

In patients with advanced PD, pramipexole was shown to improve motor function during on and off periods, decrease the total off time, and decrease the severity of off time. Further, a larger reduction in the dosage of levodopa was possible in the pramipexole-treated patients than in the placebo-treated patients.7

In a comparison of pramipexole with levodopa on the end point of motor complications of PD in 300 patients, the incidences of wearing off and dyskinesia were significantly lower in the patients randomized to pramipexole with follow-up over 4 years.8 Only 25% of patients initially treated with pramipexole exhibited dyskinesia compared with 54% of patients initially treated with levodopa. Forty-seven percent of patients in the pramipexole group experienced wearing off compared with 63% initially treated with levodopa. Pramipexole is available as tablets ranging from 0.125 mg to 1.5 mg in size. It is given in three divided daily doses with gradual increments of 0.25 mg three times a day every week. Pramipexole is now also available in an extended-release formulation for once-a-day dosing in tablets ranging in size from 0.375 mg to 4.5 mg.

Ropinirole adds clinical benefit in PD patients with motor fluctuations and also permits a reduction in the dosage of levodopa.9

In one study, ropinirole monotherapy was compared with levodopa therapy in 268 patients with early PD. By the end of the 5-year study, 45% of the levodopa patients experienced dyskinesias versus 20% of the ropinirole patients.10

Ropinirole is available as tablets ranging in size from 0.25 mg to 5 mg. It is now also available in an extended-release (XL) formulation, with tablet sizes ranging from 2 mg to 12 mg. Ropinirole XL is taken once a day.

Bromocriptine is an old ergot-derived dopamine agonist that has also been studied for monotherapy and add-on treatment in PD. Due to the potential risks of pulmonary, retroperitoneal, and heart valve fibrosis, bromocriptine is not commonly used.

Apomorphine was approved by the US Food and Drug Administration in 2004 as an acute, intermittent, subcutaneous injection for the treatment of hypomobility off episodes (end-of-dose wearing off and unpredictable on-off episodes) associated with advanced PD. Apomorphine has been shown to be beneficial in patients with unpredictable off periods.11 Its onset of action is 10 to 15 minutes, and the effects of each dose last for 60 to 90 minutes. The best tolerated dose is 4 mg to 10 mg. Apomorphine appears to be most useful as as rescue medication in the refractory off periods with severe bradykinesia and unpredictable off periods.

 

 

TREATMENT OF DYSKINESIAS

Reduction of levodopa doses will reduce the frequency of dyskinesias, but at a cost of worsened parkinsonism and increased numbers of off periods. An alternative is to spread out the doses of levodopa (more frequent smaller doses), but this practice has not achieved good results. Replacing levodopa with dopamine agonists can also reduce the frequency of dyskinesias, but control of PD symptoms is less optimal than with levodopa. Amantadine and clozapine both have been shown to reduce dyskinesias.

Amantadine

Amantadine is an N-methyl-d-aspartate antagonist with antidyskinesia effects. Metman et al12 demonstrated that amantadine reduced dyskinesia severity by 60%, without exacerbation of motor function, in a randomized placebo-controlled crossover study. Dose-response studies with amantadine have not been conducted, but 100 mg two or three times daily is used in practice. In some studies, a short duration of benefit has been a concern. Side effects of amantadine include leg edema, hallucinations, confusion, and rash.

Clozapine

Clozapine is an atypical antipsychotic that has been shown in open-label trials and a randomized, double-blind, placebo-controlled trial to reduce the duration and severity of levodopa-induced dyskinesias without worsening of parkinsonian features and with no change in motor fluctuation.13 No benefit of clozapine was observed during activation dyskinesia, however. Cloza pine carries the inconvenience of weekly blood draws to monitor for the development of agranulocytosis, which occurs rarely.

GAIT FREEZING

Gait freezing, most commonly a manifestation of off states, causes substantial disability. It has been thought to occur as a result of a loss of noradrenaline due to locus ceruleus degeneration. Improvement in gait freezing has been shown with apomorphine and methylphenidate.

CONCEPT OF CONTINUOUS DOPAMINE STIMULATION

Short-acting dopaminergic drugs have the potential for nonphysiologic pulsatile stimulation of postsynaptic receptors, leading to motor complications. Continuous dopaminergic stimulation to prevent this pulsatile stimulation would theoretically avoid motor complications.14 Continuous dopaminergic stimulation can be achieved by using the extended-release formulation of ropinirole or pramipexole or by continuous delivery of levodopa or dopamine agonists. Several double-blind controlled trials have shown that treatment with long-acting dopamine agonists lowers the risk of motor complications compared with short-acting levodopa treatment.

In 2005, Stocchi concluded that in patients with advanced PD, a continuous infusion of levodopa was more effective in reducing motor complications than standard oral formulations.15 The reduction in motor complications was attributed to avoidance of low plasma levodopa trough levels; motor complications were not affected by relatively high plasma levodopa concentrations. The authors of this study speculated that if oral levodopa could be given “in a manner that mirrors the pharmacokinetic pattern of infusion,” it might be able to reduce motor complications.

This hypothesis led to an interest in treatment with levodopa plus entacapone. A regimen of levodopa-carbidopa-entacapone, four times daily at 3.5-hour intervals, was compared with levodopa-carbidopa in 747 patients with early PD over 134 weeks.16 Initiating levodopa therapy with levodopa-carbidopa in combination with entacapone did not delay the induction of dyskinesia compared with levodopa-carbidopa alone. In fact, levodopa-carbidopa-entacapone was associated with a shorter time to onset and an increased frequency of dyskinesia compared with levodopa-carbidopa.

Potential future treatment options

An intrajejunal pump system delivers a constant-rate infusion of levodopa. A double-blind study of this system is being conducted in the United States. Implantation of the system is an invasive procedure with the potential for infection, kinking dislocation, and occlusion and reposition of the catheter.

Miniature pumps for continuous subcutaneous delivery of apomorphine, currently available only in Europe, have been shown to reverse dyskinesias and motor fluctuations. Limitations of the minipumps are the development of red itchy nodules, ulcerations, and abscesses at infusion sites.

Extended-release dopamine agonists

Extended-release formulations of the dopamine agonists ropinirole and pramipexole are easy to administer, and they maintain therapeutic plasma levels for up to 24 hours. They are unlikely to replace stronger continuous dopamine stimulation with levodopa and apomorphine.

SUMMARY

Motor complications in PD result from progression of the disease and limitations of levodopa. Although the effects of levodopa on PD eventually wane, leaving patients vulnerable to motor complications, clinicians should not undertreat patients.

Effective options for the management of motor complications include prolonging the efficacy of levodopa through the use of selective MAO-B inhibitors and COMT inhibitors as adjuncts to levodopa or continuous dopaminergic stimulation achieved by the use of long-acting dopamine agonists or continuous intraduodenal levodopa.

Emerging therapies will be more efficient for continuous delivery of dopaminergic drugs. Pump delivery systems and extended-release formulations have shown promise.

References
  1. Chapuis S, Ouchchane L, Metz O, Gerbaud L, Durif F. Impact of the motor complications of Parkinson’s disease on the quality of life. Mov Disord 2005; 20:224230.
  2. Nutt JG, Holford NHG. The response to levodopa in Parkinson’s disease: imposing pharmacological law and order. Ann Neurol 1996; 39:561573.
  3. Rascol O, Brooks DJ, Oertel W, et al. Rasagiline as an adjunct to levodopa in patients with Parkinson’s disease and motor fluctuations (LARGO, Lasting effect in Adjunct therapy with Rasagiline Given Once daily, study): a randomised, double-blind, parallel-group trial. Lancet 2005; 365:947954.
  4. Parkinson Study Group. A randomized placebo-controlled trial of rasagiline in levodopa-treated patients with Parkinson disease and motor fluctuations: the PRESTO study. Arch Neurol 2005; 62:241248.
  5. Waters CH, Sethi KD, Hauser RA, et al. Zydis selegiline reduces off time in Parkinson’s disease patients with motor fluctuations: a 3-month, randomized, placebo-controlled study. Mov Disord 2004; 19:426432.
  6. Rajput AH, Martin W, Saint-Hilaire MH, Dorflinger E, Pedder S. Tolcapone improves motor function in parkinsonian patients with the “wearing-off” phenomenon: a double-blind, placebo-controlled, multicenter trial. Neurology 1997; 49:10661071.
  7. Lieberman A, Ranhosky A, Korts D. Clinical evaluation of pramipexole in advanced Parkinson’s disease: results of a double-blind, placebo-controlled, parallel-group study. Neurology 1997; 49:162168.
  8. Parkinson Study Group. Pramipexole vs levodopa as initial treatment for Parkinson disease: a 4-year randomized controlled trial. JAMA 2004; 61:10441053.
  9. Lieberman A, Olanow CW, Sethi K, et al. A multicenter trial of ropinirole as adjunct treatment for Parkinson’s disease. Neurology 1998; 51:10571062.
  10. Rascol O, Brooks DJ, Korczyn AD, et al. A five-year study of the incidence of dyskinesia in patients with early Parkinson’s disease who were treated with ropinirole or levodopa. N Engl J Med 2000; 342:14841491.
  11. Dewey RB, Hutton JT, LeWitt PA, Factor SA. A randomized, double-blind, placebo-controlled trial of subcutaneously injected apomorphine for parkinsonian off-state events. Arch Neurol 2001; 58:13851392.
  12. Metman LV, Del Dotto P, van den Munckhof P, Fang J, Mouradian MM, Chase TN. Amantadine as treatment for dyskinesias and motor fluctuations in Parkinson’s disease. Neurology 1998; 50:13231326.
  13. Durif F, Debilly B, Galitzky M, et al. Clozapine improves dyskinesias in Parkinson disease: a double-blind, placebo-controlled study. Neurology 2004; 62:381388.
  14. Olanow CW, Obeso JA, Stocchi F. Continuous dopamine-receptor treatment of Parkinson’s disease: scientific rationale and clinical implications. Lancet Neurol 2006; 5:677687.
  15. Stocchi F, Vacca L, Ruggieri S, Olanow CW. Intermittent vs continuous levodopa administration in patients with advanced Parkinson disease: a clinical and pharmacokinetic study. Arch Neurol 2005; 62:905910.
  16. Stocchi F, Rascol O, Kieburtz K, et al. Initiating levodopa/carbidopa therapy with and without entacapone in early Parkinson disease: The STRIDE-PD study. Ann Neurol 2010; 68:1827.
References
  1. Chapuis S, Ouchchane L, Metz O, Gerbaud L, Durif F. Impact of the motor complications of Parkinson’s disease on the quality of life. Mov Disord 2005; 20:224230.
  2. Nutt JG, Holford NHG. The response to levodopa in Parkinson’s disease: imposing pharmacological law and order. Ann Neurol 1996; 39:561573.
  3. Rascol O, Brooks DJ, Oertel W, et al. Rasagiline as an adjunct to levodopa in patients with Parkinson’s disease and motor fluctuations (LARGO, Lasting effect in Adjunct therapy with Rasagiline Given Once daily, study): a randomised, double-blind, parallel-group trial. Lancet 2005; 365:947954.
  4. Parkinson Study Group. A randomized placebo-controlled trial of rasagiline in levodopa-treated patients with Parkinson disease and motor fluctuations: the PRESTO study. Arch Neurol 2005; 62:241248.
  5. Waters CH, Sethi KD, Hauser RA, et al. Zydis selegiline reduces off time in Parkinson’s disease patients with motor fluctuations: a 3-month, randomized, placebo-controlled study. Mov Disord 2004; 19:426432.
  6. Rajput AH, Martin W, Saint-Hilaire MH, Dorflinger E, Pedder S. Tolcapone improves motor function in parkinsonian patients with the “wearing-off” phenomenon: a double-blind, placebo-controlled, multicenter trial. Neurology 1997; 49:10661071.
  7. Lieberman A, Ranhosky A, Korts D. Clinical evaluation of pramipexole in advanced Parkinson’s disease: results of a double-blind, placebo-controlled, parallel-group study. Neurology 1997; 49:162168.
  8. Parkinson Study Group. Pramipexole vs levodopa as initial treatment for Parkinson disease: a 4-year randomized controlled trial. JAMA 2004; 61:10441053.
  9. Lieberman A, Olanow CW, Sethi K, et al. A multicenter trial of ropinirole as adjunct treatment for Parkinson’s disease. Neurology 1998; 51:10571062.
  10. Rascol O, Brooks DJ, Korczyn AD, et al. A five-year study of the incidence of dyskinesia in patients with early Parkinson’s disease who were treated with ropinirole or levodopa. N Engl J Med 2000; 342:14841491.
  11. Dewey RB, Hutton JT, LeWitt PA, Factor SA. A randomized, double-blind, placebo-controlled trial of subcutaneously injected apomorphine for parkinsonian off-state events. Arch Neurol 2001; 58:13851392.
  12. Metman LV, Del Dotto P, van den Munckhof P, Fang J, Mouradian MM, Chase TN. Amantadine as treatment for dyskinesias and motor fluctuations in Parkinson’s disease. Neurology 1998; 50:13231326.
  13. Durif F, Debilly B, Galitzky M, et al. Clozapine improves dyskinesias in Parkinson disease: a double-blind, placebo-controlled study. Neurology 2004; 62:381388.
  14. Olanow CW, Obeso JA, Stocchi F. Continuous dopamine-receptor treatment of Parkinson’s disease: scientific rationale and clinical implications. Lancet Neurol 2006; 5:677687.
  15. Stocchi F, Vacca L, Ruggieri S, Olanow CW. Intermittent vs continuous levodopa administration in patients with advanced Parkinson disease: a clinical and pharmacokinetic study. Arch Neurol 2005; 62:905910.
  16. Stocchi F, Rascol O, Kieburtz K, et al. Initiating levodopa/carbidopa therapy with and without entacapone in early Parkinson disease: The STRIDE-PD study. Ann Neurol 2010; 68:1827.
Page Number
S8-S13
Page Number
S8-S13
Publications
Publications
Article Type
Display Headline
Off spells and dyskinesias: Pharmacologic management of motor complications
Display Headline
Off spells and dyskinesias: Pharmacologic management of motor complications
Citation Override
Cleveland Clinic Journal of Medicine 2012 July;79(suppl 2):S8-S13
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media

Nonmotor complications of Parkinson disease

Article Type
Changed
Mon, 10/01/2018 - 15:34
Display Headline
Nonmotor complications of Parkinson disease

Although the definition of Parkinson disease (PD) is based on the presence of motor features, these are just the “tip of the iceberg.” Nonmotor manifestations are nearly ubiquitous in PD, with behavior problems often being the most malignant. Almost all patients with PD have nonmotor and neuropsychiatric features, including sleep disturbances, compulsive and impulsive behaviors, autonomic dysfunction, and psychosis.

The neuropsychiatric and behavioral features of PD can be classified as intrinsic features, which occur as part of PD, and iatrogenic features, which are complications that arise from treatments used to manage the motor symptoms of PD.

DEMENTIA IN PD

An intrinsic nonmotor feature of PD is dementia, which occurs at a rate four to six times greater in patients with PD than in age-matched controls without PD.1 The prevalence of dementia in PD varies among studies and depends on the demographics of the population being studied. The cross-sectional prevalence of dementia is 40% in patients with PD.2 Seventy-eight percent of a population-based, representative cohort of patients with PD developed dementia during an 8-year study period.3

Dementia is a burden to the caregiver, the patient, and society. Cognitive and behavioral symptoms in patients with PD are the greatest contributors to caregiver distress.4 Dementia and associated behavioral symptoms (ie, hallucinations) hasten nursing home placement, contributing to the financial burden of caring for patients with PD.5 The risk of mortality is increased when dementia develops.6

Reprinted with permission from The New England Journal of Medicine (Emre M, et al. Rivastigmine for dementia associated with Parkinson’s disease. N Engl J Med 2004; 351:2509–2518). Copyright © 2004 Massachusetts Medical Society. All rights reserved.
Figure 1. In a double-blind, placebo-controlled trial that compared rivastigmine with placebo, patients with Parkinson disease dementia who were treated with rivastigmine experienced a 3-point improvement in the Alzheimer’s Disease Assessment Scale-Cognitive Subscale (ADAS-Cog) compared with placebo.
At least one medication has shown promise in managing PD dementia. In a pivotal trial of the cholinesterase inhibitor rivastigmine, involving more than 500 patients with PD dementia, the patients randomized to rivastigmine had a 3-point improvement on the primary outcome measure—the mean change from baseline in the Alzheimer’s Disease Assessment Scale-Cognitive Subscale—compared with those randomized to placebo (Figure 1).7 This trial led to US Food and Drug Administration approval of rivastigmine for the treatment of PD dementia.

PSYCHOTIC SYMPTOMS IN PD: AN EFFECT OF EXCESS DOPAMINE STIMULATION

Most of the complications observed in PD can be explained by the dopamine effect of medications and by dopamine deficiencies. An excess of dopamine stimulation caused by administration of prodopaminergic agents manifests as dyskinesias, hallucinations, or delusions. Withdrawal of levodopa will reverse these complications but leads to dopamine deficiency and thus a worsening of PD symptoms. Most patients with PD will tolerate mild dyskinesias or hallucinations if their PD symptoms are well controlled.

The hallucinations in PD tend to be visual as opposed to auditory (as in schizophrenia). They are usually benign and involve figures of people, furry animals, or complex scenes. About 10% to 40% of hallucinations in PD are secondary auditory hallucinations, which tend to be nondistinct, non-paranoid, and often incomprehensible (ie, voices in a crowd).

In the same way, the delusions experienced in patients with PD are distinct from those in schizophrenia. The delusions in PD are usually paranoid in nature and involve stereotyped themes (ie, spousal infidelity, feelings of abandonment) rather than the grandiose delusions that are common in schizophrenia.

The reported prevalence of psychotic symptoms in PD, including hallucinations and delusions, ranges from 20% to 50%.8,9 Auditory hallucinations are a feature in about 10%, and they usually occur with visual hallucinations. Less common are delusions and hallucinations with loss of insight, which are more likely with increasing severity of dementia.

Once a PD patient experiences hallucinations, they are likely to continue. In a 6-year longitudinal study, the prevalence of hallucinations increased from 33% at baseline to 55% at 72 months.10 Persistent psychosis was found in 69% of participants in the Psychosis and Clozapine in PD Study (PSYCLOPS) with 26 months of follow-up.11

High caregiver burden

Psychotic symptoms in PD are associated with high caregiver stress and increased rates of nursing home placement. Goetz et al12 showed that PD patients with psychosis had a much greater risk of nursing home placement than those without psychosis. The prognosis for PD patients in extended-care facilities is worse for those with psychotic symptoms.13

Management of psychotic symptoms

The first step in managing psychosis in PD is to rule out other causes of changes in mental status, such as infection, electrolyte imbalance, or introduction of new medications.

Adjusting anti-PD medications to a tolerable yet effective dose may help to reduce the incidence and severity of psychotic complications. If necessary, selective discontinuation of anti-PD medications may be tried in the following sequence: anticholinergics, amantadine, monoamine oxidase B inhibitors, dopamine agonists, catechol-O-methyltransferase inhibitors, and levodopa/carbidopa.

If motor symptoms prevent dosage minimization or discontinuation of some medications, then the addition of an atypical antipsychotic medication should be considered. Before the advent of atypical antipsychotics, the management of psychosis and hallucinations in PD was unsatisfactory, reflected by a mortality of 100% within 2 years among psychotic PD patients placed in nursing homes compared with 32% among age-matched community dwellers.13 The introduction of atypical antipsychotics has improved survival among PD patients with psychosis. In one study, mortality over 5 years was 44% among PD patients taking long-term clozapine for the treatment of psychosis.14 Recurrence of psychosis is rapid (within 8 weeks) even when PD patients are slowly weaned from atypical antipsychotics.15

Receptor affinities differ among antipsychotics. Because dopamine has been implicated as the principal neurotransmitter in the development of PD psychosis, atypical antipsychotics, with milder dopamine-blocking action, have played a central role in the treatment of PD psychosis. The dopamine D2 receptor is the main target for conventional antipsychotic drugs to exert their clinical effects. Atypical antipsychotics have different affinities for the D2 receptors.16 Occupancy of D2 receptors with atypical antipsychotics is 40% to 70% (risperidone and olanzapine have higher affinity for the D2 receptor than clozapine and quetiapine), and affinity for 5-HT2A receptors can be as high as 70%. This affinity for 5-HT2A receptors relative to D2 receptors may be important for therapeutic efficacy of the atypical antipsychotics. Antagonism of muscarinic, histaminergic, noradrenergic, and other serotonergic receptors also differs among the atypical antipsychotics.

Clozapine remains the gold standard atypical antipsychotic agent, based on results from three relatively small (N = 6 to 60) double-blind, placebo-controlled studies in PD patients with dopaminergic drug-induced psychosis.17–19 Quetiapine improved psychotic symptoms associated with PD in several open-label studies, but has not demonstrated the same success in double-blind clinical trials.20,21

Loss of cholinergic neurons and implications for treatment. In autopsy studies, the loss of cholinergic neurons is more profound in PD than in Alzheimer disease, which suggests that procholinergic drugs may improve symptoms of PD dementia, a major risk factor for hallucinations. In open-label studies, acetylcholinesterase inhibitors have reduced the frequency of hallucinations in patients who have dementia with Lewy bodies (DLB) and in patients with PD dementia. Double-blind trials of patients with DLB and PD dementia concentrated on the effect of cholinesterase inhibitors on dementia and not hallucinations. One concern with the use of a procholinergic drug in patients with PD has been worsening of parkinsonism, but studies of acetylcholinesterase inhibitors have shown no worsening of parkinsonism and only transient worsening of tremor.

Ondansetron, a 5-HT3 receptor antagonist used as an antinausea medication, produced moderate improvements in hallucinations and delusions in an open-label trial for the treatment of psychosis in advanced PD.22 For PD patients with psychosis and comorbid depression, antidepressant therapy and electroconvulsive therapy may be effective options.23,24

 

 

MOOD DISTURBANCES IN PD

Depression and apathy occur more frequently in patients with PD than in those who do not have PD.

Depression

Challenges in the management of depression in PD include recognition of depression and distinguishing depressive disorders from mood fluctuations. Whereas a depressive disorder lasts from weeks to years and can occur at any stage of illness, mood fluctuations can change many times daily and appear as nonmotor manifestations during the “off” medication state. Mood fluctuations occur mostly in patients who have developed motor fluctuations. The implication for treatment is that the treatment strategy for a depressive disorder is antidepressant therapy, whereas the strategy for mood fluctuations in PD is to increase the levodopa dose.

Recognition of depression in PD is confounded by the depression criteria of the Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition; many of these criteria can be intrinsic features of PD itself—for example, anhedonia, weight/appetite loss or gain, insomnia or hypersomnia, psychomotor retardation, and fatigue. Questions such as “are you feeling sad” or “are you feeling blue” may be superior to questions about associative symptoms when evaluating PD patients for depression.

Reprinted with permission from Parkinsonism and Related Disorders (Shulman LM, et al. Non-recognition of depression and other non-motor symptoms in Parkinson’s disease. Parkinsonism Relat Disord 2002; 8:193–197). © 2002 Elsevier Science Ltd.
Figure 2. A comparison of routine office assessment with the use of standardized rating scales that identify nonmotor symptoms demonstrated superiority of rating scales compared with neurologists’ impressions in identification of depression and other nonmotor complications of Parkinson disease. Investigators used the Beck Depression Inventory, the Beck Anxiety Inventory, the Fatigue Severity Scale, and the Pittsburg Sleep Quality Index.
The value of rating scales also should not be overlooked. Shulman et al25 found that the use of standardized rating scales is superior to routine office assessment by neurologists in recognizing depression in PD patients; in more than 50% of routine office assessments, neurologists missed a diagnosis of depression (Figure 2).

Most of the medications used for the treatment of depression also work well for depression in patients with PD. Double-blind controlled studies have demonstrated superiority of nortriptyline, citalopram, desipramine, and pramipexole over placebo in improving mood.26–29

Apathy

The overlap between apathy and depressive symptoms can also complicate recognition of apathy, which can be described as a lack of motivation or failure to initiate goal-directed behavior. Apathy involves three domains30:

  • Cognitive: expressed as a loss of interest in new experience or a lack of concern about a personal problem
  • Diminished affect: flattened affect or a lack of reaction to positive or negative events
  • Final: diminished goal-directed cognition, as indicated by a lack of effort or requiring others to structure activities.

Unlike depression, which is similarly representative of PD and other episodic conditions such as dystonia, apathy is more common in PD than in dystonia. In fact, the occurrence of apathy alone distinguishes PD from dystonia. Apathy in PD has no known treatment. If it is associated with depression, apathy may respond to antidepressants.

Repetitive transcranial magnetic stimulation (rTMS) manipulates activity in specific brain neural circuits through the skull to induce changes in behavior. Some studies suggest that modulation of behavior may last beyond the actual stimulation. A randomized, sham-controlled trial of rTMS over the middorsolateral frontal cortex has been conducted with the primary aim of improving apathy in PD. Unfortunately, while patients who were randomized to rTMS experienced some improvement in apathy during the study, the improvement was not significantly different from that observed in patients who received sham treatment.31

IMPULSE CONTROL AND COMPULSIVE DISORDERS IN PD

Impulse control disorders are characterized by the inability to resist an urge to act; the resulting irrational desire to pursue self-gratification may inflict suffering on friends and relatives that compromises relationships and impairs social- and work-related functioning.

Examples of impulse control disorders in PD are pathologic gambling, hypersexuality, compulsive shopping, excessive spending, and binge eating. Patients taking dopamine agonists are two to three times more likely to develop impulse control disorders than those receiving other treatments for PD. Dopamine agonists with relative selectivity for D3 receptors have been implicated in impulse control disorders in PD because D3 receptors are abundant in a region of the brain (ventral striatum) associated with behavioral and substance addictions. Higher levodopa dosages were also associated with impulse control disorders.

Factors associated with impulse control disorders in PD are young age, being single, a family history of impulse control disorders, and levodopa treatment.32 Modifications to dopamine agonist or levodopa therapy are important in the treatment of dopamine agonist–induced impulse disorders.

Compulsive disorders have been described as a class distinct from impulse control disorders and involve repetitive stereotypes and well-ordered acts to decrease inner anxiety and avoid harm. Punding is the engagement of stereotyped behaviors that are repeated compulsively—for example, repetitive manipulation of technical equipment; continual handling, sorting, and examining of objects; grooming; and hoarding. The punder has poor insight into the disruptive and senseless nature of his or her acts. Punding has consistently been related to dopaminergic therapy. Its prevalence in PD patients on dopaminergic therapy ranges from 1.4%33 to 14%.34 An improvement in behavior is observed with a reduction in dosage or discontinuation of levodopa.

Pathologic gambling, or the inability to control gambling, can result in lying to obtain money for gambling, thereby complicating relationships. It can affect up to 8% of patients with PD.35

SUMMARY

Dementia, psychotic symptoms, mood disturbances, and impulse control disorders are important nonmotor manifestations of PD that present management challenges. Some of these manifestations are intrinsic to PD, and some are complications of therapies used to treat the motor manifestations of PD.

Dementia and psychotic symptoms extract a considerable toll on the patient, caregivers, and society. Psychotic symptoms generally manifest as hallucinations (mostly visual) and other sensory disturbances. Initial management involves adjustment of anti-PD medications. The use of atypical antipsychotic drugs has been shown to improve survival among patients with PD. Clozapine is the preferred agent.

Mood disturbances such as depression and apathy may be difficult to diagnose. Depression may be treated similarly to depression unassociated with PD.

Dopamine agonists and levodopa have been associated with impulse control disorders in PD. Compulsive disorders, which are distinct from impulse control disorders, may improve with reduction or discontinuation of levodopa therapy.

References
  1. Aarsland D, Andersen K, Larsen JP, Lolk A, Nielsen H, Kragh-Sørensen P. Risk of dementia in Parkinson’s disease: a community-based, prospective study. Neurology 2001; 56:730736.
  2. Cummings JL. Intellectual impairment in Parkinson’s disease: clinical, pathologic, and biochemical correlates. J Geriatr Psychiatry Neurol 1988; 1:2436.
  3. Aarsland D, Andersen K, Larsen JP, Lolk A, Kragh-Sørensen P. Prevalence and characteristics of dementia in Parkinson disease: an 8-year prospective study. Arch Neurol 2003; 60:387392.
  4. Aarsland D, Larsen JP, Karlsen K, Lim NG, Tandberg E. Mental symptoms in Parkinson’s disease are important contributors to caregiver distress. Int J Geriatr Psychiatry 1999; 14:866874.
  5. Aarsland D, Larsen JP, Tandberg E, Laake K. Predictors of nursing home placement in Parkinson’s disease: a population-based, prospective study. J Am Geriatr Soc 2000; 48:938942.
  6. Hughes TA, Ross HF, Mindham RH, Spokes EG. Mortality in Parkinson’s disease and its association with dementia and depression. Acta Neurol Scand 2004; 110:118123.
  7. Emre M, Aarsland D, Albanese A, et al. Rivastigmine for dementia associated with Parkinson’s disease. N Engl J Med 2004; 351:25092518.
  8. Fénelon G, Mahieux F, Huon R, Ziégler M. Hallucinations in Parkinson’s disease: prevalence, phenomenology and risk factors. Brain 2000; 123:733745.
  9. Zahodne LB, Fernandez HH. Pathophysiology and treatment of psychosis in Parkinson’s disease: a review. Drugs Aging 2008; 25:665682.
  10. Goetz CG, Wuu J, Curgian LM, Leurgans S. Hallucinations and sleep disorders in PD: six-year prospective longitudinal study. Neurology 2005; 64:8186.
  11. Factor SA, Feustel PJ, Friedman JH, et al. Longitudinal outcome of Parkinson’s disease patients with psychosis. Neurology 2003; 60:17561761.
  12. Goetz CG, Stebbins GT. Risk factors for nursing home placement in advanced Parkinson’s disease. Neurology 1993; 43:22272229.
  13. Goetz CG, Stebbins GT. Mortality and hallucinations in nursing home patients with advanced Parkinson’s disease. Neurology 1995; 45:669671.
  14. Fernandez HH, Donnelly EM, Friedman JH. Long-term outcome of clozapine use for psychosis in parkinsonian patients. Mov Disord 2004; 19:831833.
  15. Fernandez HH, Trieschmann ME, Okun MS. Rebound psychosis: effect of discontinuation of antipsychotics in Parkinson’s disease. Mov Disord 2005; 20:104115.
  16. Goldstein JM. Atypical antipsychotic drugs: beyond acute psychosis, new directions. Emerging Drugs 1999; 4:127151.
  17. Pollak P, Tison F, Rascol O, et al; on behalf of the French Clozapine Parkinson Study Group. Clozapine in drug induced psychosis in Parkinson’s disease: a randomised, placebo controlled study with open follow up. J Neurol Neurosurg Psychiatry 2004; 75:689695.
  18. The Parkinson Study Group. Low-dose clozapine for the treatment of drug-induced psychosis in Parkinson’s disease. N Engl J Med 1999; 340:757763.
  19. Wolters ECh, Hurwitz TA, Mak E, et al. Clozapine in the treatment of parkinsonian patients with dopaminomimetic psychosis. Neurology 1990; 40:832834.
  20. Ondo WG, Tintner R, Voung KD, Lai D, Ringholz G. Double-blind, placebo-controlled, unforced titration parallel trial of quetiapine for dopaminergic-induced hallucinations in Parkinson’s disease. Mov Disord 2005; 20:958963.
  21. Fernandez HH, Okun MS, Rodriguez RL, Malaty IA, Romrell J. Quetiapine improves visual hallucinations in Parkinson disease but not through normalization of sleep architecture: results from a double-blind clinical-polysomnography study. Int J Neurosci 2009; 119:21962205.
  22. Zoldan J, Friedberg G, Livneh M, Melamed E. Psychosis in advanced Parkinson’s disease: treatment with ondansetron, a 5-HT3 receptor antagonist. Neurology 1995; 45:13051308.
  23. Voon V, Lang AE. Antidepressants in the treatment of psychosis with comorbid depression in Parkinson disease. Clin Neuropharmacol 2004; 27:9092.
  24. Ozer F, Meral H, Aydin B, Hanoglu L, Aydemir T, Oral T. Electroconvulsive therapy in drug-induced psychiatric states and neuroleptic malignant syndrome. J ECT 2005; 21:125127.
  25. Shulman LM, Taback RL, Rabinstein AA, Weiner WJ. Non-recognition of depression and other non-motor symptoms in Parkinson’s disease. Parkinsonism Relat Disord 2002; 8:193197.
  26. Devos D, Dujardin K, Poirot I, et al. Comparison of desipramine and citalopram treatments for depression in Parkinson’s disease: a double-blind, randomized, placebo-controlled study. Mov Disord 2008; 23:850857.
  27. Menza M, Dobkin RD, Marin H, et al. A controlled trial of antidepressants in patients with Parkinson disease and depression. Neurology 2009; 72:886892.
  28. Barone P, Poewe W, Albrecht S, et al. Pramipexole for the treatment of depressive symptoms in patients with Parkinson’s disease: a randomised, double-blind, placebo-controlled trial. Lancet Neurol 2010; 9:573580.
  29. Fernandez HH, Merello M. Pramipexole for depression and motor symptoms in Parkinson disease: can we kill two birds with one stone? Lancet Neurol 2010; 9:556557.
  30. Marin RS. Apathy: a neuropsychiatric syndrome. J Neuropsychiatry Clin Neurosci 1991; 3:243254.
  31. Fernandez HH, Bowers D, Triggs WJ, et al. Repetitive transcranial magnetic stimulation for the treatment of apathy in Parkinson’s disease: results from a double-blind, sham-controlled, randomized, controlled trial. Neurology 2010; 74( suppl 2):352.
  32. Weintraub D, Koester J, Potenza MN, et al. Impulse control disorders in Parkinson disease: a cross-sectional study of 3090 patients. Arch Neurol 2010; 67:589595.
  33. Miyasaki JM, Al Hassan K, Lang AE, Voon V. Punding prevalence in Parkinson’s disease. Mov Disord 2007; 22:11791181.
  34. Evans AH, Katzenschlager R, Paviour D, et al. Punding in Parkinson’s disease: its relation to the dopamine dysregulation syndrome. Mov Disord 2004; 19:397405.
  35. Grosset KA, Macphee G, Pal G, et al. Problematic gambling on dopamine agonists: not such a rarity. Mov Disord 2006; 21:22062208.
Article PDF
Author and Disclosure Information

Hubert H. Fernandez, MD, FAAN, FANA
Head, Movement Disorders Program, Center for Neurological Restoration, Cleveland Clinic, Cleveland, OH

Correspondence: Hubert H. Fernandez, MD, Center for Neurological Restoration, Cleveland Clinic, 9500 Euclid Avenue, S31, Cleveland, OH 44195; fernanh@ccf.org

Dr. Fernandez reported independent contractor relationships with Abbott Laboratories, Acadia Pharmaceuticals Inc., Biotie Therapies, EMD Serono, Inc., Ipsen, Merck & Co., Inc., Merz Pharma, Novartis Corporation, Synosia Therapeutics, Schering Plough Corporation, and Teva Pharmaceuticals, Inc.; board memberships with Abbott Laboratories; and consulting and advisory committee membership with Abbott Laboratories.

This article is based on Dr. Fernandez’s presentation at “The Annual Therapy Symposium on Movement Disorders for the Modern Clinician” held in Fort Lauderdale, Florida, on January 29, 2011. The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Fernandez.

Publications
Page Number
S14-S18
Author and Disclosure Information

Hubert H. Fernandez, MD, FAAN, FANA
Head, Movement Disorders Program, Center for Neurological Restoration, Cleveland Clinic, Cleveland, OH

Correspondence: Hubert H. Fernandez, MD, Center for Neurological Restoration, Cleveland Clinic, 9500 Euclid Avenue, S31, Cleveland, OH 44195; fernanh@ccf.org

Dr. Fernandez reported independent contractor relationships with Abbott Laboratories, Acadia Pharmaceuticals Inc., Biotie Therapies, EMD Serono, Inc., Ipsen, Merck & Co., Inc., Merz Pharma, Novartis Corporation, Synosia Therapeutics, Schering Plough Corporation, and Teva Pharmaceuticals, Inc.; board memberships with Abbott Laboratories; and consulting and advisory committee membership with Abbott Laboratories.

This article is based on Dr. Fernandez’s presentation at “The Annual Therapy Symposium on Movement Disorders for the Modern Clinician” held in Fort Lauderdale, Florida, on January 29, 2011. The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Fernandez.

Author and Disclosure Information

Hubert H. Fernandez, MD, FAAN, FANA
Head, Movement Disorders Program, Center for Neurological Restoration, Cleveland Clinic, Cleveland, OH

Correspondence: Hubert H. Fernandez, MD, Center for Neurological Restoration, Cleveland Clinic, 9500 Euclid Avenue, S31, Cleveland, OH 44195; fernanh@ccf.org

Dr. Fernandez reported independent contractor relationships with Abbott Laboratories, Acadia Pharmaceuticals Inc., Biotie Therapies, EMD Serono, Inc., Ipsen, Merck & Co., Inc., Merz Pharma, Novartis Corporation, Synosia Therapeutics, Schering Plough Corporation, and Teva Pharmaceuticals, Inc.; board memberships with Abbott Laboratories; and consulting and advisory committee membership with Abbott Laboratories.

This article is based on Dr. Fernandez’s presentation at “The Annual Therapy Symposium on Movement Disorders for the Modern Clinician” held in Fort Lauderdale, Florida, on January 29, 2011. The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Fernandez.

Article PDF
Article PDF

Although the definition of Parkinson disease (PD) is based on the presence of motor features, these are just the “tip of the iceberg.” Nonmotor manifestations are nearly ubiquitous in PD, with behavior problems often being the most malignant. Almost all patients with PD have nonmotor and neuropsychiatric features, including sleep disturbances, compulsive and impulsive behaviors, autonomic dysfunction, and psychosis.

The neuropsychiatric and behavioral features of PD can be classified as intrinsic features, which occur as part of PD, and iatrogenic features, which are complications that arise from treatments used to manage the motor symptoms of PD.

DEMENTIA IN PD

An intrinsic nonmotor feature of PD is dementia, which occurs at a rate four to six times greater in patients with PD than in age-matched controls without PD.1 The prevalence of dementia in PD varies among studies and depends on the demographics of the population being studied. The cross-sectional prevalence of dementia is 40% in patients with PD.2 Seventy-eight percent of a population-based, representative cohort of patients with PD developed dementia during an 8-year study period.3

Dementia is a burden to the caregiver, the patient, and society. Cognitive and behavioral symptoms in patients with PD are the greatest contributors to caregiver distress.4 Dementia and associated behavioral symptoms (ie, hallucinations) hasten nursing home placement, contributing to the financial burden of caring for patients with PD.5 The risk of mortality is increased when dementia develops.6

Reprinted with permission from The New England Journal of Medicine (Emre M, et al. Rivastigmine for dementia associated with Parkinson’s disease. N Engl J Med 2004; 351:2509–2518). Copyright © 2004 Massachusetts Medical Society. All rights reserved.
Figure 1. In a double-blind, placebo-controlled trial that compared rivastigmine with placebo, patients with Parkinson disease dementia who were treated with rivastigmine experienced a 3-point improvement in the Alzheimer’s Disease Assessment Scale-Cognitive Subscale (ADAS-Cog) compared with placebo.
At least one medication has shown promise in managing PD dementia. In a pivotal trial of the cholinesterase inhibitor rivastigmine, involving more than 500 patients with PD dementia, the patients randomized to rivastigmine had a 3-point improvement on the primary outcome measure—the mean change from baseline in the Alzheimer’s Disease Assessment Scale-Cognitive Subscale—compared with those randomized to placebo (Figure 1).7 This trial led to US Food and Drug Administration approval of rivastigmine for the treatment of PD dementia.

PSYCHOTIC SYMPTOMS IN PD: AN EFFECT OF EXCESS DOPAMINE STIMULATION

Most of the complications observed in PD can be explained by the dopamine effect of medications and by dopamine deficiencies. An excess of dopamine stimulation caused by administration of prodopaminergic agents manifests as dyskinesias, hallucinations, or delusions. Withdrawal of levodopa will reverse these complications but leads to dopamine deficiency and thus a worsening of PD symptoms. Most patients with PD will tolerate mild dyskinesias or hallucinations if their PD symptoms are well controlled.

The hallucinations in PD tend to be visual as opposed to auditory (as in schizophrenia). They are usually benign and involve figures of people, furry animals, or complex scenes. About 10% to 40% of hallucinations in PD are secondary auditory hallucinations, which tend to be nondistinct, non-paranoid, and often incomprehensible (ie, voices in a crowd).

In the same way, the delusions experienced in patients with PD are distinct from those in schizophrenia. The delusions in PD are usually paranoid in nature and involve stereotyped themes (ie, spousal infidelity, feelings of abandonment) rather than the grandiose delusions that are common in schizophrenia.

The reported prevalence of psychotic symptoms in PD, including hallucinations and delusions, ranges from 20% to 50%.8,9 Auditory hallucinations are a feature in about 10%, and they usually occur with visual hallucinations. Less common are delusions and hallucinations with loss of insight, which are more likely with increasing severity of dementia.

Once a PD patient experiences hallucinations, they are likely to continue. In a 6-year longitudinal study, the prevalence of hallucinations increased from 33% at baseline to 55% at 72 months.10 Persistent psychosis was found in 69% of participants in the Psychosis and Clozapine in PD Study (PSYCLOPS) with 26 months of follow-up.11

High caregiver burden

Psychotic symptoms in PD are associated with high caregiver stress and increased rates of nursing home placement. Goetz et al12 showed that PD patients with psychosis had a much greater risk of nursing home placement than those without psychosis. The prognosis for PD patients in extended-care facilities is worse for those with psychotic symptoms.13

Management of psychotic symptoms

The first step in managing psychosis in PD is to rule out other causes of changes in mental status, such as infection, electrolyte imbalance, or introduction of new medications.

Adjusting anti-PD medications to a tolerable yet effective dose may help to reduce the incidence and severity of psychotic complications. If necessary, selective discontinuation of anti-PD medications may be tried in the following sequence: anticholinergics, amantadine, monoamine oxidase B inhibitors, dopamine agonists, catechol-O-methyltransferase inhibitors, and levodopa/carbidopa.

If motor symptoms prevent dosage minimization or discontinuation of some medications, then the addition of an atypical antipsychotic medication should be considered. Before the advent of atypical antipsychotics, the management of psychosis and hallucinations in PD was unsatisfactory, reflected by a mortality of 100% within 2 years among psychotic PD patients placed in nursing homes compared with 32% among age-matched community dwellers.13 The introduction of atypical antipsychotics has improved survival among PD patients with psychosis. In one study, mortality over 5 years was 44% among PD patients taking long-term clozapine for the treatment of psychosis.14 Recurrence of psychosis is rapid (within 8 weeks) even when PD patients are slowly weaned from atypical antipsychotics.15

Receptor affinities differ among antipsychotics. Because dopamine has been implicated as the principal neurotransmitter in the development of PD psychosis, atypical antipsychotics, with milder dopamine-blocking action, have played a central role in the treatment of PD psychosis. The dopamine D2 receptor is the main target for conventional antipsychotic drugs to exert their clinical effects. Atypical antipsychotics have different affinities for the D2 receptors.16 Occupancy of D2 receptors with atypical antipsychotics is 40% to 70% (risperidone and olanzapine have higher affinity for the D2 receptor than clozapine and quetiapine), and affinity for 5-HT2A receptors can be as high as 70%. This affinity for 5-HT2A receptors relative to D2 receptors may be important for therapeutic efficacy of the atypical antipsychotics. Antagonism of muscarinic, histaminergic, noradrenergic, and other serotonergic receptors also differs among the atypical antipsychotics.

Clozapine remains the gold standard atypical antipsychotic agent, based on results from three relatively small (N = 6 to 60) double-blind, placebo-controlled studies in PD patients with dopaminergic drug-induced psychosis.17–19 Quetiapine improved psychotic symptoms associated with PD in several open-label studies, but has not demonstrated the same success in double-blind clinical trials.20,21

Loss of cholinergic neurons and implications for treatment. In autopsy studies, the loss of cholinergic neurons is more profound in PD than in Alzheimer disease, which suggests that procholinergic drugs may improve symptoms of PD dementia, a major risk factor for hallucinations. In open-label studies, acetylcholinesterase inhibitors have reduced the frequency of hallucinations in patients who have dementia with Lewy bodies (DLB) and in patients with PD dementia. Double-blind trials of patients with DLB and PD dementia concentrated on the effect of cholinesterase inhibitors on dementia and not hallucinations. One concern with the use of a procholinergic drug in patients with PD has been worsening of parkinsonism, but studies of acetylcholinesterase inhibitors have shown no worsening of parkinsonism and only transient worsening of tremor.

Ondansetron, a 5-HT3 receptor antagonist used as an antinausea medication, produced moderate improvements in hallucinations and delusions in an open-label trial for the treatment of psychosis in advanced PD.22 For PD patients with psychosis and comorbid depression, antidepressant therapy and electroconvulsive therapy may be effective options.23,24

 

 

MOOD DISTURBANCES IN PD

Depression and apathy occur more frequently in patients with PD than in those who do not have PD.

Depression

Challenges in the management of depression in PD include recognition of depression and distinguishing depressive disorders from mood fluctuations. Whereas a depressive disorder lasts from weeks to years and can occur at any stage of illness, mood fluctuations can change many times daily and appear as nonmotor manifestations during the “off” medication state. Mood fluctuations occur mostly in patients who have developed motor fluctuations. The implication for treatment is that the treatment strategy for a depressive disorder is antidepressant therapy, whereas the strategy for mood fluctuations in PD is to increase the levodopa dose.

Recognition of depression in PD is confounded by the depression criteria of the Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition; many of these criteria can be intrinsic features of PD itself—for example, anhedonia, weight/appetite loss or gain, insomnia or hypersomnia, psychomotor retardation, and fatigue. Questions such as “are you feeling sad” or “are you feeling blue” may be superior to questions about associative symptoms when evaluating PD patients for depression.

Reprinted with permission from Parkinsonism and Related Disorders (Shulman LM, et al. Non-recognition of depression and other non-motor symptoms in Parkinson’s disease. Parkinsonism Relat Disord 2002; 8:193–197). © 2002 Elsevier Science Ltd.
Figure 2. A comparison of routine office assessment with the use of standardized rating scales that identify nonmotor symptoms demonstrated superiority of rating scales compared with neurologists’ impressions in identification of depression and other nonmotor complications of Parkinson disease. Investigators used the Beck Depression Inventory, the Beck Anxiety Inventory, the Fatigue Severity Scale, and the Pittsburg Sleep Quality Index.
The value of rating scales also should not be overlooked. Shulman et al25 found that the use of standardized rating scales is superior to routine office assessment by neurologists in recognizing depression in PD patients; in more than 50% of routine office assessments, neurologists missed a diagnosis of depression (Figure 2).

Most of the medications used for the treatment of depression also work well for depression in patients with PD. Double-blind controlled studies have demonstrated superiority of nortriptyline, citalopram, desipramine, and pramipexole over placebo in improving mood.26–29

Apathy

The overlap between apathy and depressive symptoms can also complicate recognition of apathy, which can be described as a lack of motivation or failure to initiate goal-directed behavior. Apathy involves three domains30:

  • Cognitive: expressed as a loss of interest in new experience or a lack of concern about a personal problem
  • Diminished affect: flattened affect or a lack of reaction to positive or negative events
  • Final: diminished goal-directed cognition, as indicated by a lack of effort or requiring others to structure activities.

Unlike depression, which is similarly representative of PD and other episodic conditions such as dystonia, apathy is more common in PD than in dystonia. In fact, the occurrence of apathy alone distinguishes PD from dystonia. Apathy in PD has no known treatment. If it is associated with depression, apathy may respond to antidepressants.

Repetitive transcranial magnetic stimulation (rTMS) manipulates activity in specific brain neural circuits through the skull to induce changes in behavior. Some studies suggest that modulation of behavior may last beyond the actual stimulation. A randomized, sham-controlled trial of rTMS over the middorsolateral frontal cortex has been conducted with the primary aim of improving apathy in PD. Unfortunately, while patients who were randomized to rTMS experienced some improvement in apathy during the study, the improvement was not significantly different from that observed in patients who received sham treatment.31

IMPULSE CONTROL AND COMPULSIVE DISORDERS IN PD

Impulse control disorders are characterized by the inability to resist an urge to act; the resulting irrational desire to pursue self-gratification may inflict suffering on friends and relatives that compromises relationships and impairs social- and work-related functioning.

Examples of impulse control disorders in PD are pathologic gambling, hypersexuality, compulsive shopping, excessive spending, and binge eating. Patients taking dopamine agonists are two to three times more likely to develop impulse control disorders than those receiving other treatments for PD. Dopamine agonists with relative selectivity for D3 receptors have been implicated in impulse control disorders in PD because D3 receptors are abundant in a region of the brain (ventral striatum) associated with behavioral and substance addictions. Higher levodopa dosages were also associated with impulse control disorders.

Factors associated with impulse control disorders in PD are young age, being single, a family history of impulse control disorders, and levodopa treatment.32 Modifications to dopamine agonist or levodopa therapy are important in the treatment of dopamine agonist–induced impulse disorders.

Compulsive disorders have been described as a class distinct from impulse control disorders and involve repetitive stereotypes and well-ordered acts to decrease inner anxiety and avoid harm. Punding is the engagement of stereotyped behaviors that are repeated compulsively—for example, repetitive manipulation of technical equipment; continual handling, sorting, and examining of objects; grooming; and hoarding. The punder has poor insight into the disruptive and senseless nature of his or her acts. Punding has consistently been related to dopaminergic therapy. Its prevalence in PD patients on dopaminergic therapy ranges from 1.4%33 to 14%.34 An improvement in behavior is observed with a reduction in dosage or discontinuation of levodopa.

Pathologic gambling, or the inability to control gambling, can result in lying to obtain money for gambling, thereby complicating relationships. It can affect up to 8% of patients with PD.35

SUMMARY

Dementia, psychotic symptoms, mood disturbances, and impulse control disorders are important nonmotor manifestations of PD that present management challenges. Some of these manifestations are intrinsic to PD, and some are complications of therapies used to treat the motor manifestations of PD.

Dementia and psychotic symptoms extract a considerable toll on the patient, caregivers, and society. Psychotic symptoms generally manifest as hallucinations (mostly visual) and other sensory disturbances. Initial management involves adjustment of anti-PD medications. The use of atypical antipsychotic drugs has been shown to improve survival among patients with PD. Clozapine is the preferred agent.

Mood disturbances such as depression and apathy may be difficult to diagnose. Depression may be treated similarly to depression unassociated with PD.

Dopamine agonists and levodopa have been associated with impulse control disorders in PD. Compulsive disorders, which are distinct from impulse control disorders, may improve with reduction or discontinuation of levodopa therapy.

Although the definition of Parkinson disease (PD) is based on the presence of motor features, these are just the “tip of the iceberg.” Nonmotor manifestations are nearly ubiquitous in PD, with behavior problems often being the most malignant. Almost all patients with PD have nonmotor and neuropsychiatric features, including sleep disturbances, compulsive and impulsive behaviors, autonomic dysfunction, and psychosis.

The neuropsychiatric and behavioral features of PD can be classified as intrinsic features, which occur as part of PD, and iatrogenic features, which are complications that arise from treatments used to manage the motor symptoms of PD.

DEMENTIA IN PD

An intrinsic nonmotor feature of PD is dementia, which occurs at a rate four to six times greater in patients with PD than in age-matched controls without PD.1 The prevalence of dementia in PD varies among studies and depends on the demographics of the population being studied. The cross-sectional prevalence of dementia is 40% in patients with PD.2 Seventy-eight percent of a population-based, representative cohort of patients with PD developed dementia during an 8-year study period.3

Dementia is a burden to the caregiver, the patient, and society. Cognitive and behavioral symptoms in patients with PD are the greatest contributors to caregiver distress.4 Dementia and associated behavioral symptoms (ie, hallucinations) hasten nursing home placement, contributing to the financial burden of caring for patients with PD.5 The risk of mortality is increased when dementia develops.6

Reprinted with permission from The New England Journal of Medicine (Emre M, et al. Rivastigmine for dementia associated with Parkinson’s disease. N Engl J Med 2004; 351:2509–2518). Copyright © 2004 Massachusetts Medical Society. All rights reserved.
Figure 1. In a double-blind, placebo-controlled trial that compared rivastigmine with placebo, patients with Parkinson disease dementia who were treated with rivastigmine experienced a 3-point improvement in the Alzheimer’s Disease Assessment Scale-Cognitive Subscale (ADAS-Cog) compared with placebo.
At least one medication has shown promise in managing PD dementia. In a pivotal trial of the cholinesterase inhibitor rivastigmine, involving more than 500 patients with PD dementia, the patients randomized to rivastigmine had a 3-point improvement on the primary outcome measure—the mean change from baseline in the Alzheimer’s Disease Assessment Scale-Cognitive Subscale—compared with those randomized to placebo (Figure 1).7 This trial led to US Food and Drug Administration approval of rivastigmine for the treatment of PD dementia.

PSYCHOTIC SYMPTOMS IN PD: AN EFFECT OF EXCESS DOPAMINE STIMULATION

Most of the complications observed in PD can be explained by the dopamine effect of medications and by dopamine deficiencies. An excess of dopamine stimulation caused by administration of prodopaminergic agents manifests as dyskinesias, hallucinations, or delusions. Withdrawal of levodopa will reverse these complications but leads to dopamine deficiency and thus a worsening of PD symptoms. Most patients with PD will tolerate mild dyskinesias or hallucinations if their PD symptoms are well controlled.

The hallucinations in PD tend to be visual as opposed to auditory (as in schizophrenia). They are usually benign and involve figures of people, furry animals, or complex scenes. About 10% to 40% of hallucinations in PD are secondary auditory hallucinations, which tend to be nondistinct, non-paranoid, and often incomprehensible (ie, voices in a crowd).

In the same way, the delusions experienced in patients with PD are distinct from those in schizophrenia. The delusions in PD are usually paranoid in nature and involve stereotyped themes (ie, spousal infidelity, feelings of abandonment) rather than the grandiose delusions that are common in schizophrenia.

The reported prevalence of psychotic symptoms in PD, including hallucinations and delusions, ranges from 20% to 50%.8,9 Auditory hallucinations are a feature in about 10%, and they usually occur with visual hallucinations. Less common are delusions and hallucinations with loss of insight, which are more likely with increasing severity of dementia.

Once a PD patient experiences hallucinations, they are likely to continue. In a 6-year longitudinal study, the prevalence of hallucinations increased from 33% at baseline to 55% at 72 months.10 Persistent psychosis was found in 69% of participants in the Psychosis and Clozapine in PD Study (PSYCLOPS) with 26 months of follow-up.11

High caregiver burden

Psychotic symptoms in PD are associated with high caregiver stress and increased rates of nursing home placement. Goetz et al12 showed that PD patients with psychosis had a much greater risk of nursing home placement than those without psychosis. The prognosis for PD patients in extended-care facilities is worse for those with psychotic symptoms.13

Management of psychotic symptoms

The first step in managing psychosis in PD is to rule out other causes of changes in mental status, such as infection, electrolyte imbalance, or introduction of new medications.

Adjusting anti-PD medications to a tolerable yet effective dose may help to reduce the incidence and severity of psychotic complications. If necessary, selective discontinuation of anti-PD medications may be tried in the following sequence: anticholinergics, amantadine, monoamine oxidase B inhibitors, dopamine agonists, catechol-O-methyltransferase inhibitors, and levodopa/carbidopa.

If motor symptoms prevent dosage minimization or discontinuation of some medications, then the addition of an atypical antipsychotic medication should be considered. Before the advent of atypical antipsychotics, the management of psychosis and hallucinations in PD was unsatisfactory, reflected by a mortality of 100% within 2 years among psychotic PD patients placed in nursing homes compared with 32% among age-matched community dwellers.13 The introduction of atypical antipsychotics has improved survival among PD patients with psychosis. In one study, mortality over 5 years was 44% among PD patients taking long-term clozapine for the treatment of psychosis.14 Recurrence of psychosis is rapid (within 8 weeks) even when PD patients are slowly weaned from atypical antipsychotics.15

Receptor affinities differ among antipsychotics. Because dopamine has been implicated as the principal neurotransmitter in the development of PD psychosis, atypical antipsychotics, with milder dopamine-blocking action, have played a central role in the treatment of PD psychosis. The dopamine D2 receptor is the main target for conventional antipsychotic drugs to exert their clinical effects. Atypical antipsychotics have different affinities for the D2 receptors.16 Occupancy of D2 receptors with atypical antipsychotics is 40% to 70% (risperidone and olanzapine have higher affinity for the D2 receptor than clozapine and quetiapine), and affinity for 5-HT2A receptors can be as high as 70%. This affinity for 5-HT2A receptors relative to D2 receptors may be important for therapeutic efficacy of the atypical antipsychotics. Antagonism of muscarinic, histaminergic, noradrenergic, and other serotonergic receptors also differs among the atypical antipsychotics.

Clozapine remains the gold standard atypical antipsychotic agent, based on results from three relatively small (N = 6 to 60) double-blind, placebo-controlled studies in PD patients with dopaminergic drug-induced psychosis.17–19 Quetiapine improved psychotic symptoms associated with PD in several open-label studies, but has not demonstrated the same success in double-blind clinical trials.20,21

Loss of cholinergic neurons and implications for treatment. In autopsy studies, the loss of cholinergic neurons is more profound in PD than in Alzheimer disease, which suggests that procholinergic drugs may improve symptoms of PD dementia, a major risk factor for hallucinations. In open-label studies, acetylcholinesterase inhibitors have reduced the frequency of hallucinations in patients who have dementia with Lewy bodies (DLB) and in patients with PD dementia. Double-blind trials of patients with DLB and PD dementia concentrated on the effect of cholinesterase inhibitors on dementia and not hallucinations. One concern with the use of a procholinergic drug in patients with PD has been worsening of parkinsonism, but studies of acetylcholinesterase inhibitors have shown no worsening of parkinsonism and only transient worsening of tremor.

Ondansetron, a 5-HT3 receptor antagonist used as an antinausea medication, produced moderate improvements in hallucinations and delusions in an open-label trial for the treatment of psychosis in advanced PD.22 For PD patients with psychosis and comorbid depression, antidepressant therapy and electroconvulsive therapy may be effective options.23,24

 

 

MOOD DISTURBANCES IN PD

Depression and apathy occur more frequently in patients with PD than in those who do not have PD.

Depression

Challenges in the management of depression in PD include recognition of depression and distinguishing depressive disorders from mood fluctuations. Whereas a depressive disorder lasts from weeks to years and can occur at any stage of illness, mood fluctuations can change many times daily and appear as nonmotor manifestations during the “off” medication state. Mood fluctuations occur mostly in patients who have developed motor fluctuations. The implication for treatment is that the treatment strategy for a depressive disorder is antidepressant therapy, whereas the strategy for mood fluctuations in PD is to increase the levodopa dose.

Recognition of depression in PD is confounded by the depression criteria of the Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition; many of these criteria can be intrinsic features of PD itself—for example, anhedonia, weight/appetite loss or gain, insomnia or hypersomnia, psychomotor retardation, and fatigue. Questions such as “are you feeling sad” or “are you feeling blue” may be superior to questions about associative symptoms when evaluating PD patients for depression.

Reprinted with permission from Parkinsonism and Related Disorders (Shulman LM, et al. Non-recognition of depression and other non-motor symptoms in Parkinson’s disease. Parkinsonism Relat Disord 2002; 8:193–197). © 2002 Elsevier Science Ltd.
Figure 2. A comparison of routine office assessment with the use of standardized rating scales that identify nonmotor symptoms demonstrated superiority of rating scales compared with neurologists’ impressions in identification of depression and other nonmotor complications of Parkinson disease. Investigators used the Beck Depression Inventory, the Beck Anxiety Inventory, the Fatigue Severity Scale, and the Pittsburg Sleep Quality Index.
The value of rating scales also should not be overlooked. Shulman et al25 found that the use of standardized rating scales is superior to routine office assessment by neurologists in recognizing depression in PD patients; in more than 50% of routine office assessments, neurologists missed a diagnosis of depression (Figure 2).

Most of the medications used for the treatment of depression also work well for depression in patients with PD. Double-blind controlled studies have demonstrated superiority of nortriptyline, citalopram, desipramine, and pramipexole over placebo in improving mood.26–29

Apathy

The overlap between apathy and depressive symptoms can also complicate recognition of apathy, which can be described as a lack of motivation or failure to initiate goal-directed behavior. Apathy involves three domains30:

  • Cognitive: expressed as a loss of interest in new experience or a lack of concern about a personal problem
  • Diminished affect: flattened affect or a lack of reaction to positive or negative events
  • Final: diminished goal-directed cognition, as indicated by a lack of effort or requiring others to structure activities.

Unlike depression, which is similarly representative of PD and other episodic conditions such as dystonia, apathy is more common in PD than in dystonia. In fact, the occurrence of apathy alone distinguishes PD from dystonia. Apathy in PD has no known treatment. If it is associated with depression, apathy may respond to antidepressants.

Repetitive transcranial magnetic stimulation (rTMS) manipulates activity in specific brain neural circuits through the skull to induce changes in behavior. Some studies suggest that modulation of behavior may last beyond the actual stimulation. A randomized, sham-controlled trial of rTMS over the middorsolateral frontal cortex has been conducted with the primary aim of improving apathy in PD. Unfortunately, while patients who were randomized to rTMS experienced some improvement in apathy during the study, the improvement was not significantly different from that observed in patients who received sham treatment.31

IMPULSE CONTROL AND COMPULSIVE DISORDERS IN PD

Impulse control disorders are characterized by the inability to resist an urge to act; the resulting irrational desire to pursue self-gratification may inflict suffering on friends and relatives that compromises relationships and impairs social- and work-related functioning.

Examples of impulse control disorders in PD are pathologic gambling, hypersexuality, compulsive shopping, excessive spending, and binge eating. Patients taking dopamine agonists are two to three times more likely to develop impulse control disorders than those receiving other treatments for PD. Dopamine agonists with relative selectivity for D3 receptors have been implicated in impulse control disorders in PD because D3 receptors are abundant in a region of the brain (ventral striatum) associated with behavioral and substance addictions. Higher levodopa dosages were also associated with impulse control disorders.

Factors associated with impulse control disorders in PD are young age, being single, a family history of impulse control disorders, and levodopa treatment.32 Modifications to dopamine agonist or levodopa therapy are important in the treatment of dopamine agonist–induced impulse disorders.

Compulsive disorders have been described as a class distinct from impulse control disorders and involve repetitive stereotypes and well-ordered acts to decrease inner anxiety and avoid harm. Punding is the engagement of stereotyped behaviors that are repeated compulsively—for example, repetitive manipulation of technical equipment; continual handling, sorting, and examining of objects; grooming; and hoarding. The punder has poor insight into the disruptive and senseless nature of his or her acts. Punding has consistently been related to dopaminergic therapy. Its prevalence in PD patients on dopaminergic therapy ranges from 1.4%33 to 14%.34 An improvement in behavior is observed with a reduction in dosage or discontinuation of levodopa.

Pathologic gambling, or the inability to control gambling, can result in lying to obtain money for gambling, thereby complicating relationships. It can affect up to 8% of patients with PD.35

SUMMARY

Dementia, psychotic symptoms, mood disturbances, and impulse control disorders are important nonmotor manifestations of PD that present management challenges. Some of these manifestations are intrinsic to PD, and some are complications of therapies used to treat the motor manifestations of PD.

Dementia and psychotic symptoms extract a considerable toll on the patient, caregivers, and society. Psychotic symptoms generally manifest as hallucinations (mostly visual) and other sensory disturbances. Initial management involves adjustment of anti-PD medications. The use of atypical antipsychotic drugs has been shown to improve survival among patients with PD. Clozapine is the preferred agent.

Mood disturbances such as depression and apathy may be difficult to diagnose. Depression may be treated similarly to depression unassociated with PD.

Dopamine agonists and levodopa have been associated with impulse control disorders in PD. Compulsive disorders, which are distinct from impulse control disorders, may improve with reduction or discontinuation of levodopa therapy.

References
  1. Aarsland D, Andersen K, Larsen JP, Lolk A, Nielsen H, Kragh-Sørensen P. Risk of dementia in Parkinson’s disease: a community-based, prospective study. Neurology 2001; 56:730736.
  2. Cummings JL. Intellectual impairment in Parkinson’s disease: clinical, pathologic, and biochemical correlates. J Geriatr Psychiatry Neurol 1988; 1:2436.
  3. Aarsland D, Andersen K, Larsen JP, Lolk A, Kragh-Sørensen P. Prevalence and characteristics of dementia in Parkinson disease: an 8-year prospective study. Arch Neurol 2003; 60:387392.
  4. Aarsland D, Larsen JP, Karlsen K, Lim NG, Tandberg E. Mental symptoms in Parkinson’s disease are important contributors to caregiver distress. Int J Geriatr Psychiatry 1999; 14:866874.
  5. Aarsland D, Larsen JP, Tandberg E, Laake K. Predictors of nursing home placement in Parkinson’s disease: a population-based, prospective study. J Am Geriatr Soc 2000; 48:938942.
  6. Hughes TA, Ross HF, Mindham RH, Spokes EG. Mortality in Parkinson’s disease and its association with dementia and depression. Acta Neurol Scand 2004; 110:118123.
  7. Emre M, Aarsland D, Albanese A, et al. Rivastigmine for dementia associated with Parkinson’s disease. N Engl J Med 2004; 351:25092518.
  8. Fénelon G, Mahieux F, Huon R, Ziégler M. Hallucinations in Parkinson’s disease: prevalence, phenomenology and risk factors. Brain 2000; 123:733745.
  9. Zahodne LB, Fernandez HH. Pathophysiology and treatment of psychosis in Parkinson’s disease: a review. Drugs Aging 2008; 25:665682.
  10. Goetz CG, Wuu J, Curgian LM, Leurgans S. Hallucinations and sleep disorders in PD: six-year prospective longitudinal study. Neurology 2005; 64:8186.
  11. Factor SA, Feustel PJ, Friedman JH, et al. Longitudinal outcome of Parkinson’s disease patients with psychosis. Neurology 2003; 60:17561761.
  12. Goetz CG, Stebbins GT. Risk factors for nursing home placement in advanced Parkinson’s disease. Neurology 1993; 43:22272229.
  13. Goetz CG, Stebbins GT. Mortality and hallucinations in nursing home patients with advanced Parkinson’s disease. Neurology 1995; 45:669671.
  14. Fernandez HH, Donnelly EM, Friedman JH. Long-term outcome of clozapine use for psychosis in parkinsonian patients. Mov Disord 2004; 19:831833.
  15. Fernandez HH, Trieschmann ME, Okun MS. Rebound psychosis: effect of discontinuation of antipsychotics in Parkinson’s disease. Mov Disord 2005; 20:104115.
  16. Goldstein JM. Atypical antipsychotic drugs: beyond acute psychosis, new directions. Emerging Drugs 1999; 4:127151.
  17. Pollak P, Tison F, Rascol O, et al; on behalf of the French Clozapine Parkinson Study Group. Clozapine in drug induced psychosis in Parkinson’s disease: a randomised, placebo controlled study with open follow up. J Neurol Neurosurg Psychiatry 2004; 75:689695.
  18. The Parkinson Study Group. Low-dose clozapine for the treatment of drug-induced psychosis in Parkinson’s disease. N Engl J Med 1999; 340:757763.
  19. Wolters ECh, Hurwitz TA, Mak E, et al. Clozapine in the treatment of parkinsonian patients with dopaminomimetic psychosis. Neurology 1990; 40:832834.
  20. Ondo WG, Tintner R, Voung KD, Lai D, Ringholz G. Double-blind, placebo-controlled, unforced titration parallel trial of quetiapine for dopaminergic-induced hallucinations in Parkinson’s disease. Mov Disord 2005; 20:958963.
  21. Fernandez HH, Okun MS, Rodriguez RL, Malaty IA, Romrell J. Quetiapine improves visual hallucinations in Parkinson disease but not through normalization of sleep architecture: results from a double-blind clinical-polysomnography study. Int J Neurosci 2009; 119:21962205.
  22. Zoldan J, Friedberg G, Livneh M, Melamed E. Psychosis in advanced Parkinson’s disease: treatment with ondansetron, a 5-HT3 receptor antagonist. Neurology 1995; 45:13051308.
  23. Voon V, Lang AE. Antidepressants in the treatment of psychosis with comorbid depression in Parkinson disease. Clin Neuropharmacol 2004; 27:9092.
  24. Ozer F, Meral H, Aydin B, Hanoglu L, Aydemir T, Oral T. Electroconvulsive therapy in drug-induced psychiatric states and neuroleptic malignant syndrome. J ECT 2005; 21:125127.
  25. Shulman LM, Taback RL, Rabinstein AA, Weiner WJ. Non-recognition of depression and other non-motor symptoms in Parkinson’s disease. Parkinsonism Relat Disord 2002; 8:193197.
  26. Devos D, Dujardin K, Poirot I, et al. Comparison of desipramine and citalopram treatments for depression in Parkinson’s disease: a double-blind, randomized, placebo-controlled study. Mov Disord 2008; 23:850857.
  27. Menza M, Dobkin RD, Marin H, et al. A controlled trial of antidepressants in patients with Parkinson disease and depression. Neurology 2009; 72:886892.
  28. Barone P, Poewe W, Albrecht S, et al. Pramipexole for the treatment of depressive symptoms in patients with Parkinson’s disease: a randomised, double-blind, placebo-controlled trial. Lancet Neurol 2010; 9:573580.
  29. Fernandez HH, Merello M. Pramipexole for depression and motor symptoms in Parkinson disease: can we kill two birds with one stone? Lancet Neurol 2010; 9:556557.
  30. Marin RS. Apathy: a neuropsychiatric syndrome. J Neuropsychiatry Clin Neurosci 1991; 3:243254.
  31. Fernandez HH, Bowers D, Triggs WJ, et al. Repetitive transcranial magnetic stimulation for the treatment of apathy in Parkinson’s disease: results from a double-blind, sham-controlled, randomized, controlled trial. Neurology 2010; 74( suppl 2):352.
  32. Weintraub D, Koester J, Potenza MN, et al. Impulse control disorders in Parkinson disease: a cross-sectional study of 3090 patients. Arch Neurol 2010; 67:589595.
  33. Miyasaki JM, Al Hassan K, Lang AE, Voon V. Punding prevalence in Parkinson’s disease. Mov Disord 2007; 22:11791181.
  34. Evans AH, Katzenschlager R, Paviour D, et al. Punding in Parkinson’s disease: its relation to the dopamine dysregulation syndrome. Mov Disord 2004; 19:397405.
  35. Grosset KA, Macphee G, Pal G, et al. Problematic gambling on dopamine agonists: not such a rarity. Mov Disord 2006; 21:22062208.
References
  1. Aarsland D, Andersen K, Larsen JP, Lolk A, Nielsen H, Kragh-Sørensen P. Risk of dementia in Parkinson’s disease: a community-based, prospective study. Neurology 2001; 56:730736.
  2. Cummings JL. Intellectual impairment in Parkinson’s disease: clinical, pathologic, and biochemical correlates. J Geriatr Psychiatry Neurol 1988; 1:2436.
  3. Aarsland D, Andersen K, Larsen JP, Lolk A, Kragh-Sørensen P. Prevalence and characteristics of dementia in Parkinson disease: an 8-year prospective study. Arch Neurol 2003; 60:387392.
  4. Aarsland D, Larsen JP, Karlsen K, Lim NG, Tandberg E. Mental symptoms in Parkinson’s disease are important contributors to caregiver distress. Int J Geriatr Psychiatry 1999; 14:866874.
  5. Aarsland D, Larsen JP, Tandberg E, Laake K. Predictors of nursing home placement in Parkinson’s disease: a population-based, prospective study. J Am Geriatr Soc 2000; 48:938942.
  6. Hughes TA, Ross HF, Mindham RH, Spokes EG. Mortality in Parkinson’s disease and its association with dementia and depression. Acta Neurol Scand 2004; 110:118123.
  7. Emre M, Aarsland D, Albanese A, et al. Rivastigmine for dementia associated with Parkinson’s disease. N Engl J Med 2004; 351:25092518.
  8. Fénelon G, Mahieux F, Huon R, Ziégler M. Hallucinations in Parkinson’s disease: prevalence, phenomenology and risk factors. Brain 2000; 123:733745.
  9. Zahodne LB, Fernandez HH. Pathophysiology and treatment of psychosis in Parkinson’s disease: a review. Drugs Aging 2008; 25:665682.
  10. Goetz CG, Wuu J, Curgian LM, Leurgans S. Hallucinations and sleep disorders in PD: six-year prospective longitudinal study. Neurology 2005; 64:8186.
  11. Factor SA, Feustel PJ, Friedman JH, et al. Longitudinal outcome of Parkinson’s disease patients with psychosis. Neurology 2003; 60:17561761.
  12. Goetz CG, Stebbins GT. Risk factors for nursing home placement in advanced Parkinson’s disease. Neurology 1993; 43:22272229.
  13. Goetz CG, Stebbins GT. Mortality and hallucinations in nursing home patients with advanced Parkinson’s disease. Neurology 1995; 45:669671.
  14. Fernandez HH, Donnelly EM, Friedman JH. Long-term outcome of clozapine use for psychosis in parkinsonian patients. Mov Disord 2004; 19:831833.
  15. Fernandez HH, Trieschmann ME, Okun MS. Rebound psychosis: effect of discontinuation of antipsychotics in Parkinson’s disease. Mov Disord 2005; 20:104115.
  16. Goldstein JM. Atypical antipsychotic drugs: beyond acute psychosis, new directions. Emerging Drugs 1999; 4:127151.
  17. Pollak P, Tison F, Rascol O, et al; on behalf of the French Clozapine Parkinson Study Group. Clozapine in drug induced psychosis in Parkinson’s disease: a randomised, placebo controlled study with open follow up. J Neurol Neurosurg Psychiatry 2004; 75:689695.
  18. The Parkinson Study Group. Low-dose clozapine for the treatment of drug-induced psychosis in Parkinson’s disease. N Engl J Med 1999; 340:757763.
  19. Wolters ECh, Hurwitz TA, Mak E, et al. Clozapine in the treatment of parkinsonian patients with dopaminomimetic psychosis. Neurology 1990; 40:832834.
  20. Ondo WG, Tintner R, Voung KD, Lai D, Ringholz G. Double-blind, placebo-controlled, unforced titration parallel trial of quetiapine for dopaminergic-induced hallucinations in Parkinson’s disease. Mov Disord 2005; 20:958963.
  21. Fernandez HH, Okun MS, Rodriguez RL, Malaty IA, Romrell J. Quetiapine improves visual hallucinations in Parkinson disease but not through normalization of sleep architecture: results from a double-blind clinical-polysomnography study. Int J Neurosci 2009; 119:21962205.
  22. Zoldan J, Friedberg G, Livneh M, Melamed E. Psychosis in advanced Parkinson’s disease: treatment with ondansetron, a 5-HT3 receptor antagonist. Neurology 1995; 45:13051308.
  23. Voon V, Lang AE. Antidepressants in the treatment of psychosis with comorbid depression in Parkinson disease. Clin Neuropharmacol 2004; 27:9092.
  24. Ozer F, Meral H, Aydin B, Hanoglu L, Aydemir T, Oral T. Electroconvulsive therapy in drug-induced psychiatric states and neuroleptic malignant syndrome. J ECT 2005; 21:125127.
  25. Shulman LM, Taback RL, Rabinstein AA, Weiner WJ. Non-recognition of depression and other non-motor symptoms in Parkinson’s disease. Parkinsonism Relat Disord 2002; 8:193197.
  26. Devos D, Dujardin K, Poirot I, et al. Comparison of desipramine and citalopram treatments for depression in Parkinson’s disease: a double-blind, randomized, placebo-controlled study. Mov Disord 2008; 23:850857.
  27. Menza M, Dobkin RD, Marin H, et al. A controlled trial of antidepressants in patients with Parkinson disease and depression. Neurology 2009; 72:886892.
  28. Barone P, Poewe W, Albrecht S, et al. Pramipexole for the treatment of depressive symptoms in patients with Parkinson’s disease: a randomised, double-blind, placebo-controlled trial. Lancet Neurol 2010; 9:573580.
  29. Fernandez HH, Merello M. Pramipexole for depression and motor symptoms in Parkinson disease: can we kill two birds with one stone? Lancet Neurol 2010; 9:556557.
  30. Marin RS. Apathy: a neuropsychiatric syndrome. J Neuropsychiatry Clin Neurosci 1991; 3:243254.
  31. Fernandez HH, Bowers D, Triggs WJ, et al. Repetitive transcranial magnetic stimulation for the treatment of apathy in Parkinson’s disease: results from a double-blind, sham-controlled, randomized, controlled trial. Neurology 2010; 74( suppl 2):352.
  32. Weintraub D, Koester J, Potenza MN, et al. Impulse control disorders in Parkinson disease: a cross-sectional study of 3090 patients. Arch Neurol 2010; 67:589595.
  33. Miyasaki JM, Al Hassan K, Lang AE, Voon V. Punding prevalence in Parkinson’s disease. Mov Disord 2007; 22:11791181.
  34. Evans AH, Katzenschlager R, Paviour D, et al. Punding in Parkinson’s disease: its relation to the dopamine dysregulation syndrome. Mov Disord 2004; 19:397405.
  35. Grosset KA, Macphee G, Pal G, et al. Problematic gambling on dopamine agonists: not such a rarity. Mov Disord 2006; 21:22062208.
Page Number
S14-S18
Page Number
S14-S18
Publications
Publications
Article Type
Display Headline
Nonmotor complications of Parkinson disease
Display Headline
Nonmotor complications of Parkinson disease
Citation Override
Cleveland Clinic Journal of Medicine 2012 July;79(suppl 2):S14-S18
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media

Deep brain stimulation for movement disorders: Patient selection and technical options

Article Type
Changed
Mon, 10/01/2018 - 15:35
Display Headline
Deep brain stimulation for movement disorders: Patient selection and technical options

Implantation of a deep brain stimulator is the most common surgical procedure performed in the United States and industrialized world for the management of advanced movement disorders. These procedures are US Food and Drug Administration (FDA)–approved for the management of the symptoms of Parkinson disease (PD) and essential tremor. Deep brain stimulation (DBS) is also approved for managing primary generalized dystonia and torticollis under a humanitarian device exemption.

Deep brain stimulation has largely replaced ablative procedures such as thalamotomy and pallidotomy. While ablative procedures can be effective for the symptoms of movement disorders, they cause a permanent lesion in the targeted nuclei and are therefore not reversible. DBS is considered safer because it can be adjusted over time and the location of the leads can be revised.1 On the other hand, regular maintenance of implanted hardware may be considered a disadvantage of DBS.

HARDWARE AND TARGETS

While ablative procedures do not require implantable hardware, DBS consists of permanently implanted neurostimulation systems. The battery-powered pulse generators typically last for several years but require multiple replacements during a lifetime. In addition, if other hardware components fail, surgical revision may be required to maintain treatment efficacy. Surgery involving implantation of hardware carries a higher risk of infection than does a nonimplantation procedure. If infections occur, removal of the hardware is often required, with reimplantation performed after the infection clears. In addition, the expense of DBS hardware may limit availability in some cases.

Three components

Figure 1. The components of an implantable deep brain stimulation system.
Permanently implanted DBS devices have three components: the DBS lead, which is inserted into the brain and extends to the outside of the skull; the implantable pulse generator, typically located in the infraclavicular area; and an extension cable that connects the two components (Figure 1). Patients may have unilateral or bilateral lead implantation and unilateral or bilateral implantation of pulse generators. A single generator may be connected to both brain leads. Patients also have the option of receiving either a nonrechargeable or a rechargeable pulse generator. The advantage of the latter is longer intervals between battery replacement surgery (up to 9 years). However, these require more maintenance by the patient, who needs to periodically recharge the generators at home using a wireless charging unit. The recharging procedure may be time consuming and difficult for patients who are challenged by new technologies. In our experience, most patients with PD and tremor prefer nonrechargeable pulse generators.

Target nuclei

Several nodes or nuclei can serve as targets for DBS. In patients with PD, the most common surgical target is the subthalamic nucleus (STN), either unilaterally or bilaterally.2 The globus pallidus pars interna (GPi) is also a viable target and is preferred for some patients with PD. The most common target for managing essential tremor is the ventral intermediate nucleus (VIM) of the thalamus, which can also be the target of choice for patients with tremor-predominant PD. However, the GPi and STN are usually preferred over the VIM in patients with PD because stimulation of these targets can relieve symptoms other than tremor, such as rigidity and bradykinesia. Bilateral stimulation of the GPi is the most frequent approach in patients with generalized torsion dystonia and torticollis, although the STN and thalamic nuclei (off-label) are also considered options.

PATIENT SELECTION

Patients are evaluated in our center at Cleveland Clinic by a multidisciplinary team that includes a movement disorder neurologist, a subspecialized neurosurgeon, a movement disorder neuropsychologist, and a psychiatrist with special interest in the behavioral comorbidities of movement disorders.3 Neuroimaging is included in this assessment. We have also included physical therapy as part of the initial evaluation in order to gain insight into the patient’s limitations and develop rehabilitation strategies that may enhance the outcomes of surgery or provide alternatives should surgery not be indicated. This evaluation provides extensive data that are then reviewed by the team in a conference dedicated to discussing candidacy for DBS or options for managing the symptoms of advanced movement disorders. Behavioral and cognitive issues are assessed in detail and, in our experience, are the most common reasons for not recommending DBS.

An important part of the evaluation of patients with PD is a formal test with rating of the motor section of the Unified Parkinson’s Disease Rating Scale (UPDRS) with the patient off medications for 8 to 12 hours and then after a test dose of levodopa. At our center, this off/on test is videotaped so that the responsiveness of individual symptoms to levodopa can be reviewed later in conference.

Risk of cognitive decline

While DBS is considered safe and effective, there is a risk of cognitive decline in some patients. In most patients, long-term stimulation-related cognitive decline may be detected with formal measures but is not clinically significant and is outweighed by the motor and quality-of-life benefits of surgery. In some patients, long-term cognitive decline can be significant and can limit function. Cognitive neuropsychologic testing provides valuable information in this regard. Patients with preserved cognitive function seldom experience significant decline with DBS while those with substantial baseline impairment are thought to be at greater risk. Patients who meet criteria for dementia are usually not considered candidates for DBS, but exceptions exist. Transient perioperative cognitive difficulties are more common than persistent deficits, and typically resolve within a few weeks (see “Complications of deep brain stimulation”).

Benefits in Parkinson disease

Deep brain stimulation can address several symptoms of PD but with varying effects. Tremor, rigidity, and bradykinesia usually improve substantially. Gait has a more variable response, and balance is typically refractory. A general rule is that symptoms that improve with a single dose of levodopa should also improve with DBS. (Tremor, however, will most often respond to DBS even if refractory to medication.) Good candidates for surgery typically have a greater than 30% improvement in UPDRS motor score with levodopa challenge, but sometimes, improvement in the total score is less informative than evaluation of the effects of levodopa on particular symptoms. Treatment effects can be compared with the patient’s expectations for surgery in order to infer whether the goals for symptom improvement are realistic.

 

 

Treatment outcomes depend on etiology

After programming, DBS can provide PD symptom control similar to that of medication “on time,” but with fewer on-off fluctuations and less on-time dyskinesia. Good surgical candidates are patients who once responded well to dopaminergic medications but who, after several years with the disease, present with increased duration of “off time,” unpredictable duration of on time, and medication side effects such as on-time dyskinesia. Patients who do not respond well to levodopa even in subscores of the UPDRS may not be good candidates for DBS, and in some cases the diagnosis itself needs to be reviewed.

Deep brain stimulation can improve quality of life and alleviate symptoms of essential tremor. Tremor control is best for the upper extremities and tends to be better for distal tremors than for proximal ones. Patients who are good candidates for surgery often have severe tremors. A substantial improvement in these symptoms often has a dramatic, positive effect on work and quality of life. In some patients, surgery is considered for mild tremor if it seriously disrupts the patient’s lifestyle or occupation and cannot be well controlled with medications. Often, in these cases, tremor that appears relatively mild to the examiner is significantly limiting for the patient.

Very severe and proximal tremor is more refractory, though it may also improve. The changes can be well documented with objective measures. In these cases, however, residual tremor can still be moderate to severe and can be functionally limiting. Head or vocal tremors are typically refractory. They may be improved with bilateral implantation, but this cannot be accurately predicted. Patients who present with head-only or head-predominant tremor are thought to be less likely to benefit than those with limb tremor. Nonetheless, tremors of the head can severely impair quality of life. Because there are few other treatment options, some patients choose DBS with the understanding that the outcome is uncertain and the benefit may be limited.

Tremor resulting from multiple sclerosis or other causes can be medically refractory and disabling. In our experience, DBS can be an off-label option for managing secondary tremors and good outcomes have been observed. However, outcomes are much less predictable and tremor control less effective than in patients with essential tremor.

Patients with primary generalized dystonia can be considered candidates for DBS and may experience improved symptom control and quality of life.4 Patients with the DYT1 mutation are more likely to respond well to DBS, as are those with other forms of primary generalized dystonia. In contrast to that seen in patients with PD and tremor, symptomatic improvement is frequently not observed during intraoperative testing. Several months of stimulation and programming may be required before significant improvements are detected.5 Surgery can also be considered for off-label use in the treatment of patients with secondary dystonia—such as that following injury or associated with cerebral palsy—but outcomes are less predictable and usually more limited. A possible exception may be seen in cases of tardive dystonia, for which there is increasing evidence6 for the effectiveness of DBS. This remains an off-label use of DBS.

Realistic expectations

An important aspect of the multidisciplinary evaluation includes a discussion of the expectations for surgery, the risks, and the requirements for postoperative care. As discussed above, DBS is reversible and adjustable, so outcomes depend not only on accurate implantation of the hardware but also on postoperative programming. Also, monitoring and maintenance of the implanted hardware are required in these patients. It is important that patients and families appreciate the fact that specialized, long-term postoperative follow-up is as much a part of the treatment as is the implantation itself.

UNILATERAL VERSUS BILATERAL DBS

Most patients with generalized dystonia undergo bilateral DBS. However, patients with PD or essential tremor may receive bilateral, staged, or unilateral implants. Some patients with PD present with either near-complete predominance of symptoms on one side or with symptoms that affect mostly the dominant extremity. In these patients, unilateral implantation is often recommended because it has less risk than the bilateral approach and may be sufficient to address the most limiting symptoms.

As the disease advances, an additional surgery may be required to accomplish bilateral symptom control. Nevertheless, we do not routinely recommend preventive implantation because it is not known whether second-side symptoms will become severe enough to require it. This strategy allows for deferring surgical risk, which is in itself advantageous. In our experience, bilateral implantation is often recommended to PD patients who present with symptoms such as freezing of gait.

Patients who have essential tremor often present with bilateral symptoms. Although many patients will indicate that they need symptom relief on both upper extremities in order to perform activities of daily living, our practice is to recommend surgery on one side at first and to suggest the patient consider contralateral implantation after weeks or months. Bilateral implantation may carry a risk for dysarthria and the risk is thought to be reduced if bilateral procedures are staged. Although high rates of dysarthria have been reported following bilateral surgery for tremor, its occurrence has been infrequent in our experience with bilateral staged DBS. Benefits of treating tremor in the dominant extremity usually exceed those of treating nondominant tremor, so most patients prefer that the dominant side be the first one treated.

TECHNICAL OPTIONS

There are several technical options for implantation of DBS systems. Stereotactic procedures rely on co-registration of preoperative imaging with external and internal fiducials, or points of reference. Targeting of the intended structures is performed by combining direct and indirect methods. Direct methods rely on identification of the target structures with imaging, such as visualization of the STN and GPi on preoperative magnetic resonance imaging (MRI). Indirect targeting relies on cadaveric anatomic atlases and coordinate systems that infer the location of the intended structures in relation to anatomical points of reference.

Frame-based systems

Figure 2. In a frame-based system, the stereotactic arc is attached to the base of the frame. Entry points of the leads are marked on the skin and on the skull.
In the most common approach to DBS surgery,7 stereotactic frames are placed over the patient’s head and secured with pins. The frame becomes the fixed point of reference for accurate stereotactic surgery and must remain in place for the duration of the procedure. Computed tomography or MRI is then performed with the frame in place, so that the images are co-registered with the fiducial points of the stereotactic frame. The targets are then selected for surgery and trajectories are chosen based on anatomic structures. The patient is positioned supine and the frame and head are secured to the operating table. The coordinates calculated by the clinical workstations are then set to the stereotactic frame and arc. The stereotactic arc (Figure 2) is attached to the base of the frame and the entry points of the leads—where the burr hole will be placed—are marked on the skin and then on the skull. Once the burr hole and opening of the meninges are completed, the targeting cannulae are inserted. The microelectrode system is then mounted for recording of the target area and subsequently for final lead implantation.

Frameless systems

Figure 3. Surgeon’s view of the frameless device, placed over the head approximately at the level of the coronal suture. The occipital area is in the bottom of the figure. When a frameless system is used, a lighter-weight structure is affixed to the head.
The workflow and overall surgical procedure for implantation of DBS with frameless systems are similar to those of the frame-based procedure. However, instead of fixing the head to a rigid frame that prevents head motion, a lighter-weight, frameless system is fixed to the head and moves with it (Figure 3). First, metal screws and fiducials are fixed to the head under local anesthesia or sedation. Preoperative imaging is then acquired with the fiducials in place and the surgical plans are completed in the same fashion as for frame-based surgery. The patient is then placed supine on the operating table and the frameless system is attached to the head with the aid of image guidance, in the location determined by target and trajectory planning.

The key advantage of the frameless system over the frame-based system is greater mobility of the head. Another important advantage is easier access to the airway, should an emergency situation occur. In our practice, patients with experience of both frameless and frame-based systems did not report significantly less discomfort with the frameless system.

The frameless system also has disadvantages, including less secure fixation of the head, which can add risk to the procedure. In addition, because of its lightweight, plastic construction, it provides less robust support to the instrumentation entering the brain than do metallic head frames and, in some cases, there is less flexibility for adjusting targets if needed during surgery. In addition, frameless systems are nonreusable and represent a substantial additional cost.

 

 

Microelectrode recording

Physiologic verification of anatomic targets identified by imaging can be accomplished with microelectrode recording (MER). This technique involves placing fine, high-impedance electrodes through the target area, so that anatomic structures can be recognized by characteristic electrical activity of individual neurons or groups of neurons. The locations of the structures are identified and the lengths of the electrode trajectories through the different structures—as well as the gaps between these structures—are recorded. The distances are then compared with the anatomy and a best-fit model is created to infer the location of the trajectory in the target area. Additional MER penetrations are made in order to further delineate the anatomy. Once a location for implantation has been selected, the DBS lead is inserted into the target area.

Electrode implantation

Lead implantation is often performed under fluoroscopic guidance in order to ensure accuracy and stability. When implanted, the electrode may cause a microlesional effect, manifested by transient improvement in symptoms.

The DBS leads are then connected to external pulse generators and assessed for clinical benefits and side effects. Amplitude, pulse width, and frequency are adjusted to test the therapeutic window of stimulation (clinical improvement thresholds versus side effect thresholds). Some PD patients develop dyskinesia during test stimulation, which may be a positive indicator for lead location. If good effects and a therapeutic window are observed, the location of the lead is considered to be satisfactory and the procedure is completed.

Pulse generator implantation

During the final step of surgery, performed under general anesthesia, the pulse generator is implanted. The extension cable that connects the DBS lead to the implantable pulse generator is tunneled subcutaneously, connecting the DBS lead to the pulse generator in the chest.

Intraoperative, real-time MRI stereotaxis

Real-time intraoperative MRI has become available for DBS implantation with devices recently cleared for use by the FDA. The procedure, typically performed in a diagnostic MRI suite, uses MR images acquired during surgery to guide DBS lead implantation in the target area and to verify implantation accuracy.8

References
  1. Rezai AR, Machado AG, Azmi H, Kubu C, Boulis NM. Surgery for movement disorders. Neurosurg 2008; 62(SHC suppl 2):SHC809SHC839.
  2. Krack P, Batir A, Van Blercom N, et al. Five-year follow-up of bilateral stimulation of the subthalamic nucleus in advanced Parkinson’s disease. N Engl J Med 2003; 349:19251934.
  3. Machado A, Fernandez HH, Deogaonkar M. Deep brain stimulation: what can patients expect from it? Cleve Clin J Med 2012; 79:113120.
  4. Vidailhet M, Vercueil L, Houeto JL, et al. Bilateral deep-brain stimulation of the globus pallidus in primary generalized dystonia. N Engl J Med 2005; 352:459467.
  5. Kupsch A, Benecke R, Müller J, et al. Pallidal deep-brain stimulation in primary generalized or segmental dystonia. N Engl J Med 2006; 355:19781990.
  6. Gruber D, Trottenberg T, Kivi A, et al. Long-term effects of pallidal deep brain stimulation in tardive dystonia. Neurology 2009; 73:5358.
  7. Gross RE, Sharan AD, Benabid AL. Deep brain stimulation for Parkinson’s disease: surgical technique and perioperative management. Mov Disord 2006; 21( suppl 14):S247S258.
  8. Starr PA, Martin AJ, Ostrem JL, et al. Subthalamic nucleus deep brain stimulator placement using high-field interventional magnetic resonance imaging and a skull-mounted aiming device: technique and application accuracy. J Neurosurg 2010; 112:479490.
Article PDF
Author and Disclosure Information

Andre G. Machado, MD, PhD
Center for Neurological Restoration, Cleveland Clinic, Cleveland, OH

Milind Deogaonkar, MD
Center for Neurological Restoration, Cleveland Clinic, Cleveland, OH

Scott Cooper, MD, PhD
Center for Neurological Restoration, Cleveland Clinic, Cleveland, OH

Correspondence: Andre Machado, MD, PhD, Center for Neurological Restoration, Cleveland Clinic, 9500 Euclid Avenue, S31, Cleveland, OH 44195; machada@ccf.org

Dr. Machado reported ownership interest in ATI Medical Equipment Corporation, Cardionomics, and Intelect Medical, Inc.; and consulting services for Monteris Medical. Dr. Deogaonkar reported intellectual property rights with Autonomic Technologies, Inc. and consulting for Medtronic, Inc. Dr. Cooper reported that he has no financial interests or relationships that pose a potential conflict of interest with this article.

This article is based on Dr. Machado’s presentation at “The Annual Therapy Symposium on Movement Disorders for the Modern Clinician” held in Fort Lauderdale, Florida, on January 29, 2011, and was written by Drs. Machado, Deogaonkar, and Cooper.

Publications
Page Number
S19-S24
Author and Disclosure Information

Andre G. Machado, MD, PhD
Center for Neurological Restoration, Cleveland Clinic, Cleveland, OH

Milind Deogaonkar, MD
Center for Neurological Restoration, Cleveland Clinic, Cleveland, OH

Scott Cooper, MD, PhD
Center for Neurological Restoration, Cleveland Clinic, Cleveland, OH

Correspondence: Andre Machado, MD, PhD, Center for Neurological Restoration, Cleveland Clinic, 9500 Euclid Avenue, S31, Cleveland, OH 44195; machada@ccf.org

Dr. Machado reported ownership interest in ATI Medical Equipment Corporation, Cardionomics, and Intelect Medical, Inc.; and consulting services for Monteris Medical. Dr. Deogaonkar reported intellectual property rights with Autonomic Technologies, Inc. and consulting for Medtronic, Inc. Dr. Cooper reported that he has no financial interests or relationships that pose a potential conflict of interest with this article.

This article is based on Dr. Machado’s presentation at “The Annual Therapy Symposium on Movement Disorders for the Modern Clinician” held in Fort Lauderdale, Florida, on January 29, 2011, and was written by Drs. Machado, Deogaonkar, and Cooper.

Author and Disclosure Information

Andre G. Machado, MD, PhD
Center for Neurological Restoration, Cleveland Clinic, Cleveland, OH

Milind Deogaonkar, MD
Center for Neurological Restoration, Cleveland Clinic, Cleveland, OH

Scott Cooper, MD, PhD
Center for Neurological Restoration, Cleveland Clinic, Cleveland, OH

Correspondence: Andre Machado, MD, PhD, Center for Neurological Restoration, Cleveland Clinic, 9500 Euclid Avenue, S31, Cleveland, OH 44195; machada@ccf.org

Dr. Machado reported ownership interest in ATI Medical Equipment Corporation, Cardionomics, and Intelect Medical, Inc.; and consulting services for Monteris Medical. Dr. Deogaonkar reported intellectual property rights with Autonomic Technologies, Inc. and consulting for Medtronic, Inc. Dr. Cooper reported that he has no financial interests or relationships that pose a potential conflict of interest with this article.

This article is based on Dr. Machado’s presentation at “The Annual Therapy Symposium on Movement Disorders for the Modern Clinician” held in Fort Lauderdale, Florida, on January 29, 2011, and was written by Drs. Machado, Deogaonkar, and Cooper.

Article PDF
Article PDF

Implantation of a deep brain stimulator is the most common surgical procedure performed in the United States and industrialized world for the management of advanced movement disorders. These procedures are US Food and Drug Administration (FDA)–approved for the management of the symptoms of Parkinson disease (PD) and essential tremor. Deep brain stimulation (DBS) is also approved for managing primary generalized dystonia and torticollis under a humanitarian device exemption.

Deep brain stimulation has largely replaced ablative procedures such as thalamotomy and pallidotomy. While ablative procedures can be effective for the symptoms of movement disorders, they cause a permanent lesion in the targeted nuclei and are therefore not reversible. DBS is considered safer because it can be adjusted over time and the location of the leads can be revised.1 On the other hand, regular maintenance of implanted hardware may be considered a disadvantage of DBS.

HARDWARE AND TARGETS

While ablative procedures do not require implantable hardware, DBS consists of permanently implanted neurostimulation systems. The battery-powered pulse generators typically last for several years but require multiple replacements during a lifetime. In addition, if other hardware components fail, surgical revision may be required to maintain treatment efficacy. Surgery involving implantation of hardware carries a higher risk of infection than does a nonimplantation procedure. If infections occur, removal of the hardware is often required, with reimplantation performed after the infection clears. In addition, the expense of DBS hardware may limit availability in some cases.

Three components

Figure 1. The components of an implantable deep brain stimulation system.
Permanently implanted DBS devices have three components: the DBS lead, which is inserted into the brain and extends to the outside of the skull; the implantable pulse generator, typically located in the infraclavicular area; and an extension cable that connects the two components (Figure 1). Patients may have unilateral or bilateral lead implantation and unilateral or bilateral implantation of pulse generators. A single generator may be connected to both brain leads. Patients also have the option of receiving either a nonrechargeable or a rechargeable pulse generator. The advantage of the latter is longer intervals between battery replacement surgery (up to 9 years). However, these require more maintenance by the patient, who needs to periodically recharge the generators at home using a wireless charging unit. The recharging procedure may be time consuming and difficult for patients who are challenged by new technologies. In our experience, most patients with PD and tremor prefer nonrechargeable pulse generators.

Target nuclei

Several nodes or nuclei can serve as targets for DBS. In patients with PD, the most common surgical target is the subthalamic nucleus (STN), either unilaterally or bilaterally.2 The globus pallidus pars interna (GPi) is also a viable target and is preferred for some patients with PD. The most common target for managing essential tremor is the ventral intermediate nucleus (VIM) of the thalamus, which can also be the target of choice for patients with tremor-predominant PD. However, the GPi and STN are usually preferred over the VIM in patients with PD because stimulation of these targets can relieve symptoms other than tremor, such as rigidity and bradykinesia. Bilateral stimulation of the GPi is the most frequent approach in patients with generalized torsion dystonia and torticollis, although the STN and thalamic nuclei (off-label) are also considered options.

PATIENT SELECTION

Patients are evaluated in our center at Cleveland Clinic by a multidisciplinary team that includes a movement disorder neurologist, a subspecialized neurosurgeon, a movement disorder neuropsychologist, and a psychiatrist with special interest in the behavioral comorbidities of movement disorders.3 Neuroimaging is included in this assessment. We have also included physical therapy as part of the initial evaluation in order to gain insight into the patient’s limitations and develop rehabilitation strategies that may enhance the outcomes of surgery or provide alternatives should surgery not be indicated. This evaluation provides extensive data that are then reviewed by the team in a conference dedicated to discussing candidacy for DBS or options for managing the symptoms of advanced movement disorders. Behavioral and cognitive issues are assessed in detail and, in our experience, are the most common reasons for not recommending DBS.

An important part of the evaluation of patients with PD is a formal test with rating of the motor section of the Unified Parkinson’s Disease Rating Scale (UPDRS) with the patient off medications for 8 to 12 hours and then after a test dose of levodopa. At our center, this off/on test is videotaped so that the responsiveness of individual symptoms to levodopa can be reviewed later in conference.

Risk of cognitive decline

While DBS is considered safe and effective, there is a risk of cognitive decline in some patients. In most patients, long-term stimulation-related cognitive decline may be detected with formal measures but is not clinically significant and is outweighed by the motor and quality-of-life benefits of surgery. In some patients, long-term cognitive decline can be significant and can limit function. Cognitive neuropsychologic testing provides valuable information in this regard. Patients with preserved cognitive function seldom experience significant decline with DBS while those with substantial baseline impairment are thought to be at greater risk. Patients who meet criteria for dementia are usually not considered candidates for DBS, but exceptions exist. Transient perioperative cognitive difficulties are more common than persistent deficits, and typically resolve within a few weeks (see “Complications of deep brain stimulation”).

Benefits in Parkinson disease

Deep brain stimulation can address several symptoms of PD but with varying effects. Tremor, rigidity, and bradykinesia usually improve substantially. Gait has a more variable response, and balance is typically refractory. A general rule is that symptoms that improve with a single dose of levodopa should also improve with DBS. (Tremor, however, will most often respond to DBS even if refractory to medication.) Good candidates for surgery typically have a greater than 30% improvement in UPDRS motor score with levodopa challenge, but sometimes, improvement in the total score is less informative than evaluation of the effects of levodopa on particular symptoms. Treatment effects can be compared with the patient’s expectations for surgery in order to infer whether the goals for symptom improvement are realistic.

 

 

Treatment outcomes depend on etiology

After programming, DBS can provide PD symptom control similar to that of medication “on time,” but with fewer on-off fluctuations and less on-time dyskinesia. Good surgical candidates are patients who once responded well to dopaminergic medications but who, after several years with the disease, present with increased duration of “off time,” unpredictable duration of on time, and medication side effects such as on-time dyskinesia. Patients who do not respond well to levodopa even in subscores of the UPDRS may not be good candidates for DBS, and in some cases the diagnosis itself needs to be reviewed.

Deep brain stimulation can improve quality of life and alleviate symptoms of essential tremor. Tremor control is best for the upper extremities and tends to be better for distal tremors than for proximal ones. Patients who are good candidates for surgery often have severe tremors. A substantial improvement in these symptoms often has a dramatic, positive effect on work and quality of life. In some patients, surgery is considered for mild tremor if it seriously disrupts the patient’s lifestyle or occupation and cannot be well controlled with medications. Often, in these cases, tremor that appears relatively mild to the examiner is significantly limiting for the patient.

Very severe and proximal tremor is more refractory, though it may also improve. The changes can be well documented with objective measures. In these cases, however, residual tremor can still be moderate to severe and can be functionally limiting. Head or vocal tremors are typically refractory. They may be improved with bilateral implantation, but this cannot be accurately predicted. Patients who present with head-only or head-predominant tremor are thought to be less likely to benefit than those with limb tremor. Nonetheless, tremors of the head can severely impair quality of life. Because there are few other treatment options, some patients choose DBS with the understanding that the outcome is uncertain and the benefit may be limited.

Tremor resulting from multiple sclerosis or other causes can be medically refractory and disabling. In our experience, DBS can be an off-label option for managing secondary tremors and good outcomes have been observed. However, outcomes are much less predictable and tremor control less effective than in patients with essential tremor.

Patients with primary generalized dystonia can be considered candidates for DBS and may experience improved symptom control and quality of life.4 Patients with the DYT1 mutation are more likely to respond well to DBS, as are those with other forms of primary generalized dystonia. In contrast to that seen in patients with PD and tremor, symptomatic improvement is frequently not observed during intraoperative testing. Several months of stimulation and programming may be required before significant improvements are detected.5 Surgery can also be considered for off-label use in the treatment of patients with secondary dystonia—such as that following injury or associated with cerebral palsy—but outcomes are less predictable and usually more limited. A possible exception may be seen in cases of tardive dystonia, for which there is increasing evidence6 for the effectiveness of DBS. This remains an off-label use of DBS.

Realistic expectations

An important aspect of the multidisciplinary evaluation includes a discussion of the expectations for surgery, the risks, and the requirements for postoperative care. As discussed above, DBS is reversible and adjustable, so outcomes depend not only on accurate implantation of the hardware but also on postoperative programming. Also, monitoring and maintenance of the implanted hardware are required in these patients. It is important that patients and families appreciate the fact that specialized, long-term postoperative follow-up is as much a part of the treatment as is the implantation itself.

UNILATERAL VERSUS BILATERAL DBS

Most patients with generalized dystonia undergo bilateral DBS. However, patients with PD or essential tremor may receive bilateral, staged, or unilateral implants. Some patients with PD present with either near-complete predominance of symptoms on one side or with symptoms that affect mostly the dominant extremity. In these patients, unilateral implantation is often recommended because it has less risk than the bilateral approach and may be sufficient to address the most limiting symptoms.

As the disease advances, an additional surgery may be required to accomplish bilateral symptom control. Nevertheless, we do not routinely recommend preventive implantation because it is not known whether second-side symptoms will become severe enough to require it. This strategy allows for deferring surgical risk, which is in itself advantageous. In our experience, bilateral implantation is often recommended to PD patients who present with symptoms such as freezing of gait.

Patients who have essential tremor often present with bilateral symptoms. Although many patients will indicate that they need symptom relief on both upper extremities in order to perform activities of daily living, our practice is to recommend surgery on one side at first and to suggest the patient consider contralateral implantation after weeks or months. Bilateral implantation may carry a risk for dysarthria and the risk is thought to be reduced if bilateral procedures are staged. Although high rates of dysarthria have been reported following bilateral surgery for tremor, its occurrence has been infrequent in our experience with bilateral staged DBS. Benefits of treating tremor in the dominant extremity usually exceed those of treating nondominant tremor, so most patients prefer that the dominant side be the first one treated.

TECHNICAL OPTIONS

There are several technical options for implantation of DBS systems. Stereotactic procedures rely on co-registration of preoperative imaging with external and internal fiducials, or points of reference. Targeting of the intended structures is performed by combining direct and indirect methods. Direct methods rely on identification of the target structures with imaging, such as visualization of the STN and GPi on preoperative magnetic resonance imaging (MRI). Indirect targeting relies on cadaveric anatomic atlases and coordinate systems that infer the location of the intended structures in relation to anatomical points of reference.

Frame-based systems

Figure 2. In a frame-based system, the stereotactic arc is attached to the base of the frame. Entry points of the leads are marked on the skin and on the skull.
In the most common approach to DBS surgery,7 stereotactic frames are placed over the patient’s head and secured with pins. The frame becomes the fixed point of reference for accurate stereotactic surgery and must remain in place for the duration of the procedure. Computed tomography or MRI is then performed with the frame in place, so that the images are co-registered with the fiducial points of the stereotactic frame. The targets are then selected for surgery and trajectories are chosen based on anatomic structures. The patient is positioned supine and the frame and head are secured to the operating table. The coordinates calculated by the clinical workstations are then set to the stereotactic frame and arc. The stereotactic arc (Figure 2) is attached to the base of the frame and the entry points of the leads—where the burr hole will be placed—are marked on the skin and then on the skull. Once the burr hole and opening of the meninges are completed, the targeting cannulae are inserted. The microelectrode system is then mounted for recording of the target area and subsequently for final lead implantation.

Frameless systems

Figure 3. Surgeon’s view of the frameless device, placed over the head approximately at the level of the coronal suture. The occipital area is in the bottom of the figure. When a frameless system is used, a lighter-weight structure is affixed to the head.
The workflow and overall surgical procedure for implantation of DBS with frameless systems are similar to those of the frame-based procedure. However, instead of fixing the head to a rigid frame that prevents head motion, a lighter-weight, frameless system is fixed to the head and moves with it (Figure 3). First, metal screws and fiducials are fixed to the head under local anesthesia or sedation. Preoperative imaging is then acquired with the fiducials in place and the surgical plans are completed in the same fashion as for frame-based surgery. The patient is then placed supine on the operating table and the frameless system is attached to the head with the aid of image guidance, in the location determined by target and trajectory planning.

The key advantage of the frameless system over the frame-based system is greater mobility of the head. Another important advantage is easier access to the airway, should an emergency situation occur. In our practice, patients with experience of both frameless and frame-based systems did not report significantly less discomfort with the frameless system.

The frameless system also has disadvantages, including less secure fixation of the head, which can add risk to the procedure. In addition, because of its lightweight, plastic construction, it provides less robust support to the instrumentation entering the brain than do metallic head frames and, in some cases, there is less flexibility for adjusting targets if needed during surgery. In addition, frameless systems are nonreusable and represent a substantial additional cost.

 

 

Microelectrode recording

Physiologic verification of anatomic targets identified by imaging can be accomplished with microelectrode recording (MER). This technique involves placing fine, high-impedance electrodes through the target area, so that anatomic structures can be recognized by characteristic electrical activity of individual neurons or groups of neurons. The locations of the structures are identified and the lengths of the electrode trajectories through the different structures—as well as the gaps between these structures—are recorded. The distances are then compared with the anatomy and a best-fit model is created to infer the location of the trajectory in the target area. Additional MER penetrations are made in order to further delineate the anatomy. Once a location for implantation has been selected, the DBS lead is inserted into the target area.

Electrode implantation

Lead implantation is often performed under fluoroscopic guidance in order to ensure accuracy and stability. When implanted, the electrode may cause a microlesional effect, manifested by transient improvement in symptoms.

The DBS leads are then connected to external pulse generators and assessed for clinical benefits and side effects. Amplitude, pulse width, and frequency are adjusted to test the therapeutic window of stimulation (clinical improvement thresholds versus side effect thresholds). Some PD patients develop dyskinesia during test stimulation, which may be a positive indicator for lead location. If good effects and a therapeutic window are observed, the location of the lead is considered to be satisfactory and the procedure is completed.

Pulse generator implantation

During the final step of surgery, performed under general anesthesia, the pulse generator is implanted. The extension cable that connects the DBS lead to the implantable pulse generator is tunneled subcutaneously, connecting the DBS lead to the pulse generator in the chest.

Intraoperative, real-time MRI stereotaxis

Real-time intraoperative MRI has become available for DBS implantation with devices recently cleared for use by the FDA. The procedure, typically performed in a diagnostic MRI suite, uses MR images acquired during surgery to guide DBS lead implantation in the target area and to verify implantation accuracy.8

Implantation of a deep brain stimulator is the most common surgical procedure performed in the United States and industrialized world for the management of advanced movement disorders. These procedures are US Food and Drug Administration (FDA)–approved for the management of the symptoms of Parkinson disease (PD) and essential tremor. Deep brain stimulation (DBS) is also approved for managing primary generalized dystonia and torticollis under a humanitarian device exemption.

Deep brain stimulation has largely replaced ablative procedures such as thalamotomy and pallidotomy. While ablative procedures can be effective for the symptoms of movement disorders, they cause a permanent lesion in the targeted nuclei and are therefore not reversible. DBS is considered safer because it can be adjusted over time and the location of the leads can be revised.1 On the other hand, regular maintenance of implanted hardware may be considered a disadvantage of DBS.

HARDWARE AND TARGETS

While ablative procedures do not require implantable hardware, DBS consists of permanently implanted neurostimulation systems. The battery-powered pulse generators typically last for several years but require multiple replacements during a lifetime. In addition, if other hardware components fail, surgical revision may be required to maintain treatment efficacy. Surgery involving implantation of hardware carries a higher risk of infection than does a nonimplantation procedure. If infections occur, removal of the hardware is often required, with reimplantation performed after the infection clears. In addition, the expense of DBS hardware may limit availability in some cases.

Three components

Figure 1. The components of an implantable deep brain stimulation system.
Permanently implanted DBS devices have three components: the DBS lead, which is inserted into the brain and extends to the outside of the skull; the implantable pulse generator, typically located in the infraclavicular area; and an extension cable that connects the two components (Figure 1). Patients may have unilateral or bilateral lead implantation and unilateral or bilateral implantation of pulse generators. A single generator may be connected to both brain leads. Patients also have the option of receiving either a nonrechargeable or a rechargeable pulse generator. The advantage of the latter is longer intervals between battery replacement surgery (up to 9 years). However, these require more maintenance by the patient, who needs to periodically recharge the generators at home using a wireless charging unit. The recharging procedure may be time consuming and difficult for patients who are challenged by new technologies. In our experience, most patients with PD and tremor prefer nonrechargeable pulse generators.

Target nuclei

Several nodes or nuclei can serve as targets for DBS. In patients with PD, the most common surgical target is the subthalamic nucleus (STN), either unilaterally or bilaterally.2 The globus pallidus pars interna (GPi) is also a viable target and is preferred for some patients with PD. The most common target for managing essential tremor is the ventral intermediate nucleus (VIM) of the thalamus, which can also be the target of choice for patients with tremor-predominant PD. However, the GPi and STN are usually preferred over the VIM in patients with PD because stimulation of these targets can relieve symptoms other than tremor, such as rigidity and bradykinesia. Bilateral stimulation of the GPi is the most frequent approach in patients with generalized torsion dystonia and torticollis, although the STN and thalamic nuclei (off-label) are also considered options.

PATIENT SELECTION

Patients are evaluated in our center at Cleveland Clinic by a multidisciplinary team that includes a movement disorder neurologist, a subspecialized neurosurgeon, a movement disorder neuropsychologist, and a psychiatrist with special interest in the behavioral comorbidities of movement disorders.3 Neuroimaging is included in this assessment. We have also included physical therapy as part of the initial evaluation in order to gain insight into the patient’s limitations and develop rehabilitation strategies that may enhance the outcomes of surgery or provide alternatives should surgery not be indicated. This evaluation provides extensive data that are then reviewed by the team in a conference dedicated to discussing candidacy for DBS or options for managing the symptoms of advanced movement disorders. Behavioral and cognitive issues are assessed in detail and, in our experience, are the most common reasons for not recommending DBS.

An important part of the evaluation of patients with PD is a formal test with rating of the motor section of the Unified Parkinson’s Disease Rating Scale (UPDRS) with the patient off medications for 8 to 12 hours and then after a test dose of levodopa. At our center, this off/on test is videotaped so that the responsiveness of individual symptoms to levodopa can be reviewed later in conference.

Risk of cognitive decline

While DBS is considered safe and effective, there is a risk of cognitive decline in some patients. In most patients, long-term stimulation-related cognitive decline may be detected with formal measures but is not clinically significant and is outweighed by the motor and quality-of-life benefits of surgery. In some patients, long-term cognitive decline can be significant and can limit function. Cognitive neuropsychologic testing provides valuable information in this regard. Patients with preserved cognitive function seldom experience significant decline with DBS while those with substantial baseline impairment are thought to be at greater risk. Patients who meet criteria for dementia are usually not considered candidates for DBS, but exceptions exist. Transient perioperative cognitive difficulties are more common than persistent deficits, and typically resolve within a few weeks (see “Complications of deep brain stimulation”).

Benefits in Parkinson disease

Deep brain stimulation can address several symptoms of PD but with varying effects. Tremor, rigidity, and bradykinesia usually improve substantially. Gait has a more variable response, and balance is typically refractory. A general rule is that symptoms that improve with a single dose of levodopa should also improve with DBS. (Tremor, however, will most often respond to DBS even if refractory to medication.) Good candidates for surgery typically have a greater than 30% improvement in UPDRS motor score with levodopa challenge, but sometimes, improvement in the total score is less informative than evaluation of the effects of levodopa on particular symptoms. Treatment effects can be compared with the patient’s expectations for surgery in order to infer whether the goals for symptom improvement are realistic.

 

 

Treatment outcomes depend on etiology

After programming, DBS can provide PD symptom control similar to that of medication “on time,” but with fewer on-off fluctuations and less on-time dyskinesia. Good surgical candidates are patients who once responded well to dopaminergic medications but who, after several years with the disease, present with increased duration of “off time,” unpredictable duration of on time, and medication side effects such as on-time dyskinesia. Patients who do not respond well to levodopa even in subscores of the UPDRS may not be good candidates for DBS, and in some cases the diagnosis itself needs to be reviewed.

Deep brain stimulation can improve quality of life and alleviate symptoms of essential tremor. Tremor control is best for the upper extremities and tends to be better for distal tremors than for proximal ones. Patients who are good candidates for surgery often have severe tremors. A substantial improvement in these symptoms often has a dramatic, positive effect on work and quality of life. In some patients, surgery is considered for mild tremor if it seriously disrupts the patient’s lifestyle or occupation and cannot be well controlled with medications. Often, in these cases, tremor that appears relatively mild to the examiner is significantly limiting for the patient.

Very severe and proximal tremor is more refractory, though it may also improve. The changes can be well documented with objective measures. In these cases, however, residual tremor can still be moderate to severe and can be functionally limiting. Head or vocal tremors are typically refractory. They may be improved with bilateral implantation, but this cannot be accurately predicted. Patients who present with head-only or head-predominant tremor are thought to be less likely to benefit than those with limb tremor. Nonetheless, tremors of the head can severely impair quality of life. Because there are few other treatment options, some patients choose DBS with the understanding that the outcome is uncertain and the benefit may be limited.

Tremor resulting from multiple sclerosis or other causes can be medically refractory and disabling. In our experience, DBS can be an off-label option for managing secondary tremors and good outcomes have been observed. However, outcomes are much less predictable and tremor control less effective than in patients with essential tremor.

Patients with primary generalized dystonia can be considered candidates for DBS and may experience improved symptom control and quality of life.4 Patients with the DYT1 mutation are more likely to respond well to DBS, as are those with other forms of primary generalized dystonia. In contrast to that seen in patients with PD and tremor, symptomatic improvement is frequently not observed during intraoperative testing. Several months of stimulation and programming may be required before significant improvements are detected.5 Surgery can also be considered for off-label use in the treatment of patients with secondary dystonia—such as that following injury or associated with cerebral palsy—but outcomes are less predictable and usually more limited. A possible exception may be seen in cases of tardive dystonia, for which there is increasing evidence6 for the effectiveness of DBS. This remains an off-label use of DBS.

Realistic expectations

An important aspect of the multidisciplinary evaluation includes a discussion of the expectations for surgery, the risks, and the requirements for postoperative care. As discussed above, DBS is reversible and adjustable, so outcomes depend not only on accurate implantation of the hardware but also on postoperative programming. Also, monitoring and maintenance of the implanted hardware are required in these patients. It is important that patients and families appreciate the fact that specialized, long-term postoperative follow-up is as much a part of the treatment as is the implantation itself.

UNILATERAL VERSUS BILATERAL DBS

Most patients with generalized dystonia undergo bilateral DBS. However, patients with PD or essential tremor may receive bilateral, staged, or unilateral implants. Some patients with PD present with either near-complete predominance of symptoms on one side or with symptoms that affect mostly the dominant extremity. In these patients, unilateral implantation is often recommended because it has less risk than the bilateral approach and may be sufficient to address the most limiting symptoms.

As the disease advances, an additional surgery may be required to accomplish bilateral symptom control. Nevertheless, we do not routinely recommend preventive implantation because it is not known whether second-side symptoms will become severe enough to require it. This strategy allows for deferring surgical risk, which is in itself advantageous. In our experience, bilateral implantation is often recommended to PD patients who present with symptoms such as freezing of gait.

Patients who have essential tremor often present with bilateral symptoms. Although many patients will indicate that they need symptom relief on both upper extremities in order to perform activities of daily living, our practice is to recommend surgery on one side at first and to suggest the patient consider contralateral implantation after weeks or months. Bilateral implantation may carry a risk for dysarthria and the risk is thought to be reduced if bilateral procedures are staged. Although high rates of dysarthria have been reported following bilateral surgery for tremor, its occurrence has been infrequent in our experience with bilateral staged DBS. Benefits of treating tremor in the dominant extremity usually exceed those of treating nondominant tremor, so most patients prefer that the dominant side be the first one treated.

TECHNICAL OPTIONS

There are several technical options for implantation of DBS systems. Stereotactic procedures rely on co-registration of preoperative imaging with external and internal fiducials, or points of reference. Targeting of the intended structures is performed by combining direct and indirect methods. Direct methods rely on identification of the target structures with imaging, such as visualization of the STN and GPi on preoperative magnetic resonance imaging (MRI). Indirect targeting relies on cadaveric anatomic atlases and coordinate systems that infer the location of the intended structures in relation to anatomical points of reference.

Frame-based systems

Figure 2. In a frame-based system, the stereotactic arc is attached to the base of the frame. Entry points of the leads are marked on the skin and on the skull.
In the most common approach to DBS surgery,7 stereotactic frames are placed over the patient’s head and secured with pins. The frame becomes the fixed point of reference for accurate stereotactic surgery and must remain in place for the duration of the procedure. Computed tomography or MRI is then performed with the frame in place, so that the images are co-registered with the fiducial points of the stereotactic frame. The targets are then selected for surgery and trajectories are chosen based on anatomic structures. The patient is positioned supine and the frame and head are secured to the operating table. The coordinates calculated by the clinical workstations are then set to the stereotactic frame and arc. The stereotactic arc (Figure 2) is attached to the base of the frame and the entry points of the leads—where the burr hole will be placed—are marked on the skin and then on the skull. Once the burr hole and opening of the meninges are completed, the targeting cannulae are inserted. The microelectrode system is then mounted for recording of the target area and subsequently for final lead implantation.

Frameless systems

Figure 3. Surgeon’s view of the frameless device, placed over the head approximately at the level of the coronal suture. The occipital area is in the bottom of the figure. When a frameless system is used, a lighter-weight structure is affixed to the head.
The workflow and overall surgical procedure for implantation of DBS with frameless systems are similar to those of the frame-based procedure. However, instead of fixing the head to a rigid frame that prevents head motion, a lighter-weight, frameless system is fixed to the head and moves with it (Figure 3). First, metal screws and fiducials are fixed to the head under local anesthesia or sedation. Preoperative imaging is then acquired with the fiducials in place and the surgical plans are completed in the same fashion as for frame-based surgery. The patient is then placed supine on the operating table and the frameless system is attached to the head with the aid of image guidance, in the location determined by target and trajectory planning.

The key advantage of the frameless system over the frame-based system is greater mobility of the head. Another important advantage is easier access to the airway, should an emergency situation occur. In our practice, patients with experience of both frameless and frame-based systems did not report significantly less discomfort with the frameless system.

The frameless system also has disadvantages, including less secure fixation of the head, which can add risk to the procedure. In addition, because of its lightweight, plastic construction, it provides less robust support to the instrumentation entering the brain than do metallic head frames and, in some cases, there is less flexibility for adjusting targets if needed during surgery. In addition, frameless systems are nonreusable and represent a substantial additional cost.

 

 

Microelectrode recording

Physiologic verification of anatomic targets identified by imaging can be accomplished with microelectrode recording (MER). This technique involves placing fine, high-impedance electrodes through the target area, so that anatomic structures can be recognized by characteristic electrical activity of individual neurons or groups of neurons. The locations of the structures are identified and the lengths of the electrode trajectories through the different structures—as well as the gaps between these structures—are recorded. The distances are then compared with the anatomy and a best-fit model is created to infer the location of the trajectory in the target area. Additional MER penetrations are made in order to further delineate the anatomy. Once a location for implantation has been selected, the DBS lead is inserted into the target area.

Electrode implantation

Lead implantation is often performed under fluoroscopic guidance in order to ensure accuracy and stability. When implanted, the electrode may cause a microlesional effect, manifested by transient improvement in symptoms.

The DBS leads are then connected to external pulse generators and assessed for clinical benefits and side effects. Amplitude, pulse width, and frequency are adjusted to test the therapeutic window of stimulation (clinical improvement thresholds versus side effect thresholds). Some PD patients develop dyskinesia during test stimulation, which may be a positive indicator for lead location. If good effects and a therapeutic window are observed, the location of the lead is considered to be satisfactory and the procedure is completed.

Pulse generator implantation

During the final step of surgery, performed under general anesthesia, the pulse generator is implanted. The extension cable that connects the DBS lead to the implantable pulse generator is tunneled subcutaneously, connecting the DBS lead to the pulse generator in the chest.

Intraoperative, real-time MRI stereotaxis

Real-time intraoperative MRI has become available for DBS implantation with devices recently cleared for use by the FDA. The procedure, typically performed in a diagnostic MRI suite, uses MR images acquired during surgery to guide DBS lead implantation in the target area and to verify implantation accuracy.8

References
  1. Rezai AR, Machado AG, Azmi H, Kubu C, Boulis NM. Surgery for movement disorders. Neurosurg 2008; 62(SHC suppl 2):SHC809SHC839.
  2. Krack P, Batir A, Van Blercom N, et al. Five-year follow-up of bilateral stimulation of the subthalamic nucleus in advanced Parkinson’s disease. N Engl J Med 2003; 349:19251934.
  3. Machado A, Fernandez HH, Deogaonkar M. Deep brain stimulation: what can patients expect from it? Cleve Clin J Med 2012; 79:113120.
  4. Vidailhet M, Vercueil L, Houeto JL, et al. Bilateral deep-brain stimulation of the globus pallidus in primary generalized dystonia. N Engl J Med 2005; 352:459467.
  5. Kupsch A, Benecke R, Müller J, et al. Pallidal deep-brain stimulation in primary generalized or segmental dystonia. N Engl J Med 2006; 355:19781990.
  6. Gruber D, Trottenberg T, Kivi A, et al. Long-term effects of pallidal deep brain stimulation in tardive dystonia. Neurology 2009; 73:5358.
  7. Gross RE, Sharan AD, Benabid AL. Deep brain stimulation for Parkinson’s disease: surgical technique and perioperative management. Mov Disord 2006; 21( suppl 14):S247S258.
  8. Starr PA, Martin AJ, Ostrem JL, et al. Subthalamic nucleus deep brain stimulator placement using high-field interventional magnetic resonance imaging and a skull-mounted aiming device: technique and application accuracy. J Neurosurg 2010; 112:479490.
References
  1. Rezai AR, Machado AG, Azmi H, Kubu C, Boulis NM. Surgery for movement disorders. Neurosurg 2008; 62(SHC suppl 2):SHC809SHC839.
  2. Krack P, Batir A, Van Blercom N, et al. Five-year follow-up of bilateral stimulation of the subthalamic nucleus in advanced Parkinson’s disease. N Engl J Med 2003; 349:19251934.
  3. Machado A, Fernandez HH, Deogaonkar M. Deep brain stimulation: what can patients expect from it? Cleve Clin J Med 2012; 79:113120.
  4. Vidailhet M, Vercueil L, Houeto JL, et al. Bilateral deep-brain stimulation of the globus pallidus in primary generalized dystonia. N Engl J Med 2005; 352:459467.
  5. Kupsch A, Benecke R, Müller J, et al. Pallidal deep-brain stimulation in primary generalized or segmental dystonia. N Engl J Med 2006; 355:19781990.
  6. Gruber D, Trottenberg T, Kivi A, et al. Long-term effects of pallidal deep brain stimulation in tardive dystonia. Neurology 2009; 73:5358.
  7. Gross RE, Sharan AD, Benabid AL. Deep brain stimulation for Parkinson’s disease: surgical technique and perioperative management. Mov Disord 2006; 21( suppl 14):S247S258.
  8. Starr PA, Martin AJ, Ostrem JL, et al. Subthalamic nucleus deep brain stimulator placement using high-field interventional magnetic resonance imaging and a skull-mounted aiming device: technique and application accuracy. J Neurosurg 2010; 112:479490.
Page Number
S19-S24
Page Number
S19-S24
Publications
Publications
Article Type
Display Headline
Deep brain stimulation for movement disorders: Patient selection and technical options
Display Headline
Deep brain stimulation for movement disorders: Patient selection and technical options
Citation Override
Cleveland Clinic Journal of Medicine 2012 July;79(suppl 2):S19-S24
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media

Use of chemodenervation in dystonic conditions

Article Type
Changed
Mon, 10/01/2018 - 15:35
Display Headline
Use of chemodenervation in dystonic conditions

Dystonia is a movement disorder in which involuntary sustained muscle contractions cause twisting movements that place the body in abnormal, sometimes painful, positions. Dystonia is believed to arise from an abnormality in the basal ganglia and an inherent or acquired defect in the processing of neurotransmitters.1

Dystonia is uncommon, although its exact prevalence is unknown. Nutt et al concluded that at least 250,000 people were affected by idiopathic dystonia in the United States, but prevalence is likely higher because misdiagnosis is not uncommon.2 A more recent European study found the prevalence of primary dystonia in the general population aged 50 years or more to be 732 per 100,000.3 The Epidemiological Study of Dystonia in Europe (ESDE) Collaborative Group found that the estimated prevalence of cervical dystonia was 50 to 200 per 1 million individuals.4 Also known as spasmodic torticollis, this is the most commonly diagnosed form of focal dystonia.

CLASSIFICATION OF DYSTONIA

Accurate classification of dystonia is important, since this informs approaches to management as well as prognosis. The three most important means by which dystonia is classified are (1) etiology, including primary dystonia, which encompasses a variety of genetic variables, and secondary dystonia; (2) bodily distribution of symptoms; and (3) age at onset.

Etiology

Most primary or idiopathic dystonia appears to be hereditary. Early-onset primary dystonia is most frequently caused by a mutation in the DYT1 gene, although other genetic mutations are possible.5 Patients with primary dystonia have no other underlying disorder; involuntary muscle contractions are the sole symptom. A thorough history should include a review of perinatal and early developmental history, prior neurologic illness, and exposure to drugs known to cause acquired dystonia. Physical examinations (encompassing intellectual, pyramidal, cerebellar, and sensory domains) and laboratory tests reveal no specific cause for the dystonic symptoms. Primary dystonia is also most frequently action-induced; at rest, the affected body region may appear to be normal.

Secondary dystonia occurs as a symptom of another disease process. Multiple sclerosis or any one of several hereditary neurologic disorders, such as Wilson disease, may be implicated. Secondary dystonia also may result from trauma to the brain, as might occur during an automobile accident; from heavy-metal or carbon monoxide poisoning; or as an adverse effect of medication. It may be psychogenic or related to Parkinson disease or Parkinson-plus syndromes, a group of neurodegenerative disorders with parkinsonian features. Tardive dystonia, the most common adult form of secondary dystonia, may occur follow ing exposure to certain neuroleptic drugs; tardive dystonia is a type of tardive dyskinesia that describes any involuntary neurologic movement disorder.

Bodily distribution

Dystonia is further classified by location of symptoms. Focal dystonias, which are usually primary dystonias, describe symptoms that are limited to a region of the body, such as a specific arm. There are several variations. Cervical dystonia affects the head and neck, is the most common adult-onset dystonia, and affects more women than men. Blepharospasm, or involuntary contractions of the eyelids, potentially leads to extended eye closure and functional blindness and often involves other facial muscles. Laryngeal dystonia affects the muscles in the larynx. Limb dystonia, such as writer’s or musician’s cramp, affects muscles in the arm, hand, leg, or foot. Limb dystonia is often task-specific action dystonia, and can be primary or secondary.

Segmental dystonia describes a group of involved muscles that are contiguous, such as cranial to neck to cervical to arm. Oromandibular dystonia, affecting the face, mouth, and jaw, often with unusual tongue movements (ie, lingual dystonia), is a type of segmental dystonia, although some consider it a focal dystonia. Meige syndrome is the combination of blepharospasm and oromandibular dystonia. Certain limb and cranial dystonias are considered segmental dystonias. Dystonia that affects two or more noncontiguous muscle groups in different parts of the body is multifocal. Hemi dystonia describes unilateral symptoms.

Symptoms that have advanced from a focal presentation to affect additional regions of the body characterize generalized dystonia. The symptoms potentially advance to include the trunk and limbs. The muscular contractions are usually sustained, are often both repetitive and painful, and worsen with activity.6 In severe cases, muscular contractions may occur even while resting. Early-onset myoclonus dystonia is a generalized hereditary dystonia whose symptoms include dystonic contractions of the neck and shoulders and rapid jerking movements.7 Of note diagnostically, early-onset dystonia in a leg typically begins at age 8 to 9 years and is more likely than other early-onset presentations to progress to generalized dystonia. Early-onset dystonia that begins in an arm typically presents later, at age 12 to 14 years, and is less likely to progress to generalized dystonia. Late-onset dystonia (> 27 years of age), by contrast, rarely begins in a leg and tends to remain either focal or segmental.8

Age of onset

A third useful classification scheme identifies early-onset (childhood to young adult) and late-onset varieties of dystonia.

THE DIAGNOSTIC CHALLENGE

Accurate diagnosis of dystonia is challenging because of its relative rarity and the variety of etiologies that pertain to this heterogeneous family of disorders. Patterns of inheritance are not straightforward and primary dystonia can be difficult to diagnose even with the benefit of genetic testing. There is no identifiable pathologic abnormality in many patients, and negative genetic tests do not necessarily mean that the dystonia is not primary. In the face of these challenges it is not surprising that dystonia is frequently misdiagnosed (Table 1). Nevertheless, certain findings can guide the diagnosis toward primary or secondary dystonia.

Consider primary dystonia if perinatal and developmental histories, intellect, strength, and perception of sensations are normal. There should be no prior history of neurologic illness or exposure to neuroleptic drugs whose adverse effects include secondary dystonia. In primary dystonia, diagnostic studies are negative and dystonia is the only symptom. If onset of symptoms is associated with activity, then primary dystonia should be considered. In the case of early- or late-onset limb dystonia, testing should be performed for the DYT1 gene. If the results are negative, then a trial for dopa-responsive dystonia should be undertaken with levodopa.

Consider secondary dystonia if the patient has been exposed to neuroleptic drugs, symptoms are distributed unilaterally, or the presentation is unusual for age or distribution of symptoms. For example, cranial dystonia in a child would raise the index of suspicion for secondary dystonia. If tardive dystonia is part of the differential diagnosis, consider magnetic resonance imaging (MRI), serum ceruloplasmin measurement, or slit-lamp diagnostic testing. Suspicion of a structural lesion affecting the central nervous system warrants examination with MRI, computed tomography, or angiography. Certain metabolic and neurologic hereditary disorders cause secondary dystonia, in which case dopa-responsive dystonia should be ruled out. Psychometric testing should also be considered.

 

 

SYMPTOMATIC TREATMENT WITH CHEMODENERVATION

In the absence of a cure, treatment options for dystonia are necessarily symptomatic and supportive. Titratable chemo denervation agents are injected directly into the muscle or motor nerve, temporarily weakening the local muscle and easing dystonia symptoms. Chemo denervation agents include phenol, ethyl alcohol, and botulinum toxin types A (BTX-A; onabotulinumtoxinA, abo botulinumtoxinA, and incobotulinumtoxinA) and B (BTX-B; rimabotulinum toxinB).

Phenol and ethyl alcohol injections targeted perineurally or as a motor point block have been employed for dystonia and cause nonselective tissue destruction, muscle necrosis, and highly variable durations of response. Perineural microcirculation may be damaged, possibly leading to long-term defects.

Clostridium botulinum bacteria produce seven serologically distinct neuroparalytic toxins. They are the most powerful such toxins currently known and temporarily prevent acetylcholine vesicles from docking into the presynaptic neuromuscular junction. Use of BTX-A for treatment of dystonia was recommended in a National Institutes of Health consensus statement in 1990.9 It has been studied for a variety of dystonias, including blepharospasm, hemifacial spasm, laryngeal dystonia, oromandibular dystonia, and cervical dystonia, among other focal dystonias. Lew et al reported in 1997 on the successful use of BTX-B for cervical dystonia in a double-blind, single-treatment study,10 and confirmatory studies followed.11,12

Varying indications for botulinum toxin

US Food and Drug Administration–approved indications for the toxins vary. The three BTX-A products and the single BTX-B product are approved for the treatment of cervical dystonia in adults to reduce the severity of abnormal head position and neck pain. OnabotulinumtoxinA is approved for treatment of blepharospasm and strabismus associated with dystonia; and incobotulinumtoxinA is approved for blepharospasm in patients who have previously been treated with onabotulinumtoxinA. BTX-A has also been found to be safe and effective for the management of focal dystonias. These botulinum toxin agents are not equivalent in dosing units, so caution must be observed when switching brands.

Patients selected to receive BTX for dystonia should meet three criteria:

  • The dystonia should interfere with their functioning, comfort, or care to the degree that causes impairment and affects activities of daily living;
  • Focal weakening following administration of the drug should not decrease their level of function; and
  • The patient should understand that use of BTX may not completely address positioning, posturing, or secondary deformities.

Contraindications include pregnancy, lactation, comorbid neuromuscular disease (eg, amyotrophic lateral sclerosis or myasthenia gravis), and use of an aminoglycoside.

The need for BTX therapy should be reevaluated prior to each treatment; clinical benefit lasts 3 months or more. Electromyography may facilitate the location of target muscles, particularly since involved musculature may not be palpable and is often not superficial.13 In-office tools that help document baseline and posttreatment results, including videotaping dystonic limb movements and the use of rating scales, can be important for evaluating the patient’s progress.14

Relief for cervical dystonia

The treatment of choice for focal dystonias and focal aspects of generalized dystonia is BTX. Both BTX-A and BTX-B offer effective palliative treatments for cervical dystonia by improving neck position, reducing pain, and decreasing disability in sufferers.11,15–18 The BTX solution is injected directly into the dystonic muscle at several locations, temporarily weakening the overactive muscle. The BTX dose is approximately proportional to the size of the muscle, although smaller muscles typically responsible for precision movement may require a relatively larger dose (Table 2). Doses may be modified according to clinical factors such as muscle bulk and severity of dystonia (Table 3).

Relief following BTX injection for cervical dystonia occurs about 1 week later, with the greatest effect seen at about 2 to 6 weeks following injection; relief may last 12 to 16 weeks. Reinjections are not normally administered prior to 12 weeks’ duration in order to reduce the possibility of antibody formation. Concomitant interventions addressing depression and anxiety may have a significant effect on overall quality of life.19 Patients may also try several sensory tricks, called gestes antagoniste, which may temporarily reduce or alleviate the dystonia. However, these tactile procedures—such as placing a hand on top of the head—lose their effectiveness over time.

Treatment of blepharospasm, focal limb dystonia

The use of BTX-A for blepharospasm is a significant improvement over the former clinical reliance on various oral medications, which, with the exception of baclofen, proved largely ineffective.20 Surgical treatments result in damage to muscular and nervous tissues, and so are reserved only for nonresponders to BTX-A therapy.21

BTX-A can provide effective relief and is the treatment of choice for focal limb dystonias.22 Goals of treatment include functional improvement, correction of abnormal posture, and relief from discomfort. Although a variety of oral medications may also be prescribed, drug toxicity and adverse effects can outweigh the benefit and are usually only used in cases of severe dystonia. Oral medications used for limb dystonia include anticholinergics, dopamine agonists and antagonists, baclofen, clonazepam or other benzodiazepines, and muscle relaxants.

Antibodies may bind to the drug in a small percentage of patients who regularly receive injections of BTX, rendering additional injections of that specific serotype of BTX ineffective. This immunoresistance can be avoided if clinicians inject only the smallest quantity of BTX that achieves clinical efficacy, avoid administering booster injections before the end of the minimum 12-week lockout period, and extend the period between treatments as long as possible. If immunoresistance does occur, the BTX should be exchanged for a different serotype.

Testing for nonresponse

Patients are said to be nonresponders to BTX therapy if at 4 to 6 weeks following injection they show no reduction in muscle tone. A functional test for nonresponse is to inject a small amount of BTX into either the frontalis or sternocleidomastoid muscle prior to starting treatment; asymmetric weakness demonstrates a response, indicating that either injection technique or muscle selection is the problem. In addition to the development of neutralizing antibodies, other possible reasons for nonresponse include a dose that is too low or an alteration in the pattern of muscles involved in the dystonic movement.

References
  1. Dystonia fact sheet. National Institute of Neurological Disorders and Stroke Web Site. http://www.ninds.nih.gov/disorders/dystonias/detail_dystonias.htm. Updated October 27, 2011. Accessed April 17, 2012.
  2. Nutt JG, Muenter MD, Aronson A, Kurland LT, Melton LJ. Epidemiology of focal and generalized dystonia in Rochester, Minnesota. Mov Disord 1988; 3:188194.
  3. Muller J, Kiechl S, Wenning GK, et al. The prevalence of primary dystonia in the general community. Neurology 2002; 59:941943.
  4. Epidemiological Study of Dystonia in Europe Collaborative Group. A prevalence study of primary dystonia in eight European countries. J Neurol 2000; 24:787793.
  5. Klein C, Kann M, Kis B, et al Genetics of dystonia. Nervenarz 2000; 71:431441.
  6. Fahn S, Marsden CD, Calne DB. Classification and investigation of dystonia. In:Marsden CD, Fahn S, eds. Movement Disorders 2. London, UK: Butterworth; 1987:332358.
  7. Doheny D, Danisi F, Smith C, et al. Clinical findings of a myoclonus-dystonia family with two distinct mutations. Neurology 2002; 59:11301131.
  8. Greene P, Kang UJ, Fahn S. Spread of symptoms in idiopathic torsion dystonia. Mov Disord 1995; 10:143152.
  9. Clinical use of botulinum toxin. NIH Consens Statement 1990; 8:120.
  10. Lew MF, Adornato BT, Duane DD, et al. Botulinum toxin type B: a double-blind, placebo-controlled, safety and efficacy study in cervical dystonia. Neurology 1997; 49:701707.
  11. Brin MF, Lew MF, Adler CH, et al. Safety and efficacy of Neuro-Bloc (botulinum toxin type B) in type A-resistant cervical dystonia. Neurology 1999; 53:14311438.
  12. Brashear A, Lew MF, Dykstra DD, et al. Safety and efficacy of NeuroBloc (botulinum toxin type B) in type A-responsive cervical dystonia. Neurology 1999; 53:14391446.
  13. Dressler D. Electromyographic evaluation of cervical dystonia for planning of botulinum toxin therapy. Eur J Neurol 2000; 7:713718.
  14. Dystonia rating scales and scoring sheets. Movement Disorders Virtual University Web site. http://www.mdvu.org/library/ratingscales/dystonia/. Updated April 21, 2008. Accessed April 18, 2012.
  15. Brashear A. The botulinum toxins in the treatment of cervical dystonia. Semin Neurol 2001; 21:8590.
  16. Brashear A, Watts MW, Marchetti A, Magar R, Lau H, Wang L. Duration of effect of botulinum toxin type A in adult patients with cervical dystonia: a retrospective chart review. Clin Ther 2000; 22:15161524.
  17. Ceballos-Baumann AO. Evidence-based medicine in botulinum toxin therapy for cervical dystonia. J Neurol 2001; 248( suppl 1):1420.
  18. Giladi N, Meer J, Kidan H, Honigman S. Long-term remission of idiopathic cervical dystonia after treatment with botulinum toxin. Eur Neurol 2000; 44:144146.
  19. Ben-Shlomo Y, Camfield L, Warner T, ESDE Collaborative Group. What are the determinants of quality of life in people with cervical dystonia? J Neurol Neurosurg Psychiatry 2002; 72:608614.
  20. Fahn S, Hening WA, Bressman S, et al. Long-term usefulness of baclofen in the treatment of essential blepharospasm. Adv Ophthal Plastic Reconstr Surg 1985; 4:219226.
  21. Callahan A. Blepharospasm with resection of part of orbicularis nerve supply. Arch Ophthalmol 1963; 70:508511.
  22. Yoshimura DM, Aminoff MJ, Olney RK. Botulinum toxin therapy for limb dystonias. Neurology 1992; 42:627630.
Article PDF
Author and Disclosure Information

Maurice Hanson, MD
Neurologist, Movement Disorders Program, Cleveland Clinic Florida, West Palm Beach, FL

Correspondence: Maurice Hanson, MD, Cleveland Clinic Florida, 525 Okeechobee Boulevard, City Place Tower, West Palm Beach, FL 33401; hansonm@ccf.org

Dr. Hanson reported that he has no financial interests or relationships that pose a potential conflict of interest with this article.

This article is based on Dr. Hanson’s presentation at “The Annual Therapy Symposium on Movement Disorders for the Modern Clinician” held in Fort Lauderdale, Florida, on January 29, 2011. The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Hanson.

Publications
Page Number
S25-S29
Author and Disclosure Information

Maurice Hanson, MD
Neurologist, Movement Disorders Program, Cleveland Clinic Florida, West Palm Beach, FL

Correspondence: Maurice Hanson, MD, Cleveland Clinic Florida, 525 Okeechobee Boulevard, City Place Tower, West Palm Beach, FL 33401; hansonm@ccf.org

Dr. Hanson reported that he has no financial interests or relationships that pose a potential conflict of interest with this article.

This article is based on Dr. Hanson’s presentation at “The Annual Therapy Symposium on Movement Disorders for the Modern Clinician” held in Fort Lauderdale, Florida, on January 29, 2011. The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Hanson.

Author and Disclosure Information

Maurice Hanson, MD
Neurologist, Movement Disorders Program, Cleveland Clinic Florida, West Palm Beach, FL

Correspondence: Maurice Hanson, MD, Cleveland Clinic Florida, 525 Okeechobee Boulevard, City Place Tower, West Palm Beach, FL 33401; hansonm@ccf.org

Dr. Hanson reported that he has no financial interests or relationships that pose a potential conflict of interest with this article.

This article is based on Dr. Hanson’s presentation at “The Annual Therapy Symposium on Movement Disorders for the Modern Clinician” held in Fort Lauderdale, Florida, on January 29, 2011. The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Hanson.

Article PDF
Article PDF

Dystonia is a movement disorder in which involuntary sustained muscle contractions cause twisting movements that place the body in abnormal, sometimes painful, positions. Dystonia is believed to arise from an abnormality in the basal ganglia and an inherent or acquired defect in the processing of neurotransmitters.1

Dystonia is uncommon, although its exact prevalence is unknown. Nutt et al concluded that at least 250,000 people were affected by idiopathic dystonia in the United States, but prevalence is likely higher because misdiagnosis is not uncommon.2 A more recent European study found the prevalence of primary dystonia in the general population aged 50 years or more to be 732 per 100,000.3 The Epidemiological Study of Dystonia in Europe (ESDE) Collaborative Group found that the estimated prevalence of cervical dystonia was 50 to 200 per 1 million individuals.4 Also known as spasmodic torticollis, this is the most commonly diagnosed form of focal dystonia.

CLASSIFICATION OF DYSTONIA

Accurate classification of dystonia is important, since this informs approaches to management as well as prognosis. The three most important means by which dystonia is classified are (1) etiology, including primary dystonia, which encompasses a variety of genetic variables, and secondary dystonia; (2) bodily distribution of symptoms; and (3) age at onset.

Etiology

Most primary or idiopathic dystonia appears to be hereditary. Early-onset primary dystonia is most frequently caused by a mutation in the DYT1 gene, although other genetic mutations are possible.5 Patients with primary dystonia have no other underlying disorder; involuntary muscle contractions are the sole symptom. A thorough history should include a review of perinatal and early developmental history, prior neurologic illness, and exposure to drugs known to cause acquired dystonia. Physical examinations (encompassing intellectual, pyramidal, cerebellar, and sensory domains) and laboratory tests reveal no specific cause for the dystonic symptoms. Primary dystonia is also most frequently action-induced; at rest, the affected body region may appear to be normal.

Secondary dystonia occurs as a symptom of another disease process. Multiple sclerosis or any one of several hereditary neurologic disorders, such as Wilson disease, may be implicated. Secondary dystonia also may result from trauma to the brain, as might occur during an automobile accident; from heavy-metal or carbon monoxide poisoning; or as an adverse effect of medication. It may be psychogenic or related to Parkinson disease or Parkinson-plus syndromes, a group of neurodegenerative disorders with parkinsonian features. Tardive dystonia, the most common adult form of secondary dystonia, may occur follow ing exposure to certain neuroleptic drugs; tardive dystonia is a type of tardive dyskinesia that describes any involuntary neurologic movement disorder.

Bodily distribution

Dystonia is further classified by location of symptoms. Focal dystonias, which are usually primary dystonias, describe symptoms that are limited to a region of the body, such as a specific arm. There are several variations. Cervical dystonia affects the head and neck, is the most common adult-onset dystonia, and affects more women than men. Blepharospasm, or involuntary contractions of the eyelids, potentially leads to extended eye closure and functional blindness and often involves other facial muscles. Laryngeal dystonia affects the muscles in the larynx. Limb dystonia, such as writer’s or musician’s cramp, affects muscles in the arm, hand, leg, or foot. Limb dystonia is often task-specific action dystonia, and can be primary or secondary.

Segmental dystonia describes a group of involved muscles that are contiguous, such as cranial to neck to cervical to arm. Oromandibular dystonia, affecting the face, mouth, and jaw, often with unusual tongue movements (ie, lingual dystonia), is a type of segmental dystonia, although some consider it a focal dystonia. Meige syndrome is the combination of blepharospasm and oromandibular dystonia. Certain limb and cranial dystonias are considered segmental dystonias. Dystonia that affects two or more noncontiguous muscle groups in different parts of the body is multifocal. Hemi dystonia describes unilateral symptoms.

Symptoms that have advanced from a focal presentation to affect additional regions of the body characterize generalized dystonia. The symptoms potentially advance to include the trunk and limbs. The muscular contractions are usually sustained, are often both repetitive and painful, and worsen with activity.6 In severe cases, muscular contractions may occur even while resting. Early-onset myoclonus dystonia is a generalized hereditary dystonia whose symptoms include dystonic contractions of the neck and shoulders and rapid jerking movements.7 Of note diagnostically, early-onset dystonia in a leg typically begins at age 8 to 9 years and is more likely than other early-onset presentations to progress to generalized dystonia. Early-onset dystonia that begins in an arm typically presents later, at age 12 to 14 years, and is less likely to progress to generalized dystonia. Late-onset dystonia (> 27 years of age), by contrast, rarely begins in a leg and tends to remain either focal or segmental.8

Age of onset

A third useful classification scheme identifies early-onset (childhood to young adult) and late-onset varieties of dystonia.

THE DIAGNOSTIC CHALLENGE

Accurate diagnosis of dystonia is challenging because of its relative rarity and the variety of etiologies that pertain to this heterogeneous family of disorders. Patterns of inheritance are not straightforward and primary dystonia can be difficult to diagnose even with the benefit of genetic testing. There is no identifiable pathologic abnormality in many patients, and negative genetic tests do not necessarily mean that the dystonia is not primary. In the face of these challenges it is not surprising that dystonia is frequently misdiagnosed (Table 1). Nevertheless, certain findings can guide the diagnosis toward primary or secondary dystonia.

Consider primary dystonia if perinatal and developmental histories, intellect, strength, and perception of sensations are normal. There should be no prior history of neurologic illness or exposure to neuroleptic drugs whose adverse effects include secondary dystonia. In primary dystonia, diagnostic studies are negative and dystonia is the only symptom. If onset of symptoms is associated with activity, then primary dystonia should be considered. In the case of early- or late-onset limb dystonia, testing should be performed for the DYT1 gene. If the results are negative, then a trial for dopa-responsive dystonia should be undertaken with levodopa.

Consider secondary dystonia if the patient has been exposed to neuroleptic drugs, symptoms are distributed unilaterally, or the presentation is unusual for age or distribution of symptoms. For example, cranial dystonia in a child would raise the index of suspicion for secondary dystonia. If tardive dystonia is part of the differential diagnosis, consider magnetic resonance imaging (MRI), serum ceruloplasmin measurement, or slit-lamp diagnostic testing. Suspicion of a structural lesion affecting the central nervous system warrants examination with MRI, computed tomography, or angiography. Certain metabolic and neurologic hereditary disorders cause secondary dystonia, in which case dopa-responsive dystonia should be ruled out. Psychometric testing should also be considered.

 

 

SYMPTOMATIC TREATMENT WITH CHEMODENERVATION

In the absence of a cure, treatment options for dystonia are necessarily symptomatic and supportive. Titratable chemo denervation agents are injected directly into the muscle or motor nerve, temporarily weakening the local muscle and easing dystonia symptoms. Chemo denervation agents include phenol, ethyl alcohol, and botulinum toxin types A (BTX-A; onabotulinumtoxinA, abo botulinumtoxinA, and incobotulinumtoxinA) and B (BTX-B; rimabotulinum toxinB).

Phenol and ethyl alcohol injections targeted perineurally or as a motor point block have been employed for dystonia and cause nonselective tissue destruction, muscle necrosis, and highly variable durations of response. Perineural microcirculation may be damaged, possibly leading to long-term defects.

Clostridium botulinum bacteria produce seven serologically distinct neuroparalytic toxins. They are the most powerful such toxins currently known and temporarily prevent acetylcholine vesicles from docking into the presynaptic neuromuscular junction. Use of BTX-A for treatment of dystonia was recommended in a National Institutes of Health consensus statement in 1990.9 It has been studied for a variety of dystonias, including blepharospasm, hemifacial spasm, laryngeal dystonia, oromandibular dystonia, and cervical dystonia, among other focal dystonias. Lew et al reported in 1997 on the successful use of BTX-B for cervical dystonia in a double-blind, single-treatment study,10 and confirmatory studies followed.11,12

Varying indications for botulinum toxin

US Food and Drug Administration–approved indications for the toxins vary. The three BTX-A products and the single BTX-B product are approved for the treatment of cervical dystonia in adults to reduce the severity of abnormal head position and neck pain. OnabotulinumtoxinA is approved for treatment of blepharospasm and strabismus associated with dystonia; and incobotulinumtoxinA is approved for blepharospasm in patients who have previously been treated with onabotulinumtoxinA. BTX-A has also been found to be safe and effective for the management of focal dystonias. These botulinum toxin agents are not equivalent in dosing units, so caution must be observed when switching brands.

Patients selected to receive BTX for dystonia should meet three criteria:

  • The dystonia should interfere with their functioning, comfort, or care to the degree that causes impairment and affects activities of daily living;
  • Focal weakening following administration of the drug should not decrease their level of function; and
  • The patient should understand that use of BTX may not completely address positioning, posturing, or secondary deformities.

Contraindications include pregnancy, lactation, comorbid neuromuscular disease (eg, amyotrophic lateral sclerosis or myasthenia gravis), and use of an aminoglycoside.

The need for BTX therapy should be reevaluated prior to each treatment; clinical benefit lasts 3 months or more. Electromyography may facilitate the location of target muscles, particularly since involved musculature may not be palpable and is often not superficial.13 In-office tools that help document baseline and posttreatment results, including videotaping dystonic limb movements and the use of rating scales, can be important for evaluating the patient’s progress.14

Relief for cervical dystonia

The treatment of choice for focal dystonias and focal aspects of generalized dystonia is BTX. Both BTX-A and BTX-B offer effective palliative treatments for cervical dystonia by improving neck position, reducing pain, and decreasing disability in sufferers.11,15–18 The BTX solution is injected directly into the dystonic muscle at several locations, temporarily weakening the overactive muscle. The BTX dose is approximately proportional to the size of the muscle, although smaller muscles typically responsible for precision movement may require a relatively larger dose (Table 2). Doses may be modified according to clinical factors such as muscle bulk and severity of dystonia (Table 3).

Relief following BTX injection for cervical dystonia occurs about 1 week later, with the greatest effect seen at about 2 to 6 weeks following injection; relief may last 12 to 16 weeks. Reinjections are not normally administered prior to 12 weeks’ duration in order to reduce the possibility of antibody formation. Concomitant interventions addressing depression and anxiety may have a significant effect on overall quality of life.19 Patients may also try several sensory tricks, called gestes antagoniste, which may temporarily reduce or alleviate the dystonia. However, these tactile procedures—such as placing a hand on top of the head—lose their effectiveness over time.

Treatment of blepharospasm, focal limb dystonia

The use of BTX-A for blepharospasm is a significant improvement over the former clinical reliance on various oral medications, which, with the exception of baclofen, proved largely ineffective.20 Surgical treatments result in damage to muscular and nervous tissues, and so are reserved only for nonresponders to BTX-A therapy.21

BTX-A can provide effective relief and is the treatment of choice for focal limb dystonias.22 Goals of treatment include functional improvement, correction of abnormal posture, and relief from discomfort. Although a variety of oral medications may also be prescribed, drug toxicity and adverse effects can outweigh the benefit and are usually only used in cases of severe dystonia. Oral medications used for limb dystonia include anticholinergics, dopamine agonists and antagonists, baclofen, clonazepam or other benzodiazepines, and muscle relaxants.

Antibodies may bind to the drug in a small percentage of patients who regularly receive injections of BTX, rendering additional injections of that specific serotype of BTX ineffective. This immunoresistance can be avoided if clinicians inject only the smallest quantity of BTX that achieves clinical efficacy, avoid administering booster injections before the end of the minimum 12-week lockout period, and extend the period between treatments as long as possible. If immunoresistance does occur, the BTX should be exchanged for a different serotype.

Testing for nonresponse

Patients are said to be nonresponders to BTX therapy if at 4 to 6 weeks following injection they show no reduction in muscle tone. A functional test for nonresponse is to inject a small amount of BTX into either the frontalis or sternocleidomastoid muscle prior to starting treatment; asymmetric weakness demonstrates a response, indicating that either injection technique or muscle selection is the problem. In addition to the development of neutralizing antibodies, other possible reasons for nonresponse include a dose that is too low or an alteration in the pattern of muscles involved in the dystonic movement.

Dystonia is a movement disorder in which involuntary sustained muscle contractions cause twisting movements that place the body in abnormal, sometimes painful, positions. Dystonia is believed to arise from an abnormality in the basal ganglia and an inherent or acquired defect in the processing of neurotransmitters.1

Dystonia is uncommon, although its exact prevalence is unknown. Nutt et al concluded that at least 250,000 people were affected by idiopathic dystonia in the United States, but prevalence is likely higher because misdiagnosis is not uncommon.2 A more recent European study found the prevalence of primary dystonia in the general population aged 50 years or more to be 732 per 100,000.3 The Epidemiological Study of Dystonia in Europe (ESDE) Collaborative Group found that the estimated prevalence of cervical dystonia was 50 to 200 per 1 million individuals.4 Also known as spasmodic torticollis, this is the most commonly diagnosed form of focal dystonia.

CLASSIFICATION OF DYSTONIA

Accurate classification of dystonia is important, since this informs approaches to management as well as prognosis. The three most important means by which dystonia is classified are (1) etiology, including primary dystonia, which encompasses a variety of genetic variables, and secondary dystonia; (2) bodily distribution of symptoms; and (3) age at onset.

Etiology

Most primary or idiopathic dystonia appears to be hereditary. Early-onset primary dystonia is most frequently caused by a mutation in the DYT1 gene, although other genetic mutations are possible.5 Patients with primary dystonia have no other underlying disorder; involuntary muscle contractions are the sole symptom. A thorough history should include a review of perinatal and early developmental history, prior neurologic illness, and exposure to drugs known to cause acquired dystonia. Physical examinations (encompassing intellectual, pyramidal, cerebellar, and sensory domains) and laboratory tests reveal no specific cause for the dystonic symptoms. Primary dystonia is also most frequently action-induced; at rest, the affected body region may appear to be normal.

Secondary dystonia occurs as a symptom of another disease process. Multiple sclerosis or any one of several hereditary neurologic disorders, such as Wilson disease, may be implicated. Secondary dystonia also may result from trauma to the brain, as might occur during an automobile accident; from heavy-metal or carbon monoxide poisoning; or as an adverse effect of medication. It may be psychogenic or related to Parkinson disease or Parkinson-plus syndromes, a group of neurodegenerative disorders with parkinsonian features. Tardive dystonia, the most common adult form of secondary dystonia, may occur follow ing exposure to certain neuroleptic drugs; tardive dystonia is a type of tardive dyskinesia that describes any involuntary neurologic movement disorder.

Bodily distribution

Dystonia is further classified by location of symptoms. Focal dystonias, which are usually primary dystonias, describe symptoms that are limited to a region of the body, such as a specific arm. There are several variations. Cervical dystonia affects the head and neck, is the most common adult-onset dystonia, and affects more women than men. Blepharospasm, or involuntary contractions of the eyelids, potentially leads to extended eye closure and functional blindness and often involves other facial muscles. Laryngeal dystonia affects the muscles in the larynx. Limb dystonia, such as writer’s or musician’s cramp, affects muscles in the arm, hand, leg, or foot. Limb dystonia is often task-specific action dystonia, and can be primary or secondary.

Segmental dystonia describes a group of involved muscles that are contiguous, such as cranial to neck to cervical to arm. Oromandibular dystonia, affecting the face, mouth, and jaw, often with unusual tongue movements (ie, lingual dystonia), is a type of segmental dystonia, although some consider it a focal dystonia. Meige syndrome is the combination of blepharospasm and oromandibular dystonia. Certain limb and cranial dystonias are considered segmental dystonias. Dystonia that affects two or more noncontiguous muscle groups in different parts of the body is multifocal. Hemi dystonia describes unilateral symptoms.

Symptoms that have advanced from a focal presentation to affect additional regions of the body characterize generalized dystonia. The symptoms potentially advance to include the trunk and limbs. The muscular contractions are usually sustained, are often both repetitive and painful, and worsen with activity.6 In severe cases, muscular contractions may occur even while resting. Early-onset myoclonus dystonia is a generalized hereditary dystonia whose symptoms include dystonic contractions of the neck and shoulders and rapid jerking movements.7 Of note diagnostically, early-onset dystonia in a leg typically begins at age 8 to 9 years and is more likely than other early-onset presentations to progress to generalized dystonia. Early-onset dystonia that begins in an arm typically presents later, at age 12 to 14 years, and is less likely to progress to generalized dystonia. Late-onset dystonia (> 27 years of age), by contrast, rarely begins in a leg and tends to remain either focal or segmental.8

Age of onset

A third useful classification scheme identifies early-onset (childhood to young adult) and late-onset varieties of dystonia.

THE DIAGNOSTIC CHALLENGE

Accurate diagnosis of dystonia is challenging because of its relative rarity and the variety of etiologies that pertain to this heterogeneous family of disorders. Patterns of inheritance are not straightforward and primary dystonia can be difficult to diagnose even with the benefit of genetic testing. There is no identifiable pathologic abnormality in many patients, and negative genetic tests do not necessarily mean that the dystonia is not primary. In the face of these challenges it is not surprising that dystonia is frequently misdiagnosed (Table 1). Nevertheless, certain findings can guide the diagnosis toward primary or secondary dystonia.

Consider primary dystonia if perinatal and developmental histories, intellect, strength, and perception of sensations are normal. There should be no prior history of neurologic illness or exposure to neuroleptic drugs whose adverse effects include secondary dystonia. In primary dystonia, diagnostic studies are negative and dystonia is the only symptom. If onset of symptoms is associated with activity, then primary dystonia should be considered. In the case of early- or late-onset limb dystonia, testing should be performed for the DYT1 gene. If the results are negative, then a trial for dopa-responsive dystonia should be undertaken with levodopa.

Consider secondary dystonia if the patient has been exposed to neuroleptic drugs, symptoms are distributed unilaterally, or the presentation is unusual for age or distribution of symptoms. For example, cranial dystonia in a child would raise the index of suspicion for secondary dystonia. If tardive dystonia is part of the differential diagnosis, consider magnetic resonance imaging (MRI), serum ceruloplasmin measurement, or slit-lamp diagnostic testing. Suspicion of a structural lesion affecting the central nervous system warrants examination with MRI, computed tomography, or angiography. Certain metabolic and neurologic hereditary disorders cause secondary dystonia, in which case dopa-responsive dystonia should be ruled out. Psychometric testing should also be considered.

 

 

SYMPTOMATIC TREATMENT WITH CHEMODENERVATION

In the absence of a cure, treatment options for dystonia are necessarily symptomatic and supportive. Titratable chemo denervation agents are injected directly into the muscle or motor nerve, temporarily weakening the local muscle and easing dystonia symptoms. Chemo denervation agents include phenol, ethyl alcohol, and botulinum toxin types A (BTX-A; onabotulinumtoxinA, abo botulinumtoxinA, and incobotulinumtoxinA) and B (BTX-B; rimabotulinum toxinB).

Phenol and ethyl alcohol injections targeted perineurally or as a motor point block have been employed for dystonia and cause nonselective tissue destruction, muscle necrosis, and highly variable durations of response. Perineural microcirculation may be damaged, possibly leading to long-term defects.

Clostridium botulinum bacteria produce seven serologically distinct neuroparalytic toxins. They are the most powerful such toxins currently known and temporarily prevent acetylcholine vesicles from docking into the presynaptic neuromuscular junction. Use of BTX-A for treatment of dystonia was recommended in a National Institutes of Health consensus statement in 1990.9 It has been studied for a variety of dystonias, including blepharospasm, hemifacial spasm, laryngeal dystonia, oromandibular dystonia, and cervical dystonia, among other focal dystonias. Lew et al reported in 1997 on the successful use of BTX-B for cervical dystonia in a double-blind, single-treatment study,10 and confirmatory studies followed.11,12

Varying indications for botulinum toxin

US Food and Drug Administration–approved indications for the toxins vary. The three BTX-A products and the single BTX-B product are approved for the treatment of cervical dystonia in adults to reduce the severity of abnormal head position and neck pain. OnabotulinumtoxinA is approved for treatment of blepharospasm and strabismus associated with dystonia; and incobotulinumtoxinA is approved for blepharospasm in patients who have previously been treated with onabotulinumtoxinA. BTX-A has also been found to be safe and effective for the management of focal dystonias. These botulinum toxin agents are not equivalent in dosing units, so caution must be observed when switching brands.

Patients selected to receive BTX for dystonia should meet three criteria:

  • The dystonia should interfere with their functioning, comfort, or care to the degree that causes impairment and affects activities of daily living;
  • Focal weakening following administration of the drug should not decrease their level of function; and
  • The patient should understand that use of BTX may not completely address positioning, posturing, or secondary deformities.

Contraindications include pregnancy, lactation, comorbid neuromuscular disease (eg, amyotrophic lateral sclerosis or myasthenia gravis), and use of an aminoglycoside.

The need for BTX therapy should be reevaluated prior to each treatment; clinical benefit lasts 3 months or more. Electromyography may facilitate the location of target muscles, particularly since involved musculature may not be palpable and is often not superficial.13 In-office tools that help document baseline and posttreatment results, including videotaping dystonic limb movements and the use of rating scales, can be important for evaluating the patient’s progress.14

Relief for cervical dystonia

The treatment of choice for focal dystonias and focal aspects of generalized dystonia is BTX. Both BTX-A and BTX-B offer effective palliative treatments for cervical dystonia by improving neck position, reducing pain, and decreasing disability in sufferers.11,15–18 The BTX solution is injected directly into the dystonic muscle at several locations, temporarily weakening the overactive muscle. The BTX dose is approximately proportional to the size of the muscle, although smaller muscles typically responsible for precision movement may require a relatively larger dose (Table 2). Doses may be modified according to clinical factors such as muscle bulk and severity of dystonia (Table 3).

Relief following BTX injection for cervical dystonia occurs about 1 week later, with the greatest effect seen at about 2 to 6 weeks following injection; relief may last 12 to 16 weeks. Reinjections are not normally administered prior to 12 weeks’ duration in order to reduce the possibility of antibody formation. Concomitant interventions addressing depression and anxiety may have a significant effect on overall quality of life.19 Patients may also try several sensory tricks, called gestes antagoniste, which may temporarily reduce or alleviate the dystonia. However, these tactile procedures—such as placing a hand on top of the head—lose their effectiveness over time.

Treatment of blepharospasm, focal limb dystonia

The use of BTX-A for blepharospasm is a significant improvement over the former clinical reliance on various oral medications, which, with the exception of baclofen, proved largely ineffective.20 Surgical treatments result in damage to muscular and nervous tissues, and so are reserved only for nonresponders to BTX-A therapy.21

BTX-A can provide effective relief and is the treatment of choice for focal limb dystonias.22 Goals of treatment include functional improvement, correction of abnormal posture, and relief from discomfort. Although a variety of oral medications may also be prescribed, drug toxicity and adverse effects can outweigh the benefit and are usually only used in cases of severe dystonia. Oral medications used for limb dystonia include anticholinergics, dopamine agonists and antagonists, baclofen, clonazepam or other benzodiazepines, and muscle relaxants.

Antibodies may bind to the drug in a small percentage of patients who regularly receive injections of BTX, rendering additional injections of that specific serotype of BTX ineffective. This immunoresistance can be avoided if clinicians inject only the smallest quantity of BTX that achieves clinical efficacy, avoid administering booster injections before the end of the minimum 12-week lockout period, and extend the period between treatments as long as possible. If immunoresistance does occur, the BTX should be exchanged for a different serotype.

Testing for nonresponse

Patients are said to be nonresponders to BTX therapy if at 4 to 6 weeks following injection they show no reduction in muscle tone. A functional test for nonresponse is to inject a small amount of BTX into either the frontalis or sternocleidomastoid muscle prior to starting treatment; asymmetric weakness demonstrates a response, indicating that either injection technique or muscle selection is the problem. In addition to the development of neutralizing antibodies, other possible reasons for nonresponse include a dose that is too low or an alteration in the pattern of muscles involved in the dystonic movement.

References
  1. Dystonia fact sheet. National Institute of Neurological Disorders and Stroke Web Site. http://www.ninds.nih.gov/disorders/dystonias/detail_dystonias.htm. Updated October 27, 2011. Accessed April 17, 2012.
  2. Nutt JG, Muenter MD, Aronson A, Kurland LT, Melton LJ. Epidemiology of focal and generalized dystonia in Rochester, Minnesota. Mov Disord 1988; 3:188194.
  3. Muller J, Kiechl S, Wenning GK, et al. The prevalence of primary dystonia in the general community. Neurology 2002; 59:941943.
  4. Epidemiological Study of Dystonia in Europe Collaborative Group. A prevalence study of primary dystonia in eight European countries. J Neurol 2000; 24:787793.
  5. Klein C, Kann M, Kis B, et al Genetics of dystonia. Nervenarz 2000; 71:431441.
  6. Fahn S, Marsden CD, Calne DB. Classification and investigation of dystonia. In:Marsden CD, Fahn S, eds. Movement Disorders 2. London, UK: Butterworth; 1987:332358.
  7. Doheny D, Danisi F, Smith C, et al. Clinical findings of a myoclonus-dystonia family with two distinct mutations. Neurology 2002; 59:11301131.
  8. Greene P, Kang UJ, Fahn S. Spread of symptoms in idiopathic torsion dystonia. Mov Disord 1995; 10:143152.
  9. Clinical use of botulinum toxin. NIH Consens Statement 1990; 8:120.
  10. Lew MF, Adornato BT, Duane DD, et al. Botulinum toxin type B: a double-blind, placebo-controlled, safety and efficacy study in cervical dystonia. Neurology 1997; 49:701707.
  11. Brin MF, Lew MF, Adler CH, et al. Safety and efficacy of Neuro-Bloc (botulinum toxin type B) in type A-resistant cervical dystonia. Neurology 1999; 53:14311438.
  12. Brashear A, Lew MF, Dykstra DD, et al. Safety and efficacy of NeuroBloc (botulinum toxin type B) in type A-responsive cervical dystonia. Neurology 1999; 53:14391446.
  13. Dressler D. Electromyographic evaluation of cervical dystonia for planning of botulinum toxin therapy. Eur J Neurol 2000; 7:713718.
  14. Dystonia rating scales and scoring sheets. Movement Disorders Virtual University Web site. http://www.mdvu.org/library/ratingscales/dystonia/. Updated April 21, 2008. Accessed April 18, 2012.
  15. Brashear A. The botulinum toxins in the treatment of cervical dystonia. Semin Neurol 2001; 21:8590.
  16. Brashear A, Watts MW, Marchetti A, Magar R, Lau H, Wang L. Duration of effect of botulinum toxin type A in adult patients with cervical dystonia: a retrospective chart review. Clin Ther 2000; 22:15161524.
  17. Ceballos-Baumann AO. Evidence-based medicine in botulinum toxin therapy for cervical dystonia. J Neurol 2001; 248( suppl 1):1420.
  18. Giladi N, Meer J, Kidan H, Honigman S. Long-term remission of idiopathic cervical dystonia after treatment with botulinum toxin. Eur Neurol 2000; 44:144146.
  19. Ben-Shlomo Y, Camfield L, Warner T, ESDE Collaborative Group. What are the determinants of quality of life in people with cervical dystonia? J Neurol Neurosurg Psychiatry 2002; 72:608614.
  20. Fahn S, Hening WA, Bressman S, et al. Long-term usefulness of baclofen in the treatment of essential blepharospasm. Adv Ophthal Plastic Reconstr Surg 1985; 4:219226.
  21. Callahan A. Blepharospasm with resection of part of orbicularis nerve supply. Arch Ophthalmol 1963; 70:508511.
  22. Yoshimura DM, Aminoff MJ, Olney RK. Botulinum toxin therapy for limb dystonias. Neurology 1992; 42:627630.
References
  1. Dystonia fact sheet. National Institute of Neurological Disorders and Stroke Web Site. http://www.ninds.nih.gov/disorders/dystonias/detail_dystonias.htm. Updated October 27, 2011. Accessed April 17, 2012.
  2. Nutt JG, Muenter MD, Aronson A, Kurland LT, Melton LJ. Epidemiology of focal and generalized dystonia in Rochester, Minnesota. Mov Disord 1988; 3:188194.
  3. Muller J, Kiechl S, Wenning GK, et al. The prevalence of primary dystonia in the general community. Neurology 2002; 59:941943.
  4. Epidemiological Study of Dystonia in Europe Collaborative Group. A prevalence study of primary dystonia in eight European countries. J Neurol 2000; 24:787793.
  5. Klein C, Kann M, Kis B, et al Genetics of dystonia. Nervenarz 2000; 71:431441.
  6. Fahn S, Marsden CD, Calne DB. Classification and investigation of dystonia. In:Marsden CD, Fahn S, eds. Movement Disorders 2. London, UK: Butterworth; 1987:332358.
  7. Doheny D, Danisi F, Smith C, et al. Clinical findings of a myoclonus-dystonia family with two distinct mutations. Neurology 2002; 59:11301131.
  8. Greene P, Kang UJ, Fahn S. Spread of symptoms in idiopathic torsion dystonia. Mov Disord 1995; 10:143152.
  9. Clinical use of botulinum toxin. NIH Consens Statement 1990; 8:120.
  10. Lew MF, Adornato BT, Duane DD, et al. Botulinum toxin type B: a double-blind, placebo-controlled, safety and efficacy study in cervical dystonia. Neurology 1997; 49:701707.
  11. Brin MF, Lew MF, Adler CH, et al. Safety and efficacy of Neuro-Bloc (botulinum toxin type B) in type A-resistant cervical dystonia. Neurology 1999; 53:14311438.
  12. Brashear A, Lew MF, Dykstra DD, et al. Safety and efficacy of NeuroBloc (botulinum toxin type B) in type A-responsive cervical dystonia. Neurology 1999; 53:14391446.
  13. Dressler D. Electromyographic evaluation of cervical dystonia for planning of botulinum toxin therapy. Eur J Neurol 2000; 7:713718.
  14. Dystonia rating scales and scoring sheets. Movement Disorders Virtual University Web site. http://www.mdvu.org/library/ratingscales/dystonia/. Updated April 21, 2008. Accessed April 18, 2012.
  15. Brashear A. The botulinum toxins in the treatment of cervical dystonia. Semin Neurol 2001; 21:8590.
  16. Brashear A, Watts MW, Marchetti A, Magar R, Lau H, Wang L. Duration of effect of botulinum toxin type A in adult patients with cervical dystonia: a retrospective chart review. Clin Ther 2000; 22:15161524.
  17. Ceballos-Baumann AO. Evidence-based medicine in botulinum toxin therapy for cervical dystonia. J Neurol 2001; 248( suppl 1):1420.
  18. Giladi N, Meer J, Kidan H, Honigman S. Long-term remission of idiopathic cervical dystonia after treatment with botulinum toxin. Eur Neurol 2000; 44:144146.
  19. Ben-Shlomo Y, Camfield L, Warner T, ESDE Collaborative Group. What are the determinants of quality of life in people with cervical dystonia? J Neurol Neurosurg Psychiatry 2002; 72:608614.
  20. Fahn S, Hening WA, Bressman S, et al. Long-term usefulness of baclofen in the treatment of essential blepharospasm. Adv Ophthal Plastic Reconstr Surg 1985; 4:219226.
  21. Callahan A. Blepharospasm with resection of part of orbicularis nerve supply. Arch Ophthalmol 1963; 70:508511.
  22. Yoshimura DM, Aminoff MJ, Olney RK. Botulinum toxin therapy for limb dystonias. Neurology 1992; 42:627630.
Page Number
S25-S29
Page Number
S25-S29
Publications
Publications
Article Type
Display Headline
Use of chemodenervation in dystonic conditions
Display Headline
Use of chemodenervation in dystonic conditions
Citation Override
Cleveland Clinic Journal of Medicine 2012 July;79(suppl 2):S25-S29
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media

Comprehensive treatment of Huntington disease and other choreic disorders

Article Type
Changed
Mon, 10/01/2018 - 15:36
Display Headline
Comprehensive treatment of Huntington disease and other choreic disorders

Chorea is characterized by continuous, random, brief, involuntary muscle contractions that result from a variety of causes.1 These involuntary movements are nonstereotyped and irregular. Although choreic disorders are among the most common involuntary movement disorders, their diagnosis and treatment present several important challenges, including identifying and removing the cause if possible, controlling and preventing motor symptoms, and managing neuropsychologic complications.1 This article provides an overview of the diagnosis and treatment of choreic disorders, using Sydenham chorea to illustrate the management of autoimmune choreas and Huntington disease as the model for the management of heritable choreas.

Management of choreic disorders begins with a first-pass diagnosis and the use of symptomatic therapies. Even if this first pass yields no firm diagnosis, it at least rules out causes that have the most practical significance. A subsequent second-pass evaluation can be undertaken to look for rarer causes. Symptomatic therapies are continued throughout the diagnostic period. More specific therapies can be administered if an etiologic or pathogenic mechanism is determined (eg, postinfectious, autoimmune, metabolic).

CHOREIC DISORDERS: A PRACTICAL DIAGNOSTIC APPROACH

In general, choreic disorders may be subdivided into six categories:

1. Heredodegenerative disorders, such as Huntington disease and other genetically heterogeneous choreas, include Huntington disease–like 2 (HDL2) and benign hereditary chorea.1 Sporadic cases include those of unknown paternity; X-linked disorders (eg, McLeod syndrome); and autosomal-recessive disorders such as chorea-acanthocytosis, which is characterized by chorea, dystonia with prominent orofacial involvement, self-mutilation, myopathy, and neuropathy.2

2. Drug-induced choreas include neurolepticinduced tardive dyskinesia and nontardive hyperkinetic drug-related choreas, the most common of which is levodopa-induced dyskinesias.3 Tardive drug-induced choreas may occur while using the culprit drug, while tapering the drug, or after it has been discontinued. The culprit drugs are represented by dopamine-receptor blockers and include the first-generation neuroleptics (eg, phenothiazines, haloperidol), antidepressants (lox-apine), and gastrointestinal agents (metoclopramide, prochlorperazine). Drug-induced choreas are possible with a wide range of pharmacologic agents, including antiparkinsonian drugs (eg, levodopa, dopamine agonists, anticholinergics), sympathomimetics (eg, amphetamines, cocaine), anticonvulsants, calcium channel blockers, and oral contraceptives.1

3. Autoimmune choreas include Sydenham chorea, systemic lupus erythematosus, and antiphospholipid antibody syndromes. The latter encompass lupus anticoagulant and anticardiolipin antibodies.

4. Metabolic choreas are most often associated with hyperthyroidism, although case reports have described choreas in patients with vitamin B12 deficiency. A variety of hereditary metabolic diseases are also included in this category.

5. Vascular choreas include polycythemia vera and cerebrovascular accidents, the latter frequently presenting as hemiballismus. Polycythemia vera is associated with a high incidence of neurologic symptoms, including a reported incidence of chorea of 0.5% to 5%,4 and should be considered as a potential cause of chorea.

6. Other choreic disorders include a variety of entities such as rare paraneoplastic disorder/syndrome, and posttraumatic and postanoxic presentations.

The first-pass diagnostic approach includes a family history, drug history, and brain magnetic resonance imaging to identify potential structural causes of chorea. Genetic testing for Huntington disease or other choreic disorders may also be performed, although it is essential to consider the potential implications of a positive test result. Intensive pretest and posttest counseling is important both for the patient and for currently asymptomatic family members who may also be affected.1

Other testing includes:

  • Complete blood count
  • Creatine phosphokinase
  • Peripheral smear for acanthocytes
  • Comprehensive metabolic panel
  • Ceruloplasmin level
  • Measurement of thyroxine (T4) and triiodothyronine (T3)
  • B12 tests
  • Antinuclear antibody sedimentation rate
  • Lupus anticoagulant-anticardiolipin antibodies
  • Antistreptolysin O (ASO) titer
  • Anti-DNase-B titer.

GENERAL CONSIDERATIONS FOR THE TREATMENT OF CHOREIC DISORDERS

In some cases, choreic disorders have a treatable underlying etiology, such as thyroid disease or vitamin B12 deficiency. Tardive syndromes may require treatment beyond drug discontinuation, including use of dopamine depleters for the classic tardive dyskinetic syndromes and anticholinergics for the tardive dystonic syndromes. Levodopa dyskinesia may be treated using amantadine, clozapine, or deep brain stimulation.5,6 The treatment of patients with autoimmune choreas is not well defined. It may include anticoagulation in patients with positive anticardiolipin antibodies to prevent venous or arterial thromboembolism,7 but the risk of arterial thromboembolism is uncertain, and it is unclear whether chorea is truly a harbinger of vascular events.

A negative ASO titer does not exclude Sydenham chorea, a result of childhood infection with group-A beta-hemolytic streptococcus, and antibiotics should be considered in the appropriate context. Some researchers have argued that immune responses associated with acute infections may result in autoimmune neuropsychiatric symptoms. In pediatric patients, this has been referred to as pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections (PANDAS).8

A related phenomenon has been proposed as a potential mechanism of some types of chorea, although the relationship between acute infection and chorea is controversial. Patients with elevated ASO titer or anti-DNase-B titers may be candidates for antibiotics. By 6 weeks after the onset of infection, these titers will fall and a diagnosis of Sydenham chorea can be postulated or based exclusively on clinical judgment.

 

 

SYDENHAM CHOREA

Unique to Sydenham chorea is the use of penicillin as prophylaxis. Other than that, the management of Sydenham chorea exemplifies the management approach for the larger category of autoimmune choreic disorders. Pathogenic-based treatment options include immune modulation with cortico steroids, intravenous immunoglobulin (IVIG), and plasma exchange; all treatments must be administered in the appropriate clinical context.

One double-blind clinical trial examined the effectiveness of corticosteroid treatment in children with Sydenham chorea randomly assigned to receive either prednisone (n = 22) or placebo (n = 15).9 Prednisone was administered at a dose of 2 mg/kg/day for 4 weeks, followed by gradual tapering and discontinuation. The median time to remission of chorea was significantly lower for patients in the prednisone group (54.3 days) compared with those in the placebo group (119.9 days; P < .001). Patients in the prednisone group also exhibited significantly better scores on a chorea intensity rating scale at 8 weeks and 12 weeks (P < .001). Potential limitations of this approach include relapse of chorea symptoms and corticosteroid-related adverse events (eg, Cushing syndrome, hypertension).

A second study compared the effectiveness of three modalities: IVIG at a dose of 1 g/kg/day for 2 days (n = 4), plasma exchange (n = 8), and prednisone (n = 6).10 Although differences between treatment groups were not statistically significant, the authors noted that the clinical improvement in chorea symptoms tended to be greater for patients who received IVIG or plasma exchange than for those who received prednisone. Mean chorea scores improved from baseline by 72% for the IVIG group, 50% for the plasma exchange group, and 29% for the prednisone group.

After etiology-dependent treatments have been considered, several other options may be effective regardless of the specific etiology. These include symptomatic treatments such as haloperidol, atypical neuroleptics, and amantadine.11 Antiepileptic medications or benzodiazepines may also help to control symptoms, although less information is available about the use of these agents for the treatment of Sydenham chorea. Tetrabenazine may be considered for patients who will require long-term treatment.

HUNTINGTON DISEASE

Pharmacotherapy of Huntington disease may be unnecessary if symptoms are mild or not bothersome. Symptomatic treatment options include tetrabenazine, amantadine, and either first-generation neuroleptics (eg, haloperidol) or second-generation atypical neuroleptics (eg, olanzapine, quetiapine, risperidone, ziprasidone).

Treating choreas with tetrabenazine or amantadine

Considerable recent attention has focused on the efficacy and safety of tetrabenazine for the treatment of Huntington disease and other choreic disorders. Tetrabenazine is a central monoamine depleter that reversibly binds to the type-2 vesicular monoamine transporter.12 The TETRA-HD study examined the efficacy and safety of tetrabenazine for the short- and long-term control of Huntington disease.12 An initial study compared tetrabenazine with placebo in 75 patients who were treated for up to 13 weeks. In an extension study, all patients received individualized tetrabenazine doses for up to 80 weeks.

Figure 1. Mean total maximal chorea scores decreased markedly during the first 10 weeks of tetrabenazine treatment for Huntington disease, remained below baseline through 80 weeks of treatment, and then returned to baseline after tetrabenazine discontinuation.12

The mean total maximal chorea (TMC) scores from the Unified Huntington Disease Rating Scale (UHDRS) decreased markedly during the first 10 weeks of tetrabenazine treatment, remained lower than baseline throughout 80 weeks, and then returned to baseline levels after tetrabenazine discontinuation (Figure 1). At week 80, the mean TMC score was reduced by 4.6 UHDRS units compared with baseline (P < .001) The long-term extension phase was completed by 45 of 75 patients. Treatment-related adverse events that prompted discontinuation included depression, delusions, and vocal tics. The most commonly reported adverse events included sedation or somnolence (n = 18), depressed mood (n = 17), anxiety (n = 13), insomnia (n = 10), and akathisia (n = 9). Scores of parkinsonism and dysphagia increased significantly from baseline over the 80-week study.

Amantadine is an option for patients who cannot tolerate tetrabenazine. A double-blind, placebo-controlled study performed by researchers at the National Institutes of Health (NIH) examined the efficacy and safety of amantadine in 24 patients with Huntington disease.13 Patients were treated with oral amantadine 400 mg/day or placebo for 2 weeks, and were then crossed over to the other treatment. Amantadine was associated with a median reduction in extremity chorea score at rest of 36% from baseline (P = .04), versus 0% improvement with placebo. The mean improvement with amantadine was 56% for the 10 patients with the highest drug plasma levels.

Improvement in chorea scores from baseline for amantadine compared with placebo was rated with four different methods: (1) maximal chorea severity measured from video recordings; (2) maximal chorea severity measured by live raters; (3), chorea severity at rest measured from video recordings; and (4) extremity chorea at rest measured from video recordings. Amantadine was superior to placebo according to all four rating methods. Treatment was generally safe and well tolerated, and no consistent changes in cognitive function were noted with amantadine therapy.

Reprinted with permission from Neurology (Lucetti C, et al. IV amantadine improves chorea in Huntington’s disease: an acute randomized, controlled study. Neurology 2003; 60:1995–1997). Copyright © 2003 by AAN Enterprises, Inc.
Figure 2. During the randomized placebo-controlled phase of a study that compared amantadine with placebo, mean dyskinesia scores, measured using the Abnormal Involuntary Movement Scale scores, decreased significantly 90 minutes after initiation of amantadine infusion compared with placebo.14aP < .05
A second study examined the effects of amantadine as a 2-hour IV infusion in nine patients with Huntington disease.14 Amantadine or placebo was administered in a randomized, double-blind manner on the first day of the study, and patients were then crossed over to the other treatment on the second day. All patients then received open-label oral amantadine for an additional 1 year. During the randomized placebo-controlled phase, mean dyskinesia scores, evaluated using the Abnormal Involuntary Movement Scale, were significantly lower for patients randomly assigned to amantadine compared with placebo. During the randomized placebo-controlled phase, the decrease in mean dyskinesia score was significantly greater 90 minutes after treatment with amantadine compared with placebo (Figure 2). In the open-label amantadine continuation phase, oral amantadine was associated with a further gradual improvement in symptoms over 3 to 6 months. No significant changes were observed in neuropsychologic tests or psychiatric rating scales.

 

 

Managing nonmotor complications

In addition to addressing chorea, it is also important to manage nonmotor complications of Huntington disease, including cognition, mood, and thought disorders. Rivastigmine was assessed for the treatment of motor symptoms, functional disability, and cognitive impairment associated with Huntington disease in an open-label study of 18 patients; 11 received rivastigmine 6 mg/day and 7 control patients did not.15 Motor and cognitive function were assessed for up to 2 years by raters who were blinded to treatment assignment. Ratings on a global motor performance scale were significantly better for patients who received rivastigmine than for control subjects. Rivastigmine treatment was also associated with trends toward improvements in functional disability and cognitive impairment, although these differences were not statistically significant.

A small open-label study examined the effects of donepezil for movement and cognitive symptoms associated with Huntington disease.16 Donepezil did not significantly improve cognitive symptoms, although the study enrolled only eight patients. All patients tolerated oral donepezil at a dose of 5 mg/day, but four patients withdrew from the study when the dose was increased to 10 mg/day. In two patients, chorea worsened and falls increased, moderate to severe diarrhea developed in three patients, and one patient reported anxiety and irritability.

Depression is another common complication of Huntington disease. The incidence of depression among patients with Huntington disease is approximately 40%, and the risk of suicide is at least eightfold greater than that among the general population.17 Treatment must be guided by clinical judgment. Selective serotonin reuptake inhibitor antidepressants have been recommended. Other options to manage depression include mirtazapine, monoamine oxidase inhibitors, or electroshock therapy. Mood-stabilizing agents (eg, carbamazepine, lamotrigine, valproate) may also be indicated in helping with impulse control. Haloperidol and second-generation antipsychotics are used for the treatment of a broad range of psychiatric conditions, many of which may overlap with Huntington disease, including schizophrenia and schizophreniform disorder, schizoaffective disorder, bipolar disorder, dementia, and disruptive behavior.18 The risk of tardive dyskinesia may be as much as fivefold lower with second-generation antipsychotics.18 Many patients with Huntington disease require treatment for aggression. A variety of approaches are available, including behavior modification, the antidepressant sertraline, buspirone, antipsychotic agents (eg, risperidone, olanzapine), propranolol, and lithium (combined with haloperidol).

Long-term care considerations

As a consequence of the diverse clinical manifestations of choreic disorders in movement, function, mood, and cognition, the treatment of Huntington disease requires a multidisciplinary approach that involves a number of different health care specialties across the long-term course of the disorder. Members of the Huntington disease treatment team may include neurologists, psychiatrists, nurses, social workers, geneticists, physical therapists, occupational therapists, speech therapists, dietitians, and other supporting groups or professional societies. The clinical manifestations of Huntington disease may evolve over time, as symptoms such as bradykinesia, dystonia, rigidity, cognitive decline, and gait instability become more significant.19 As a result, optimal management strategies for patients with Huntington disease may change significantly across the long-term course of the disease. During the early course of the disease, the typical clinical presentation is largely hyperkinesis, irritability, and distractibility. These patients will require initiation of drug therapy and linkage to sources of support. In the later stages of the disease, the presentation shifts to a more hypokinetic and apathetic profile, and patients are more likely to require drug regimen review and modification, nursing home placement, and palliative care services.19

Another important concern in Huntington disease treatment is care of the caregiver. Surveys show that the key concerns of caregivers include the expertise of the health care professionals who are treating the patient and the availability of sufficient services in the community.20 Several resources are available for Huntington disease caregivers, including local support groups, the Huntington’s Disease Society of America, Q Foundation, and the Huntington Study Group. The Lundbeck pharmaceutical company operates a patient assistance program (LundbeckShare. com) as well as an information center that can be accessed toll free at (888)457-4273. Approximately 90% of patients who request copayment assistance qualify for aid, regardless of the type of insurance they carry.

SUMMARY AND CONCLUSIONS

The approach to a patient with chorea starts with a search for specifically treatable etiologies. Autoimmune, metabolic, and vascular causes should be sought first and treated. The symptomatic treatment of all choreas is based on the model described here for Huntington disease, and includes attention to cognitive, psychiatric, and social support issues. The recommended approach is multidisciplinary, with a change in the mix of services as the disease progresses. It is also important to recognize the burden of Huntington disease on the caregiver and consider steps to make this burden more manageable.

References
  1. Cardoso F, Seppi K, Mair KJ, Wenning GK, Poewe W. Seminar on choreas. Lancet Neurol 2006; 5:589602.
  2. Schneider SA, Walker RH, Bhatia KP. The Huntington’s disease-like syndromes: what to consider in patients with a negative Huntington’s disease gene test. Nat Clin Pract Neurol 2007; 3:517525.
  3. Zesiewicz TA, Sullivan KL. Drug-induced hyperkinetic movement disorders by nonneuroleptic agents. Handb Clin Neurol 2011; 100:347363.
  4. Margolis RL, Ross CA. Diagnosis of Huntington disease. Clin Chem 2003; 49:17261732.
  5. Rascol O. The pharmacological therapeutic management of levodopa-induced dyskinesias in patients with Parkinson’s disease. J Neurol 2000; 247 (suppl 2):II51II57.
  6. Fabbrini G, Brotchie JM, Grandas F, Nomoto M, Goetz CG. Levodopa-induced dyskinesias. Mov Disord 2007; 22:13791389.
  7. Lim W, Crowther MA, Eifelboom JW. Management of antiphospholipid antibody syndrome: a systematic review. JAMA 2006; 295:10501057.
  8. Mink J, Kurlan R. Acute postinfectious movement and psychiatric disorders in children and adolescents. J Child Neurol 2011; 26:214217.
  9. Paz JA, Silva CA, Marques-Dias MJ. Randomized double-blind study with prednisone in Sydenham’s chorea. Pediatr Neurol 2006; 34:264269.
  10. Garvey MA, Snider LA, Leitman SF, Werden R, Swedo SE. Treatment of Sydenham’s chorea with intravenous immunoglobulin, plasma exchange, or prednisone. J Child Neurol 2005; 20:424429.
  11. Walker KG, Wilmshurst JM. An update on the treatment of Sydenham’s chorea: the evidence for established and evolving interventions. Ther Adv Neurol Disord 2010; 3:301309.
  12. Frank S. Tetrabenazine as anti-chorea therapy in Huntington disease: an open-label continuation study. Huntington Study Group/TETRA-HD Investigators. BMC Neurol 2009; 9:62.
  13. Verhagen Metman L, Morris MJ, Farmer C, et al. Huntington’s disease: a randomized, controlled trial using the NMDA-antagonist amantadine. Neurology 2002; 59:694699.
  14. Lucetti C, Del Dotto P, Gamabaccini G, et al. IV amantadine improves chorea in Huntington’s disease: an acute randomized, controlled study. Neurology 2003; 60:19951997.
  15. de Tommaso M, Difruscolo O, Scirruicchio V, Specchio N, Livrea P. Two years’ follow-up of rivastigmine treatment in Huntington disease. Clin Neuropharmacol 2007; 30:4346.
  16. Fernandez HH, Friedman JH, Grace J, Beasin-Hazen S. Donepezil for Huntington’s disease. Mov Disord 2000; 15:173176.
  17. Schoenfeld M, Myers RH, Cupples LA, Berkman B, Sax DS, Clark E. Increased rate of suicide among patients with Huntington’s disease. J Neurol Neurosurg Psychiatry 1984; 47:12831287.
  18. Correll CU, Leucht S, Kane JM. Lower risk for tardive dyskinesia associated with second-generation antipsychotics: a systematic review of 1-year studies. Am J Psychiatry 2004; 161:414425.
  19. Phillips W, Shannon KM, Barker RA. The current clinical management of Huntington’s disease. Mov Disord 2008; 23:14911504.
  20. Skirton H, Williams JK, Jackson Barnette J, Paulsen JS. Huntington disease: families’ experiences of healthcare services. J Adv Nursing 2010; 66:500510.
Article PDF
Author and Disclosure Information

Carlos Singer, MD
Professor of Neurology, Chief, Center for Parkinson’s Disease and Movement Disorders, Leonard M. Miller School of Medicine, University of Miami, Miami, FL

Correspondence: Carlos Singer, MD, Center for Parkinson’s Disease and Movement Disorders, Clinical Research Building, Leonard M. Miller School of Medicine, University of Miami, 1120 NW 14th Street, 13th Floor, Miami, FL 33136; CSinger@med.miami.edu

Dr. Singer reported receiving grant support from Boehringer Ingelheim and Teva Pharmaceuticals, Inc.; and advisory committee or review panel membership for Lundbeck.

This article is based on Dr. Singer’s presentation at “The Annual Therapy Symposium on Movement Disorders for the Modern Clinician” held in Fort Lauderdale, Florida, on January 29, 2011. The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Singer.

Publications
Page Number
S30-S34
Author and Disclosure Information

Carlos Singer, MD
Professor of Neurology, Chief, Center for Parkinson’s Disease and Movement Disorders, Leonard M. Miller School of Medicine, University of Miami, Miami, FL

Correspondence: Carlos Singer, MD, Center for Parkinson’s Disease and Movement Disorders, Clinical Research Building, Leonard M. Miller School of Medicine, University of Miami, 1120 NW 14th Street, 13th Floor, Miami, FL 33136; CSinger@med.miami.edu

Dr. Singer reported receiving grant support from Boehringer Ingelheim and Teva Pharmaceuticals, Inc.; and advisory committee or review panel membership for Lundbeck.

This article is based on Dr. Singer’s presentation at “The Annual Therapy Symposium on Movement Disorders for the Modern Clinician” held in Fort Lauderdale, Florida, on January 29, 2011. The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Singer.

Author and Disclosure Information

Carlos Singer, MD
Professor of Neurology, Chief, Center for Parkinson’s Disease and Movement Disorders, Leonard M. Miller School of Medicine, University of Miami, Miami, FL

Correspondence: Carlos Singer, MD, Center for Parkinson’s Disease and Movement Disorders, Clinical Research Building, Leonard M. Miller School of Medicine, University of Miami, 1120 NW 14th Street, 13th Floor, Miami, FL 33136; CSinger@med.miami.edu

Dr. Singer reported receiving grant support from Boehringer Ingelheim and Teva Pharmaceuticals, Inc.; and advisory committee or review panel membership for Lundbeck.

This article is based on Dr. Singer’s presentation at “The Annual Therapy Symposium on Movement Disorders for the Modern Clinician” held in Fort Lauderdale, Florida, on January 29, 2011. The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Singer.

Article PDF
Article PDF

Chorea is characterized by continuous, random, brief, involuntary muscle contractions that result from a variety of causes.1 These involuntary movements are nonstereotyped and irregular. Although choreic disorders are among the most common involuntary movement disorders, their diagnosis and treatment present several important challenges, including identifying and removing the cause if possible, controlling and preventing motor symptoms, and managing neuropsychologic complications.1 This article provides an overview of the diagnosis and treatment of choreic disorders, using Sydenham chorea to illustrate the management of autoimmune choreas and Huntington disease as the model for the management of heritable choreas.

Management of choreic disorders begins with a first-pass diagnosis and the use of symptomatic therapies. Even if this first pass yields no firm diagnosis, it at least rules out causes that have the most practical significance. A subsequent second-pass evaluation can be undertaken to look for rarer causes. Symptomatic therapies are continued throughout the diagnostic period. More specific therapies can be administered if an etiologic or pathogenic mechanism is determined (eg, postinfectious, autoimmune, metabolic).

CHOREIC DISORDERS: A PRACTICAL DIAGNOSTIC APPROACH

In general, choreic disorders may be subdivided into six categories:

1. Heredodegenerative disorders, such as Huntington disease and other genetically heterogeneous choreas, include Huntington disease–like 2 (HDL2) and benign hereditary chorea.1 Sporadic cases include those of unknown paternity; X-linked disorders (eg, McLeod syndrome); and autosomal-recessive disorders such as chorea-acanthocytosis, which is characterized by chorea, dystonia with prominent orofacial involvement, self-mutilation, myopathy, and neuropathy.2

2. Drug-induced choreas include neurolepticinduced tardive dyskinesia and nontardive hyperkinetic drug-related choreas, the most common of which is levodopa-induced dyskinesias.3 Tardive drug-induced choreas may occur while using the culprit drug, while tapering the drug, or after it has been discontinued. The culprit drugs are represented by dopamine-receptor blockers and include the first-generation neuroleptics (eg, phenothiazines, haloperidol), antidepressants (lox-apine), and gastrointestinal agents (metoclopramide, prochlorperazine). Drug-induced choreas are possible with a wide range of pharmacologic agents, including antiparkinsonian drugs (eg, levodopa, dopamine agonists, anticholinergics), sympathomimetics (eg, amphetamines, cocaine), anticonvulsants, calcium channel blockers, and oral contraceptives.1

3. Autoimmune choreas include Sydenham chorea, systemic lupus erythematosus, and antiphospholipid antibody syndromes. The latter encompass lupus anticoagulant and anticardiolipin antibodies.

4. Metabolic choreas are most often associated with hyperthyroidism, although case reports have described choreas in patients with vitamin B12 deficiency. A variety of hereditary metabolic diseases are also included in this category.

5. Vascular choreas include polycythemia vera and cerebrovascular accidents, the latter frequently presenting as hemiballismus. Polycythemia vera is associated with a high incidence of neurologic symptoms, including a reported incidence of chorea of 0.5% to 5%,4 and should be considered as a potential cause of chorea.

6. Other choreic disorders include a variety of entities such as rare paraneoplastic disorder/syndrome, and posttraumatic and postanoxic presentations.

The first-pass diagnostic approach includes a family history, drug history, and brain magnetic resonance imaging to identify potential structural causes of chorea. Genetic testing for Huntington disease or other choreic disorders may also be performed, although it is essential to consider the potential implications of a positive test result. Intensive pretest and posttest counseling is important both for the patient and for currently asymptomatic family members who may also be affected.1

Other testing includes:

  • Complete blood count
  • Creatine phosphokinase
  • Peripheral smear for acanthocytes
  • Comprehensive metabolic panel
  • Ceruloplasmin level
  • Measurement of thyroxine (T4) and triiodothyronine (T3)
  • B12 tests
  • Antinuclear antibody sedimentation rate
  • Lupus anticoagulant-anticardiolipin antibodies
  • Antistreptolysin O (ASO) titer
  • Anti-DNase-B titer.

GENERAL CONSIDERATIONS FOR THE TREATMENT OF CHOREIC DISORDERS

In some cases, choreic disorders have a treatable underlying etiology, such as thyroid disease or vitamin B12 deficiency. Tardive syndromes may require treatment beyond drug discontinuation, including use of dopamine depleters for the classic tardive dyskinetic syndromes and anticholinergics for the tardive dystonic syndromes. Levodopa dyskinesia may be treated using amantadine, clozapine, or deep brain stimulation.5,6 The treatment of patients with autoimmune choreas is not well defined. It may include anticoagulation in patients with positive anticardiolipin antibodies to prevent venous or arterial thromboembolism,7 but the risk of arterial thromboembolism is uncertain, and it is unclear whether chorea is truly a harbinger of vascular events.

A negative ASO titer does not exclude Sydenham chorea, a result of childhood infection with group-A beta-hemolytic streptococcus, and antibiotics should be considered in the appropriate context. Some researchers have argued that immune responses associated with acute infections may result in autoimmune neuropsychiatric symptoms. In pediatric patients, this has been referred to as pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections (PANDAS).8

A related phenomenon has been proposed as a potential mechanism of some types of chorea, although the relationship between acute infection and chorea is controversial. Patients with elevated ASO titer or anti-DNase-B titers may be candidates for antibiotics. By 6 weeks after the onset of infection, these titers will fall and a diagnosis of Sydenham chorea can be postulated or based exclusively on clinical judgment.

 

 

SYDENHAM CHOREA

Unique to Sydenham chorea is the use of penicillin as prophylaxis. Other than that, the management of Sydenham chorea exemplifies the management approach for the larger category of autoimmune choreic disorders. Pathogenic-based treatment options include immune modulation with cortico steroids, intravenous immunoglobulin (IVIG), and plasma exchange; all treatments must be administered in the appropriate clinical context.

One double-blind clinical trial examined the effectiveness of corticosteroid treatment in children with Sydenham chorea randomly assigned to receive either prednisone (n = 22) or placebo (n = 15).9 Prednisone was administered at a dose of 2 mg/kg/day for 4 weeks, followed by gradual tapering and discontinuation. The median time to remission of chorea was significantly lower for patients in the prednisone group (54.3 days) compared with those in the placebo group (119.9 days; P < .001). Patients in the prednisone group also exhibited significantly better scores on a chorea intensity rating scale at 8 weeks and 12 weeks (P < .001). Potential limitations of this approach include relapse of chorea symptoms and corticosteroid-related adverse events (eg, Cushing syndrome, hypertension).

A second study compared the effectiveness of three modalities: IVIG at a dose of 1 g/kg/day for 2 days (n = 4), plasma exchange (n = 8), and prednisone (n = 6).10 Although differences between treatment groups were not statistically significant, the authors noted that the clinical improvement in chorea symptoms tended to be greater for patients who received IVIG or plasma exchange than for those who received prednisone. Mean chorea scores improved from baseline by 72% for the IVIG group, 50% for the plasma exchange group, and 29% for the prednisone group.

After etiology-dependent treatments have been considered, several other options may be effective regardless of the specific etiology. These include symptomatic treatments such as haloperidol, atypical neuroleptics, and amantadine.11 Antiepileptic medications or benzodiazepines may also help to control symptoms, although less information is available about the use of these agents for the treatment of Sydenham chorea. Tetrabenazine may be considered for patients who will require long-term treatment.

HUNTINGTON DISEASE

Pharmacotherapy of Huntington disease may be unnecessary if symptoms are mild or not bothersome. Symptomatic treatment options include tetrabenazine, amantadine, and either first-generation neuroleptics (eg, haloperidol) or second-generation atypical neuroleptics (eg, olanzapine, quetiapine, risperidone, ziprasidone).

Treating choreas with tetrabenazine or amantadine

Considerable recent attention has focused on the efficacy and safety of tetrabenazine for the treatment of Huntington disease and other choreic disorders. Tetrabenazine is a central monoamine depleter that reversibly binds to the type-2 vesicular monoamine transporter.12 The TETRA-HD study examined the efficacy and safety of tetrabenazine for the short- and long-term control of Huntington disease.12 An initial study compared tetrabenazine with placebo in 75 patients who were treated for up to 13 weeks. In an extension study, all patients received individualized tetrabenazine doses for up to 80 weeks.

Figure 1. Mean total maximal chorea scores decreased markedly during the first 10 weeks of tetrabenazine treatment for Huntington disease, remained below baseline through 80 weeks of treatment, and then returned to baseline after tetrabenazine discontinuation.12

The mean total maximal chorea (TMC) scores from the Unified Huntington Disease Rating Scale (UHDRS) decreased markedly during the first 10 weeks of tetrabenazine treatment, remained lower than baseline throughout 80 weeks, and then returned to baseline levels after tetrabenazine discontinuation (Figure 1). At week 80, the mean TMC score was reduced by 4.6 UHDRS units compared with baseline (P < .001) The long-term extension phase was completed by 45 of 75 patients. Treatment-related adverse events that prompted discontinuation included depression, delusions, and vocal tics. The most commonly reported adverse events included sedation or somnolence (n = 18), depressed mood (n = 17), anxiety (n = 13), insomnia (n = 10), and akathisia (n = 9). Scores of parkinsonism and dysphagia increased significantly from baseline over the 80-week study.

Amantadine is an option for patients who cannot tolerate tetrabenazine. A double-blind, placebo-controlled study performed by researchers at the National Institutes of Health (NIH) examined the efficacy and safety of amantadine in 24 patients with Huntington disease.13 Patients were treated with oral amantadine 400 mg/day or placebo for 2 weeks, and were then crossed over to the other treatment. Amantadine was associated with a median reduction in extremity chorea score at rest of 36% from baseline (P = .04), versus 0% improvement with placebo. The mean improvement with amantadine was 56% for the 10 patients with the highest drug plasma levels.

Improvement in chorea scores from baseline for amantadine compared with placebo was rated with four different methods: (1) maximal chorea severity measured from video recordings; (2) maximal chorea severity measured by live raters; (3), chorea severity at rest measured from video recordings; and (4) extremity chorea at rest measured from video recordings. Amantadine was superior to placebo according to all four rating methods. Treatment was generally safe and well tolerated, and no consistent changes in cognitive function were noted with amantadine therapy.

Reprinted with permission from Neurology (Lucetti C, et al. IV amantadine improves chorea in Huntington’s disease: an acute randomized, controlled study. Neurology 2003; 60:1995–1997). Copyright © 2003 by AAN Enterprises, Inc.
Figure 2. During the randomized placebo-controlled phase of a study that compared amantadine with placebo, mean dyskinesia scores, measured using the Abnormal Involuntary Movement Scale scores, decreased significantly 90 minutes after initiation of amantadine infusion compared with placebo.14aP < .05
A second study examined the effects of amantadine as a 2-hour IV infusion in nine patients with Huntington disease.14 Amantadine or placebo was administered in a randomized, double-blind manner on the first day of the study, and patients were then crossed over to the other treatment on the second day. All patients then received open-label oral amantadine for an additional 1 year. During the randomized placebo-controlled phase, mean dyskinesia scores, evaluated using the Abnormal Involuntary Movement Scale, were significantly lower for patients randomly assigned to amantadine compared with placebo. During the randomized placebo-controlled phase, the decrease in mean dyskinesia score was significantly greater 90 minutes after treatment with amantadine compared with placebo (Figure 2). In the open-label amantadine continuation phase, oral amantadine was associated with a further gradual improvement in symptoms over 3 to 6 months. No significant changes were observed in neuropsychologic tests or psychiatric rating scales.

 

 

Managing nonmotor complications

In addition to addressing chorea, it is also important to manage nonmotor complications of Huntington disease, including cognition, mood, and thought disorders. Rivastigmine was assessed for the treatment of motor symptoms, functional disability, and cognitive impairment associated with Huntington disease in an open-label study of 18 patients; 11 received rivastigmine 6 mg/day and 7 control patients did not.15 Motor and cognitive function were assessed for up to 2 years by raters who were blinded to treatment assignment. Ratings on a global motor performance scale were significantly better for patients who received rivastigmine than for control subjects. Rivastigmine treatment was also associated with trends toward improvements in functional disability and cognitive impairment, although these differences were not statistically significant.

A small open-label study examined the effects of donepezil for movement and cognitive symptoms associated with Huntington disease.16 Donepezil did not significantly improve cognitive symptoms, although the study enrolled only eight patients. All patients tolerated oral donepezil at a dose of 5 mg/day, but four patients withdrew from the study when the dose was increased to 10 mg/day. In two patients, chorea worsened and falls increased, moderate to severe diarrhea developed in three patients, and one patient reported anxiety and irritability.

Depression is another common complication of Huntington disease. The incidence of depression among patients with Huntington disease is approximately 40%, and the risk of suicide is at least eightfold greater than that among the general population.17 Treatment must be guided by clinical judgment. Selective serotonin reuptake inhibitor antidepressants have been recommended. Other options to manage depression include mirtazapine, monoamine oxidase inhibitors, or electroshock therapy. Mood-stabilizing agents (eg, carbamazepine, lamotrigine, valproate) may also be indicated in helping with impulse control. Haloperidol and second-generation antipsychotics are used for the treatment of a broad range of psychiatric conditions, many of which may overlap with Huntington disease, including schizophrenia and schizophreniform disorder, schizoaffective disorder, bipolar disorder, dementia, and disruptive behavior.18 The risk of tardive dyskinesia may be as much as fivefold lower with second-generation antipsychotics.18 Many patients with Huntington disease require treatment for aggression. A variety of approaches are available, including behavior modification, the antidepressant sertraline, buspirone, antipsychotic agents (eg, risperidone, olanzapine), propranolol, and lithium (combined with haloperidol).

Long-term care considerations

As a consequence of the diverse clinical manifestations of choreic disorders in movement, function, mood, and cognition, the treatment of Huntington disease requires a multidisciplinary approach that involves a number of different health care specialties across the long-term course of the disorder. Members of the Huntington disease treatment team may include neurologists, psychiatrists, nurses, social workers, geneticists, physical therapists, occupational therapists, speech therapists, dietitians, and other supporting groups or professional societies. The clinical manifestations of Huntington disease may evolve over time, as symptoms such as bradykinesia, dystonia, rigidity, cognitive decline, and gait instability become more significant.19 As a result, optimal management strategies for patients with Huntington disease may change significantly across the long-term course of the disease. During the early course of the disease, the typical clinical presentation is largely hyperkinesis, irritability, and distractibility. These patients will require initiation of drug therapy and linkage to sources of support. In the later stages of the disease, the presentation shifts to a more hypokinetic and apathetic profile, and patients are more likely to require drug regimen review and modification, nursing home placement, and palliative care services.19

Another important concern in Huntington disease treatment is care of the caregiver. Surveys show that the key concerns of caregivers include the expertise of the health care professionals who are treating the patient and the availability of sufficient services in the community.20 Several resources are available for Huntington disease caregivers, including local support groups, the Huntington’s Disease Society of America, Q Foundation, and the Huntington Study Group. The Lundbeck pharmaceutical company operates a patient assistance program (LundbeckShare. com) as well as an information center that can be accessed toll free at (888)457-4273. Approximately 90% of patients who request copayment assistance qualify for aid, regardless of the type of insurance they carry.

SUMMARY AND CONCLUSIONS

The approach to a patient with chorea starts with a search for specifically treatable etiologies. Autoimmune, metabolic, and vascular causes should be sought first and treated. The symptomatic treatment of all choreas is based on the model described here for Huntington disease, and includes attention to cognitive, psychiatric, and social support issues. The recommended approach is multidisciplinary, with a change in the mix of services as the disease progresses. It is also important to recognize the burden of Huntington disease on the caregiver and consider steps to make this burden more manageable.

Chorea is characterized by continuous, random, brief, involuntary muscle contractions that result from a variety of causes.1 These involuntary movements are nonstereotyped and irregular. Although choreic disorders are among the most common involuntary movement disorders, their diagnosis and treatment present several important challenges, including identifying and removing the cause if possible, controlling and preventing motor symptoms, and managing neuropsychologic complications.1 This article provides an overview of the diagnosis and treatment of choreic disorders, using Sydenham chorea to illustrate the management of autoimmune choreas and Huntington disease as the model for the management of heritable choreas.

Management of choreic disorders begins with a first-pass diagnosis and the use of symptomatic therapies. Even if this first pass yields no firm diagnosis, it at least rules out causes that have the most practical significance. A subsequent second-pass evaluation can be undertaken to look for rarer causes. Symptomatic therapies are continued throughout the diagnostic period. More specific therapies can be administered if an etiologic or pathogenic mechanism is determined (eg, postinfectious, autoimmune, metabolic).

CHOREIC DISORDERS: A PRACTICAL DIAGNOSTIC APPROACH

In general, choreic disorders may be subdivided into six categories:

1. Heredodegenerative disorders, such as Huntington disease and other genetically heterogeneous choreas, include Huntington disease–like 2 (HDL2) and benign hereditary chorea.1 Sporadic cases include those of unknown paternity; X-linked disorders (eg, McLeod syndrome); and autosomal-recessive disorders such as chorea-acanthocytosis, which is characterized by chorea, dystonia with prominent orofacial involvement, self-mutilation, myopathy, and neuropathy.2

2. Drug-induced choreas include neurolepticinduced tardive dyskinesia and nontardive hyperkinetic drug-related choreas, the most common of which is levodopa-induced dyskinesias.3 Tardive drug-induced choreas may occur while using the culprit drug, while tapering the drug, or after it has been discontinued. The culprit drugs are represented by dopamine-receptor blockers and include the first-generation neuroleptics (eg, phenothiazines, haloperidol), antidepressants (lox-apine), and gastrointestinal agents (metoclopramide, prochlorperazine). Drug-induced choreas are possible with a wide range of pharmacologic agents, including antiparkinsonian drugs (eg, levodopa, dopamine agonists, anticholinergics), sympathomimetics (eg, amphetamines, cocaine), anticonvulsants, calcium channel blockers, and oral contraceptives.1

3. Autoimmune choreas include Sydenham chorea, systemic lupus erythematosus, and antiphospholipid antibody syndromes. The latter encompass lupus anticoagulant and anticardiolipin antibodies.

4. Metabolic choreas are most often associated with hyperthyroidism, although case reports have described choreas in patients with vitamin B12 deficiency. A variety of hereditary metabolic diseases are also included in this category.

5. Vascular choreas include polycythemia vera and cerebrovascular accidents, the latter frequently presenting as hemiballismus. Polycythemia vera is associated with a high incidence of neurologic symptoms, including a reported incidence of chorea of 0.5% to 5%,4 and should be considered as a potential cause of chorea.

6. Other choreic disorders include a variety of entities such as rare paraneoplastic disorder/syndrome, and posttraumatic and postanoxic presentations.

The first-pass diagnostic approach includes a family history, drug history, and brain magnetic resonance imaging to identify potential structural causes of chorea. Genetic testing for Huntington disease or other choreic disorders may also be performed, although it is essential to consider the potential implications of a positive test result. Intensive pretest and posttest counseling is important both for the patient and for currently asymptomatic family members who may also be affected.1

Other testing includes:

  • Complete blood count
  • Creatine phosphokinase
  • Peripheral smear for acanthocytes
  • Comprehensive metabolic panel
  • Ceruloplasmin level
  • Measurement of thyroxine (T4) and triiodothyronine (T3)
  • B12 tests
  • Antinuclear antibody sedimentation rate
  • Lupus anticoagulant-anticardiolipin antibodies
  • Antistreptolysin O (ASO) titer
  • Anti-DNase-B titer.

GENERAL CONSIDERATIONS FOR THE TREATMENT OF CHOREIC DISORDERS

In some cases, choreic disorders have a treatable underlying etiology, such as thyroid disease or vitamin B12 deficiency. Tardive syndromes may require treatment beyond drug discontinuation, including use of dopamine depleters for the classic tardive dyskinetic syndromes and anticholinergics for the tardive dystonic syndromes. Levodopa dyskinesia may be treated using amantadine, clozapine, or deep brain stimulation.5,6 The treatment of patients with autoimmune choreas is not well defined. It may include anticoagulation in patients with positive anticardiolipin antibodies to prevent venous or arterial thromboembolism,7 but the risk of arterial thromboembolism is uncertain, and it is unclear whether chorea is truly a harbinger of vascular events.

A negative ASO titer does not exclude Sydenham chorea, a result of childhood infection with group-A beta-hemolytic streptococcus, and antibiotics should be considered in the appropriate context. Some researchers have argued that immune responses associated with acute infections may result in autoimmune neuropsychiatric symptoms. In pediatric patients, this has been referred to as pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections (PANDAS).8

A related phenomenon has been proposed as a potential mechanism of some types of chorea, although the relationship between acute infection and chorea is controversial. Patients with elevated ASO titer or anti-DNase-B titers may be candidates for antibiotics. By 6 weeks after the onset of infection, these titers will fall and a diagnosis of Sydenham chorea can be postulated or based exclusively on clinical judgment.

 

 

SYDENHAM CHOREA

Unique to Sydenham chorea is the use of penicillin as prophylaxis. Other than that, the management of Sydenham chorea exemplifies the management approach for the larger category of autoimmune choreic disorders. Pathogenic-based treatment options include immune modulation with cortico steroids, intravenous immunoglobulin (IVIG), and plasma exchange; all treatments must be administered in the appropriate clinical context.

One double-blind clinical trial examined the effectiveness of corticosteroid treatment in children with Sydenham chorea randomly assigned to receive either prednisone (n = 22) or placebo (n = 15).9 Prednisone was administered at a dose of 2 mg/kg/day for 4 weeks, followed by gradual tapering and discontinuation. The median time to remission of chorea was significantly lower for patients in the prednisone group (54.3 days) compared with those in the placebo group (119.9 days; P < .001). Patients in the prednisone group also exhibited significantly better scores on a chorea intensity rating scale at 8 weeks and 12 weeks (P < .001). Potential limitations of this approach include relapse of chorea symptoms and corticosteroid-related adverse events (eg, Cushing syndrome, hypertension).

A second study compared the effectiveness of three modalities: IVIG at a dose of 1 g/kg/day for 2 days (n = 4), plasma exchange (n = 8), and prednisone (n = 6).10 Although differences between treatment groups were not statistically significant, the authors noted that the clinical improvement in chorea symptoms tended to be greater for patients who received IVIG or plasma exchange than for those who received prednisone. Mean chorea scores improved from baseline by 72% for the IVIG group, 50% for the plasma exchange group, and 29% for the prednisone group.

After etiology-dependent treatments have been considered, several other options may be effective regardless of the specific etiology. These include symptomatic treatments such as haloperidol, atypical neuroleptics, and amantadine.11 Antiepileptic medications or benzodiazepines may also help to control symptoms, although less information is available about the use of these agents for the treatment of Sydenham chorea. Tetrabenazine may be considered for patients who will require long-term treatment.

HUNTINGTON DISEASE

Pharmacotherapy of Huntington disease may be unnecessary if symptoms are mild or not bothersome. Symptomatic treatment options include tetrabenazine, amantadine, and either first-generation neuroleptics (eg, haloperidol) or second-generation atypical neuroleptics (eg, olanzapine, quetiapine, risperidone, ziprasidone).

Treating choreas with tetrabenazine or amantadine

Considerable recent attention has focused on the efficacy and safety of tetrabenazine for the treatment of Huntington disease and other choreic disorders. Tetrabenazine is a central monoamine depleter that reversibly binds to the type-2 vesicular monoamine transporter.12 The TETRA-HD study examined the efficacy and safety of tetrabenazine for the short- and long-term control of Huntington disease.12 An initial study compared tetrabenazine with placebo in 75 patients who were treated for up to 13 weeks. In an extension study, all patients received individualized tetrabenazine doses for up to 80 weeks.

Figure 1. Mean total maximal chorea scores decreased markedly during the first 10 weeks of tetrabenazine treatment for Huntington disease, remained below baseline through 80 weeks of treatment, and then returned to baseline after tetrabenazine discontinuation.12

The mean total maximal chorea (TMC) scores from the Unified Huntington Disease Rating Scale (UHDRS) decreased markedly during the first 10 weeks of tetrabenazine treatment, remained lower than baseline throughout 80 weeks, and then returned to baseline levels after tetrabenazine discontinuation (Figure 1). At week 80, the mean TMC score was reduced by 4.6 UHDRS units compared with baseline (P < .001) The long-term extension phase was completed by 45 of 75 patients. Treatment-related adverse events that prompted discontinuation included depression, delusions, and vocal tics. The most commonly reported adverse events included sedation or somnolence (n = 18), depressed mood (n = 17), anxiety (n = 13), insomnia (n = 10), and akathisia (n = 9). Scores of parkinsonism and dysphagia increased significantly from baseline over the 80-week study.

Amantadine is an option for patients who cannot tolerate tetrabenazine. A double-blind, placebo-controlled study performed by researchers at the National Institutes of Health (NIH) examined the efficacy and safety of amantadine in 24 patients with Huntington disease.13 Patients were treated with oral amantadine 400 mg/day or placebo for 2 weeks, and were then crossed over to the other treatment. Amantadine was associated with a median reduction in extremity chorea score at rest of 36% from baseline (P = .04), versus 0% improvement with placebo. The mean improvement with amantadine was 56% for the 10 patients with the highest drug plasma levels.

Improvement in chorea scores from baseline for amantadine compared with placebo was rated with four different methods: (1) maximal chorea severity measured from video recordings; (2) maximal chorea severity measured by live raters; (3), chorea severity at rest measured from video recordings; and (4) extremity chorea at rest measured from video recordings. Amantadine was superior to placebo according to all four rating methods. Treatment was generally safe and well tolerated, and no consistent changes in cognitive function were noted with amantadine therapy.

Reprinted with permission from Neurology (Lucetti C, et al. IV amantadine improves chorea in Huntington’s disease: an acute randomized, controlled study. Neurology 2003; 60:1995–1997). Copyright © 2003 by AAN Enterprises, Inc.
Figure 2. During the randomized placebo-controlled phase of a study that compared amantadine with placebo, mean dyskinesia scores, measured using the Abnormal Involuntary Movement Scale scores, decreased significantly 90 minutes after initiation of amantadine infusion compared with placebo.14aP < .05
A second study examined the effects of amantadine as a 2-hour IV infusion in nine patients with Huntington disease.14 Amantadine or placebo was administered in a randomized, double-blind manner on the first day of the study, and patients were then crossed over to the other treatment on the second day. All patients then received open-label oral amantadine for an additional 1 year. During the randomized placebo-controlled phase, mean dyskinesia scores, evaluated using the Abnormal Involuntary Movement Scale, were significantly lower for patients randomly assigned to amantadine compared with placebo. During the randomized placebo-controlled phase, the decrease in mean dyskinesia score was significantly greater 90 minutes after treatment with amantadine compared with placebo (Figure 2). In the open-label amantadine continuation phase, oral amantadine was associated with a further gradual improvement in symptoms over 3 to 6 months. No significant changes were observed in neuropsychologic tests or psychiatric rating scales.

 

 

Managing nonmotor complications

In addition to addressing chorea, it is also important to manage nonmotor complications of Huntington disease, including cognition, mood, and thought disorders. Rivastigmine was assessed for the treatment of motor symptoms, functional disability, and cognitive impairment associated with Huntington disease in an open-label study of 18 patients; 11 received rivastigmine 6 mg/day and 7 control patients did not.15 Motor and cognitive function were assessed for up to 2 years by raters who were blinded to treatment assignment. Ratings on a global motor performance scale were significantly better for patients who received rivastigmine than for control subjects. Rivastigmine treatment was also associated with trends toward improvements in functional disability and cognitive impairment, although these differences were not statistically significant.

A small open-label study examined the effects of donepezil for movement and cognitive symptoms associated with Huntington disease.16 Donepezil did not significantly improve cognitive symptoms, although the study enrolled only eight patients. All patients tolerated oral donepezil at a dose of 5 mg/day, but four patients withdrew from the study when the dose was increased to 10 mg/day. In two patients, chorea worsened and falls increased, moderate to severe diarrhea developed in three patients, and one patient reported anxiety and irritability.

Depression is another common complication of Huntington disease. The incidence of depression among patients with Huntington disease is approximately 40%, and the risk of suicide is at least eightfold greater than that among the general population.17 Treatment must be guided by clinical judgment. Selective serotonin reuptake inhibitor antidepressants have been recommended. Other options to manage depression include mirtazapine, monoamine oxidase inhibitors, or electroshock therapy. Mood-stabilizing agents (eg, carbamazepine, lamotrigine, valproate) may also be indicated in helping with impulse control. Haloperidol and second-generation antipsychotics are used for the treatment of a broad range of psychiatric conditions, many of which may overlap with Huntington disease, including schizophrenia and schizophreniform disorder, schizoaffective disorder, bipolar disorder, dementia, and disruptive behavior.18 The risk of tardive dyskinesia may be as much as fivefold lower with second-generation antipsychotics.18 Many patients with Huntington disease require treatment for aggression. A variety of approaches are available, including behavior modification, the antidepressant sertraline, buspirone, antipsychotic agents (eg, risperidone, olanzapine), propranolol, and lithium (combined with haloperidol).

Long-term care considerations

As a consequence of the diverse clinical manifestations of choreic disorders in movement, function, mood, and cognition, the treatment of Huntington disease requires a multidisciplinary approach that involves a number of different health care specialties across the long-term course of the disorder. Members of the Huntington disease treatment team may include neurologists, psychiatrists, nurses, social workers, geneticists, physical therapists, occupational therapists, speech therapists, dietitians, and other supporting groups or professional societies. The clinical manifestations of Huntington disease may evolve over time, as symptoms such as bradykinesia, dystonia, rigidity, cognitive decline, and gait instability become more significant.19 As a result, optimal management strategies for patients with Huntington disease may change significantly across the long-term course of the disease. During the early course of the disease, the typical clinical presentation is largely hyperkinesis, irritability, and distractibility. These patients will require initiation of drug therapy and linkage to sources of support. In the later stages of the disease, the presentation shifts to a more hypokinetic and apathetic profile, and patients are more likely to require drug regimen review and modification, nursing home placement, and palliative care services.19

Another important concern in Huntington disease treatment is care of the caregiver. Surveys show that the key concerns of caregivers include the expertise of the health care professionals who are treating the patient and the availability of sufficient services in the community.20 Several resources are available for Huntington disease caregivers, including local support groups, the Huntington’s Disease Society of America, Q Foundation, and the Huntington Study Group. The Lundbeck pharmaceutical company operates a patient assistance program (LundbeckShare. com) as well as an information center that can be accessed toll free at (888)457-4273. Approximately 90% of patients who request copayment assistance qualify for aid, regardless of the type of insurance they carry.

SUMMARY AND CONCLUSIONS

The approach to a patient with chorea starts with a search for specifically treatable etiologies. Autoimmune, metabolic, and vascular causes should be sought first and treated. The symptomatic treatment of all choreas is based on the model described here for Huntington disease, and includes attention to cognitive, psychiatric, and social support issues. The recommended approach is multidisciplinary, with a change in the mix of services as the disease progresses. It is also important to recognize the burden of Huntington disease on the caregiver and consider steps to make this burden more manageable.

References
  1. Cardoso F, Seppi K, Mair KJ, Wenning GK, Poewe W. Seminar on choreas. Lancet Neurol 2006; 5:589602.
  2. Schneider SA, Walker RH, Bhatia KP. The Huntington’s disease-like syndromes: what to consider in patients with a negative Huntington’s disease gene test. Nat Clin Pract Neurol 2007; 3:517525.
  3. Zesiewicz TA, Sullivan KL. Drug-induced hyperkinetic movement disorders by nonneuroleptic agents. Handb Clin Neurol 2011; 100:347363.
  4. Margolis RL, Ross CA. Diagnosis of Huntington disease. Clin Chem 2003; 49:17261732.
  5. Rascol O. The pharmacological therapeutic management of levodopa-induced dyskinesias in patients with Parkinson’s disease. J Neurol 2000; 247 (suppl 2):II51II57.
  6. Fabbrini G, Brotchie JM, Grandas F, Nomoto M, Goetz CG. Levodopa-induced dyskinesias. Mov Disord 2007; 22:13791389.
  7. Lim W, Crowther MA, Eifelboom JW. Management of antiphospholipid antibody syndrome: a systematic review. JAMA 2006; 295:10501057.
  8. Mink J, Kurlan R. Acute postinfectious movement and psychiatric disorders in children and adolescents. J Child Neurol 2011; 26:214217.
  9. Paz JA, Silva CA, Marques-Dias MJ. Randomized double-blind study with prednisone in Sydenham’s chorea. Pediatr Neurol 2006; 34:264269.
  10. Garvey MA, Snider LA, Leitman SF, Werden R, Swedo SE. Treatment of Sydenham’s chorea with intravenous immunoglobulin, plasma exchange, or prednisone. J Child Neurol 2005; 20:424429.
  11. Walker KG, Wilmshurst JM. An update on the treatment of Sydenham’s chorea: the evidence for established and evolving interventions. Ther Adv Neurol Disord 2010; 3:301309.
  12. Frank S. Tetrabenazine as anti-chorea therapy in Huntington disease: an open-label continuation study. Huntington Study Group/TETRA-HD Investigators. BMC Neurol 2009; 9:62.
  13. Verhagen Metman L, Morris MJ, Farmer C, et al. Huntington’s disease: a randomized, controlled trial using the NMDA-antagonist amantadine. Neurology 2002; 59:694699.
  14. Lucetti C, Del Dotto P, Gamabaccini G, et al. IV amantadine improves chorea in Huntington’s disease: an acute randomized, controlled study. Neurology 2003; 60:19951997.
  15. de Tommaso M, Difruscolo O, Scirruicchio V, Specchio N, Livrea P. Two years’ follow-up of rivastigmine treatment in Huntington disease. Clin Neuropharmacol 2007; 30:4346.
  16. Fernandez HH, Friedman JH, Grace J, Beasin-Hazen S. Donepezil for Huntington’s disease. Mov Disord 2000; 15:173176.
  17. Schoenfeld M, Myers RH, Cupples LA, Berkman B, Sax DS, Clark E. Increased rate of suicide among patients with Huntington’s disease. J Neurol Neurosurg Psychiatry 1984; 47:12831287.
  18. Correll CU, Leucht S, Kane JM. Lower risk for tardive dyskinesia associated with second-generation antipsychotics: a systematic review of 1-year studies. Am J Psychiatry 2004; 161:414425.
  19. Phillips W, Shannon KM, Barker RA. The current clinical management of Huntington’s disease. Mov Disord 2008; 23:14911504.
  20. Skirton H, Williams JK, Jackson Barnette J, Paulsen JS. Huntington disease: families’ experiences of healthcare services. J Adv Nursing 2010; 66:500510.
References
  1. Cardoso F, Seppi K, Mair KJ, Wenning GK, Poewe W. Seminar on choreas. Lancet Neurol 2006; 5:589602.
  2. Schneider SA, Walker RH, Bhatia KP. The Huntington’s disease-like syndromes: what to consider in patients with a negative Huntington’s disease gene test. Nat Clin Pract Neurol 2007; 3:517525.
  3. Zesiewicz TA, Sullivan KL. Drug-induced hyperkinetic movement disorders by nonneuroleptic agents. Handb Clin Neurol 2011; 100:347363.
  4. Margolis RL, Ross CA. Diagnosis of Huntington disease. Clin Chem 2003; 49:17261732.
  5. Rascol O. The pharmacological therapeutic management of levodopa-induced dyskinesias in patients with Parkinson’s disease. J Neurol 2000; 247 (suppl 2):II51II57.
  6. Fabbrini G, Brotchie JM, Grandas F, Nomoto M, Goetz CG. Levodopa-induced dyskinesias. Mov Disord 2007; 22:13791389.
  7. Lim W, Crowther MA, Eifelboom JW. Management of antiphospholipid antibody syndrome: a systematic review. JAMA 2006; 295:10501057.
  8. Mink J, Kurlan R. Acute postinfectious movement and psychiatric disorders in children and adolescents. J Child Neurol 2011; 26:214217.
  9. Paz JA, Silva CA, Marques-Dias MJ. Randomized double-blind study with prednisone in Sydenham’s chorea. Pediatr Neurol 2006; 34:264269.
  10. Garvey MA, Snider LA, Leitman SF, Werden R, Swedo SE. Treatment of Sydenham’s chorea with intravenous immunoglobulin, plasma exchange, or prednisone. J Child Neurol 2005; 20:424429.
  11. Walker KG, Wilmshurst JM. An update on the treatment of Sydenham’s chorea: the evidence for established and evolving interventions. Ther Adv Neurol Disord 2010; 3:301309.
  12. Frank S. Tetrabenazine as anti-chorea therapy in Huntington disease: an open-label continuation study. Huntington Study Group/TETRA-HD Investigators. BMC Neurol 2009; 9:62.
  13. Verhagen Metman L, Morris MJ, Farmer C, et al. Huntington’s disease: a randomized, controlled trial using the NMDA-antagonist amantadine. Neurology 2002; 59:694699.
  14. Lucetti C, Del Dotto P, Gamabaccini G, et al. IV amantadine improves chorea in Huntington’s disease: an acute randomized, controlled study. Neurology 2003; 60:19951997.
  15. de Tommaso M, Difruscolo O, Scirruicchio V, Specchio N, Livrea P. Two years’ follow-up of rivastigmine treatment in Huntington disease. Clin Neuropharmacol 2007; 30:4346.
  16. Fernandez HH, Friedman JH, Grace J, Beasin-Hazen S. Donepezil for Huntington’s disease. Mov Disord 2000; 15:173176.
  17. Schoenfeld M, Myers RH, Cupples LA, Berkman B, Sax DS, Clark E. Increased rate of suicide among patients with Huntington’s disease. J Neurol Neurosurg Psychiatry 1984; 47:12831287.
  18. Correll CU, Leucht S, Kane JM. Lower risk for tardive dyskinesia associated with second-generation antipsychotics: a systematic review of 1-year studies. Am J Psychiatry 2004; 161:414425.
  19. Phillips W, Shannon KM, Barker RA. The current clinical management of Huntington’s disease. Mov Disord 2008; 23:14911504.
  20. Skirton H, Williams JK, Jackson Barnette J, Paulsen JS. Huntington disease: families’ experiences of healthcare services. J Adv Nursing 2010; 66:500510.
Page Number
S30-S34
Page Number
S30-S34
Publications
Publications
Article Type
Display Headline
Comprehensive treatment of Huntington disease and other choreic disorders
Display Headline
Comprehensive treatment of Huntington disease and other choreic disorders
Citation Override
Cleveland Clinic Journal of Medicine 2012 July;79(suppl 2):S30-S34
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media

Tics and Tourette syndrome: An adult perspective

Article Type
Changed
Mon, 10/01/2018 - 15:36
Display Headline
Tics and Tourette syndrome: An adult perspective

Tourette syndrome (TS) is part of a spectrum of tic disorders. Tics are sudden, rapid, stereotyped, repetitive, nonrhythmic movements or vocalizations affecting discrete muscle groups, and are preceded by a sensory component. Patients in whom tic suppression is attempted report the experience of a sensation of inner pressure that must be released. This eventually results in the performance of motor movement or vocal sounds. TS is a disorder of childhood onset that is characterized by multiple motor and vocal tics. In some cases, there are features of obsessive compulsive disorder (OCD), attention deficit hyperactivity disorder (ADHD), or other behavioral manifestations such as coprolalia, echopraxia, palilalia, and self-injury.1,2 The spectrum of tic disorders includes:

  • Transient tics of childhood (tic duration less than 12 months)
  • Chronic motor or vocal tics (lasting more than 12 months), and
  • TS (variable motor and vocal tics lasting more than 12 months).

Many children meet the diagnostic criteria for TS between the ages of 6 and 9 years, but symptoms may improve by adulthood. The eventual loss of tics over time reflects the maturation of brain systems that control ballistic action.3

The tics that accompany TS may be defined as simple or complex, and as motor or vocal. Simple motor tics involve only a few muscles, such as eye blinking, shoulder shrugging, or facial grimacing. Complex motor tics involve multiple groups of muscles that are recruited in orchestrated bouts (eg, hand gestures, jumping, touching, or pressing), and may include copropraxia (a sudden tic-like vulgar, sexual, or obscene gesture) or echopraxia (involuntary, spontaneous imitation of someone else’s movements). Simple vocal tics are meaningless sounds such as throat clearing, grunting, sniffing, snorting, and chirping. Complex vocal tics involve speech and language such as sudden, spontaneous expression of single words or phrases, or speech blocking.4

Tics may be acquired as a consequence of other disorders, including head trauma, encephalitis, stroke, carbon monoxide poisoning, Creutzfeldt-Jakob disease, neurosyphilis, hypoglycemia, or Sydenham chorea.5 Genetic disorders such as Huntington disease may be associated with tics. Tics may also occur with certain chromosomal abnormalities or be associated with some neuropsychiatric disorders. Finally, tics may be caused by a large number of medications or illicit drugs, including cocaine, amphetamines, antipsychotics, and antidepressants.

The prevalence of all types of tics in childhood is approximately 6% to 12%, although the prevalence of chronic vocal tics is approximately 1 to 10 per 1,000 children and adolescents.6 TS is especially common among autistic children and in those with Asperger syndrome and other autistic spectrum disorders. A survey of patients at Cleveland Clinic Florida found that tics and TS accounted for 8% of all patients with movement disorders. Of patients with tics or TS who were older than 18 years, 70% were male.

PATHOPHYSIOLOGY OF TOURETTE SYNDROME: ROLE OF THE BASAL GANGLIA

Although the pathophysiology of TS is not completely understood, abnormal function of the basal ganglia is thought to be a central component of the disorder. The basal ganglia normally act to facilitate voluntary movements while suppressing competing involuntary ones. Abnormalities of basal ganglia activity are important in several disorders of motor function.7 Output neurons from the basal ganglia inhibit thalamic motor nuclei and midbrain neurons of the extrapyramidal motor system, and act to inhibit motor pattern generators in the cerebral cortex and brainstem. Hyperkinetic disorders, including tics, chorea, and dystonia, are thought to result at least in part from impaired inhibition of unwanted motor activity from the basal ganglia to downstream motor centers.7

Family heritability studies provide strong support that TS is a genetic disorder. For example, the concordance rate is 86% for monozygotic twins versus 20% for dizygotic twins.8 Chromosomes linked to TS include 2p32.2 and 13q31.1.9,10 Interactions between genetics and environment are also thought to play a significant role. The concept of pediatric autoimmune neuropsychiatric disorders associated with streptococcal (PANDAS) infections has been proposed to explain an apparent temporal association between streptococcal infections and exacerbation of tics. According to this model, molecular mimicry between streptococcal antigens and endogenous brain antigens results in an autoimmune attack.11 However, the identification of specific antibodies against basal ganglia cells remains controversial.12

MANAGEMENT OVERVIEW

Accurate diagnosis of TS is essential, and includes a complete history and neurologic examination. The tic phenomenology (complex vs simple) should be characterized, and the patient should be carefully questioned to identify the symptoms that are most bothersome (eg, motor or vocal tics, OCD, or ADHD). Pharmacotherapy should be reserved for problems that are functionally disabling and not remediable by nonpharmacologic interventions.

Treatment may also be required for other neuropsychiatric symptoms. Anxiety and depression have been reported in 19% to 80% of patients with tics, and depression is strongly correlated with the duration and severity of tics.13,14 Episodic outburst (rage), self-injurious, OCD, antisocial, and oppositional behaviors are all more common among individuals with tic disorders.15 Personality disorders may be related to OCD, ADHD, or to family or economic issues. Tic disorders are also associated with an increased incidence of somatic complaints, as well as higher rates of academic difficulties, which may be related to ADHD or medications. Sleep disturbances affect an estimated 20% to 50% of patients, and may include difficulty initiating or maintaining sleep, restlessness, movement-related arousal, or parasomnia.16

Education is an important part of treatment, and may include the patient, family members, teachers or other school staff, and work colleagues. A number of behavioral or psychosocial approaches may help to improve tics, including conditioning techniques, relaxation training, biofeedback, habit reversal, awareness training, and hypnosis.17

 

 

PHARMACOLOGIC TREATMENT: THREE TIERS

Options for the pharmacologic treatment of tics and TS include dopamine blockers, dopamine depleters, benzodiazepines, central alpha-adrenergic blockers, and botulinum toxin. Pharmacotherapy options can be divided into three tiers (Table), with first-tier drugs considered first-choice treatments.

First-tier therapies

The alpha-adrenergic blockers clonidine and guanfacine are first-tier therapies. Treatment should be initiated at a low dose and escalated gradually according to response, which is determined by the severity, and not the presence, of tics. Clonidine may be administered at a dose of 0.025 mg two or three times daily or, for maintenance, 0.1 mg three times daily; another option is 0.1, 0.2, or 0.3 mg weekly by transdermal administration. Guanfacine may be administered at a dose of 1 mg once daily. Alpha-adrenergic blockers are useful for the treatment of mild tics, and are considered first-line therapy for tic suppression. Side effects may include dry mouth, somnolence, and, rarely, blood pressure fluctuations.

Agents that affect gamma-amino butyric acid (GABA) neurotransmission have been associated with improved symptoms of tic disorders.18 For example, both clonazepam and diazepam have been reported to reduce TS symptoms.18 Both of these benzodiazepines are associated with sedation, blunting of cognition, and exacerbation of depression, however.19,20

Second-tier therapies

Second-tier therapies, consisting of neuroleptics, induce a rapid treatment response. Haloperidol may be started at a dose of 0.25 mg once daily, with a maintenance dosage of 0.5 to 3.0 mg/day. Cognitive blunting or extrapyramidal side effects are rare in patients with TS, but the potential for these side effects should be thoroughly discussed with the patient or parent/guardian before treatment. Pimozide 0.5 mg (2 to 6 mg/day for maintenance) may be associated with tremor or parkinsonian symptoms (predominantly akinesia). Risperidone 0.25 mg/day (0.5 to 4 mg/day for maintenance), olanzapine 2.5 mg/day (5 to 10 mg/day for maintenance), and quetiapine 25 mg twice daily (100 to 300 mg/day for maintenance) are associated with potential adverse effects of extrapyramidal symptoms, weight gain, and diabetes.

Third-tier therapies

Dopamine agonists (reserpine and tetrabenazine) and botulinum toxin are third-tier therapies. Reserpine, although rarely used in current clinical practice, may be administered at doses of 0.1 to 0.25 mg/day, titrating upward on the basis of clinical response. Tetrabenazine may be administered at a starting dose of 12.5 mg/day, with higher doses as needed depending on the response to treatment. Adverse effects include hypotension, sedation, extrapyramidal symptoms (predominantly parkinsonism), and depression.21

The exact mechanism by which tetrabenazine produces this suppression effect is unknown, but it is believed to be related to its effect of reversibly depleting monoamines. At least three neuronal protein classes regulate the effects of dopamine on voluntary and involuntary movement.22 The two presynaptic proteins are vesicular monoamine transporter subtype 2 (VMAT2) and dopamine transporter (DAT). Postsynaptically, dopamine activity is regulated by G-protein–linked dopamine receptors (eg, the D2 receptor). Tetrabenazine reduces the uptake of monoamines (including dopamine) into synaptic vesicles by reversibly binding to VMAT2, resulting in degradation of dopamine within axon terminals by monoamine oxidases.23 By blocking dopamine transport, tetrabenazine depletes dopamine with greater selectivity than it does other monoamines.24

The dosage of tetrabenazine for the treatment of motor disorders, particularly chorea, was established in the Huntington Study Group (HSG) clinical trial.25 In the HSG trial, a starting dose of 12.5 mg on day 1 was increased to 12.5 mg twice daily on days 2 to 7, and then by 12.5 mg/day at weekly intervals until the desired clinical effect, intolerable adverse effects, or a maximum dose of 100 mg/day was reached. Daily dosages of 37.5 mg or more are administered in three divided doses. Adverse events (reported in 70% of patients who received placebo and 91% of patients who received tetrabenazine) include sedation or somnolence, insomnia, and fatigue.21 These findings may be carried over to patients with tics.

Botulinum toxin may also help to control tics—especially dystonic tics. The premonitory symptoms of TS are usually unaffected by botulinum toxin.26 The adverse effect profile for patients with TS is similar to that of patients with dystonia or facial dyskinesia, and may include soreness, transient weakness, ptosis (if injected for eye blinking), and mild transient dysphagia (if injected into the larynx).27

MANAGING COMORBID CONDITIONS

Approximately 30% of patients with TS also have OCD.28 Treatment options include selective serotonin reuptake inhibitors at standard doses, and the tricyclic antidepressant clomipramine (25 mg once or twice daily, or 75 mg/day in sustained-release form). Trazodone, a serotonin antagonist and reuptake inhibitor that is associated with a lower incidence of anticholinergic effects, may be initiated at a dose of 50 mg/day and slowly increased to 150 to 400 mg/day depending on clinical response.

As many as 60% of patients with TS may also have ADHD.28 Methylphenidate is helpful for the treatment of ADHD and does not exacerbate tics, but it is a restricted medication. The recommended dose is 20 mg once daily, titrated upward as needed based on response. Atomoxetine carries a warning regarding increased risk of suicide. It has also been associated with an increased risk of sexual dysfunction and behavioral changes, including aggressive behaviors, agitation, and irratibility.29

 

 

DEEP BRAIN STIMULATION

Deep brain stimulation (DBS) has been shown to improve TS in single-case studies and in small series, although the long-term benefit is unclear. Potential targets of stimulation include midline thalamic centromedian-parafascicular (CM-PF) nuclei, the ventralis oralis complex of the thalamus, motor and limbic globus pallidus pars interna (GPi), and the anterior limb of the internal capsule.30 In particular, stimulation of the sensorimotor GPi may ameliorate hyperkinetic states.

One report described the results of DBS implantation in a 15-year-old boy with TS who had not responded to several pharmacologic treatment options.31 Six months after implantation, the patient exhibited markedly improved tic severity as measured using the Yale Global Tic Severity Scale, including a 76% reduction in motor tic severity, 68% reduction in vocal tics, and a complete resolution of impairment.31

Published consensus criteria for the selection of suitable candidates for DBS include age greater than 25 years, chronic and severe tics with severe functional impairment for at least 12 months, tics that are frequent and noticeable in most situations most of the time, failure of conventional medical therapy, medical stability for 6 months, and willingness to participate in ongoing psychologic interventions.32 Exclusion criteria include the presence of another medical condition that could explain the tics, an unstable medical condition, being considered likely to benefit from psychologic interventions, psychosocial factors that may complicate the recovery process or make it difficult to assess outcome, and unwillingness to participate in ongoing treatment for psychosocial problems or risk factors. Other factors that should be considered include comorbidities, the variability in tic severity over time, the involvement of a multidisciplinary treatment team, results of a thorough neuropsychologic assessment, expertise of the surgical team, and access to imaging facilities for presurgical mapping and postsurgical evaluation.

SUMMARY AND CONCLUSIONS

Tourette syndrome is not uncommon among the adult population of a typical neurology practice, and should not be considered exclusively a pediatric diagnosis. Several treatment options are available, including behavioral approaches and several medications. Treatment should focus on the most disabling symptoms. Neuropsychologic assessment and psychiatric support may be necessary for some patients. The same comorbidities that are encountered in children are usually evident in adult patients as well. In medically refractory cases, DBS surgery may be helpful.

References
  1. Robertson MM. Annotation: Gilles de la Tourette syndrome—an update. J Child Psychol Psychiatry 1994; 35:597611.
  2. Robertson MM, Althoff RR, Hafez A, Pauls DL. Principal components analysis of a large cohort with Tourette syndrome. Br J Psychiatry 2008; 193:3136.
  3. Flaherty AW. Movement disorders. In: Stern TA, Rosenbaum JF, Fava M, Biederman J, Rauch SL, eds. Massachusetts General Hospital: Comprehensive Clinical Psychiatry E-Book. Philadelphia, PA: Mosby Elsevier; 2008.
  4. Müller N. Tourette’s syndrome: clinical features, pathophysiology, and therapeutic approaches. Dialogues Clin Neurosci 2007; 9:161171.
  5. Bagheri MM, Kerbeshian J, Burd L. Recognition and management of Tourette’s syndrome and tic disorders. Am Fam Physician 1999; 59:22632272,2274.
  6. Lombroso PJ, Scahill L. Tourette syndrome and obsessive–compulsive disorder [published online ahead of print October 15, 2007]. Brain Dev 2008; 30:231237. 10.1016/j.braindev.2007.09.001
  7. Mink JW. The basal ganglia and involuntary movements: impaired inhibition of competing motor patterns. Arch Neurol 2003; 60:13651368.
  8. Singer HS, Smith-Hicks C, Lieberman D. Tourette syndrome. In: LeDoux M, ed. Animal Models of Movement Disorders. Academic Press; 2005.
  9. Tourette Syndrome Association International Consortium for Genetics. Genome scan for Tourette disorder in affected-sibling-pair and multigenerational families. Am J Hum Genet 2007; 80:265272.
  10. Abelson JF, Kwan KY, O’Roak BJ, et al. Sequence variants in SLITRK1 are associated with Tourette’s syndrome. Science 2005; 310:317320.
  11. Kurlan R. Tourette’s syndrome and ‘PANDAS’: will the relation bear out? Pediatric autoimmune neuropsychiatric disorders associated with streptococcal infection. Neurology 1998; 50:15301534.
  12. Morris CM, Pardo-Villamizar C, Gause CD, Singer HS. Serum autoantibodies measured by immunofluorescence confirm a failure to differentiate PANDAS and Tourette syndrome from controls [published online ahead of print September 27, 2008]. J Neurol Sci 2009; 276:4548. 10.1016/j.jns.2008.08.032
  13. Comings BG, Comings DE. A controlled study of Tourette syndrome. V. Depression and mania. Am J Hum Genet 1987; 41:804821.
  14. Robertson MM, Williamson F, Eapen V. Depressive symptomatology in young people with Gilles de la Tourette syndrome—a comparison of self-report scales [published online ahead of print February 7, 2006]. J Affect Disord 2006; 91:265268. 10.1016/j.jad.2005.12.046
  15. Budman CL, Bruun RD, Park KS, Lesser M, Olson M. Explosive outbursts in children with Tourette’s disorder. J Am Acad Child Adolesc Psychiatry 2000; 39:12701276.
  16. Kostanecka-Endress T, Banaschewski T, Kinkelbur J, et al. Disturbed sleep in children with Tourette syndrome: a polysomnographic study. J Psychosom Res 2003; 55:2329.
  17. Peterson AL. Psychosocial management of tics and intentional repetitive behaviors associated with Tourette syndrome. In:Woods DW, Piacentini J, Walkup JT, eds. Treating Tourette Syndrome and Tic Disorders: A Guide for Practitioners. Guilford Press; 2007.
  18. Robertson MM. Tourette syndrome, associated conditions and the complexities of treatment. Brain 2000; 123:425462.
  19. Klonopin [package insert]. South San Francisco, CA: Genentech USA, Inc.; 2010.
  20. Valium [package insert]. Nutley, NJ: Roche Laboratories, Inc.; 2008.
  21. Xenazine [package insert]. Deerfield, IL: Lundbeck Inc.; 2011.
  22. Schmitz Y, Benoit-Marand M, Gonon F, Sulzer D. Presynaptic regulation of dopaminergic neurotransmission. J Neurochem 2003; 87:273289.
  23. Morrow T. Gene therapy offers HD patients relief from some symptoms. Tetrabenazine inhibits the transport of a molecule called vesicular monoamine transporter type 2 or VMAT2. Manag Care 2008; 17:4647.
  24. Pearson SJ, Reynolds GP. Depletion of monoamine transmitters by tetrabenazine in brain tissue in Huntington’s disease. Neuropharmacology 1988; 27:717719.
  25. Huntington Study Group. Tetrabenazine as antichorea therapy in Huntington disease: a randomized controlled trial. Neurology 2006; 66:366372.
  26. Jankovic J, Kurlan R. Tourette syndrome: evolving concepts [published online ahead of print April 11, 2011]. Mov Disord 2011; 26:11491156. 10.1002/mds.23618
  27. Swain JE, Leckman JF. Tourette syndrome and tic disorders: overview and practical guide to diagnosis and treatment. Psychiatry (Edgmont) 2005; 2:2636.
  28. Gorman DA, Thompson N, Plessen KJ, Robertson MM, Leckman JF, Peterson BS. Psychosocial outcome and psychiatric comorbidity in older adolescents with Tourette syndrome: controlled study. Br J Psychiatry 2010; 197:3644.
  29. Strattera [package insert]. Indianapolis, IN: Eli Lilly and Company; 2011.
  30. Ackermans L, Temel Y, Visser-Vandewalle V. Deep brain stimulation in Tourette’s syndrome. Neurotherapeutics 2008; 5:339344.
  31. Shahed J, Poysky J, Kenney C, Simpson R, Jankovic J. GPi deep brain stimulation for Tourette syndrome improves tics and psychiatric comorbidities. Neurology 2007; 68:159160.
  32. Mink JW, Walkup J, Frey KA, et al .Patient selection and assessment recommendations for deep brain stimulation in Tourette syndrome. Mov Disord 2006; 21:18311838.
Article PDF
Author and Disclosure Information

Nestor Galvez-Jimenez, MD, MSc, MS(HSA), FACP
Pauline M. Braathen Endowed Chair in Movement Disorders, Chairman, Department of Neurology, Chief, Movement Disorders Section, Cleveland Clinic Florida, Weston, FL

Correspondence: Nestor Galvez-Jimenez, MD, Cleveland Clinic Florida, 2950 Cleveland Clinic Boulevard, Weston, FL 33331; galvezn@ccf.org

Dr. Galvez-Jimenez reported teaching and speaking services for Allergan, Inc. and Lundbeck.

This article is based on Dr. Galvez-Jimenez’s presentation at “The Annual Therapy Symposium on Movement Disorders for the Modern Clinician” held in Fort Lauderdale, Florida, on January 29, 2011. The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Galvez-Jimenez.

Publications
Page Number
S35-S39
Author and Disclosure Information

Nestor Galvez-Jimenez, MD, MSc, MS(HSA), FACP
Pauline M. Braathen Endowed Chair in Movement Disorders, Chairman, Department of Neurology, Chief, Movement Disorders Section, Cleveland Clinic Florida, Weston, FL

Correspondence: Nestor Galvez-Jimenez, MD, Cleveland Clinic Florida, 2950 Cleveland Clinic Boulevard, Weston, FL 33331; galvezn@ccf.org

Dr. Galvez-Jimenez reported teaching and speaking services for Allergan, Inc. and Lundbeck.

This article is based on Dr. Galvez-Jimenez’s presentation at “The Annual Therapy Symposium on Movement Disorders for the Modern Clinician” held in Fort Lauderdale, Florida, on January 29, 2011. The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Galvez-Jimenez.

Author and Disclosure Information

Nestor Galvez-Jimenez, MD, MSc, MS(HSA), FACP
Pauline M. Braathen Endowed Chair in Movement Disorders, Chairman, Department of Neurology, Chief, Movement Disorders Section, Cleveland Clinic Florida, Weston, FL

Correspondence: Nestor Galvez-Jimenez, MD, Cleveland Clinic Florida, 2950 Cleveland Clinic Boulevard, Weston, FL 33331; galvezn@ccf.org

Dr. Galvez-Jimenez reported teaching and speaking services for Allergan, Inc. and Lundbeck.

This article is based on Dr. Galvez-Jimenez’s presentation at “The Annual Therapy Symposium on Movement Disorders for the Modern Clinician” held in Fort Lauderdale, Florida, on January 29, 2011. The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Galvez-Jimenez.

Article PDF
Article PDF

Tourette syndrome (TS) is part of a spectrum of tic disorders. Tics are sudden, rapid, stereotyped, repetitive, nonrhythmic movements or vocalizations affecting discrete muscle groups, and are preceded by a sensory component. Patients in whom tic suppression is attempted report the experience of a sensation of inner pressure that must be released. This eventually results in the performance of motor movement or vocal sounds. TS is a disorder of childhood onset that is characterized by multiple motor and vocal tics. In some cases, there are features of obsessive compulsive disorder (OCD), attention deficit hyperactivity disorder (ADHD), or other behavioral manifestations such as coprolalia, echopraxia, palilalia, and self-injury.1,2 The spectrum of tic disorders includes:

  • Transient tics of childhood (tic duration less than 12 months)
  • Chronic motor or vocal tics (lasting more than 12 months), and
  • TS (variable motor and vocal tics lasting more than 12 months).

Many children meet the diagnostic criteria for TS between the ages of 6 and 9 years, but symptoms may improve by adulthood. The eventual loss of tics over time reflects the maturation of brain systems that control ballistic action.3

The tics that accompany TS may be defined as simple or complex, and as motor or vocal. Simple motor tics involve only a few muscles, such as eye blinking, shoulder shrugging, or facial grimacing. Complex motor tics involve multiple groups of muscles that are recruited in orchestrated bouts (eg, hand gestures, jumping, touching, or pressing), and may include copropraxia (a sudden tic-like vulgar, sexual, or obscene gesture) or echopraxia (involuntary, spontaneous imitation of someone else’s movements). Simple vocal tics are meaningless sounds such as throat clearing, grunting, sniffing, snorting, and chirping. Complex vocal tics involve speech and language such as sudden, spontaneous expression of single words or phrases, or speech blocking.4

Tics may be acquired as a consequence of other disorders, including head trauma, encephalitis, stroke, carbon monoxide poisoning, Creutzfeldt-Jakob disease, neurosyphilis, hypoglycemia, or Sydenham chorea.5 Genetic disorders such as Huntington disease may be associated with tics. Tics may also occur with certain chromosomal abnormalities or be associated with some neuropsychiatric disorders. Finally, tics may be caused by a large number of medications or illicit drugs, including cocaine, amphetamines, antipsychotics, and antidepressants.

The prevalence of all types of tics in childhood is approximately 6% to 12%, although the prevalence of chronic vocal tics is approximately 1 to 10 per 1,000 children and adolescents.6 TS is especially common among autistic children and in those with Asperger syndrome and other autistic spectrum disorders. A survey of patients at Cleveland Clinic Florida found that tics and TS accounted for 8% of all patients with movement disorders. Of patients with tics or TS who were older than 18 years, 70% were male.

PATHOPHYSIOLOGY OF TOURETTE SYNDROME: ROLE OF THE BASAL GANGLIA

Although the pathophysiology of TS is not completely understood, abnormal function of the basal ganglia is thought to be a central component of the disorder. The basal ganglia normally act to facilitate voluntary movements while suppressing competing involuntary ones. Abnormalities of basal ganglia activity are important in several disorders of motor function.7 Output neurons from the basal ganglia inhibit thalamic motor nuclei and midbrain neurons of the extrapyramidal motor system, and act to inhibit motor pattern generators in the cerebral cortex and brainstem. Hyperkinetic disorders, including tics, chorea, and dystonia, are thought to result at least in part from impaired inhibition of unwanted motor activity from the basal ganglia to downstream motor centers.7

Family heritability studies provide strong support that TS is a genetic disorder. For example, the concordance rate is 86% for monozygotic twins versus 20% for dizygotic twins.8 Chromosomes linked to TS include 2p32.2 and 13q31.1.9,10 Interactions between genetics and environment are also thought to play a significant role. The concept of pediatric autoimmune neuropsychiatric disorders associated with streptococcal (PANDAS) infections has been proposed to explain an apparent temporal association between streptococcal infections and exacerbation of tics. According to this model, molecular mimicry between streptococcal antigens and endogenous brain antigens results in an autoimmune attack.11 However, the identification of specific antibodies against basal ganglia cells remains controversial.12

MANAGEMENT OVERVIEW

Accurate diagnosis of TS is essential, and includes a complete history and neurologic examination. The tic phenomenology (complex vs simple) should be characterized, and the patient should be carefully questioned to identify the symptoms that are most bothersome (eg, motor or vocal tics, OCD, or ADHD). Pharmacotherapy should be reserved for problems that are functionally disabling and not remediable by nonpharmacologic interventions.

Treatment may also be required for other neuropsychiatric symptoms. Anxiety and depression have been reported in 19% to 80% of patients with tics, and depression is strongly correlated with the duration and severity of tics.13,14 Episodic outburst (rage), self-injurious, OCD, antisocial, and oppositional behaviors are all more common among individuals with tic disorders.15 Personality disorders may be related to OCD, ADHD, or to family or economic issues. Tic disorders are also associated with an increased incidence of somatic complaints, as well as higher rates of academic difficulties, which may be related to ADHD or medications. Sleep disturbances affect an estimated 20% to 50% of patients, and may include difficulty initiating or maintaining sleep, restlessness, movement-related arousal, or parasomnia.16

Education is an important part of treatment, and may include the patient, family members, teachers or other school staff, and work colleagues. A number of behavioral or psychosocial approaches may help to improve tics, including conditioning techniques, relaxation training, biofeedback, habit reversal, awareness training, and hypnosis.17

 

 

PHARMACOLOGIC TREATMENT: THREE TIERS

Options for the pharmacologic treatment of tics and TS include dopamine blockers, dopamine depleters, benzodiazepines, central alpha-adrenergic blockers, and botulinum toxin. Pharmacotherapy options can be divided into three tiers (Table), with first-tier drugs considered first-choice treatments.

First-tier therapies

The alpha-adrenergic blockers clonidine and guanfacine are first-tier therapies. Treatment should be initiated at a low dose and escalated gradually according to response, which is determined by the severity, and not the presence, of tics. Clonidine may be administered at a dose of 0.025 mg two or three times daily or, for maintenance, 0.1 mg three times daily; another option is 0.1, 0.2, or 0.3 mg weekly by transdermal administration. Guanfacine may be administered at a dose of 1 mg once daily. Alpha-adrenergic blockers are useful for the treatment of mild tics, and are considered first-line therapy for tic suppression. Side effects may include dry mouth, somnolence, and, rarely, blood pressure fluctuations.

Agents that affect gamma-amino butyric acid (GABA) neurotransmission have been associated with improved symptoms of tic disorders.18 For example, both clonazepam and diazepam have been reported to reduce TS symptoms.18 Both of these benzodiazepines are associated with sedation, blunting of cognition, and exacerbation of depression, however.19,20

Second-tier therapies

Second-tier therapies, consisting of neuroleptics, induce a rapid treatment response. Haloperidol may be started at a dose of 0.25 mg once daily, with a maintenance dosage of 0.5 to 3.0 mg/day. Cognitive blunting or extrapyramidal side effects are rare in patients with TS, but the potential for these side effects should be thoroughly discussed with the patient or parent/guardian before treatment. Pimozide 0.5 mg (2 to 6 mg/day for maintenance) may be associated with tremor or parkinsonian symptoms (predominantly akinesia). Risperidone 0.25 mg/day (0.5 to 4 mg/day for maintenance), olanzapine 2.5 mg/day (5 to 10 mg/day for maintenance), and quetiapine 25 mg twice daily (100 to 300 mg/day for maintenance) are associated with potential adverse effects of extrapyramidal symptoms, weight gain, and diabetes.

Third-tier therapies

Dopamine agonists (reserpine and tetrabenazine) and botulinum toxin are third-tier therapies. Reserpine, although rarely used in current clinical practice, may be administered at doses of 0.1 to 0.25 mg/day, titrating upward on the basis of clinical response. Tetrabenazine may be administered at a starting dose of 12.5 mg/day, with higher doses as needed depending on the response to treatment. Adverse effects include hypotension, sedation, extrapyramidal symptoms (predominantly parkinsonism), and depression.21

The exact mechanism by which tetrabenazine produces this suppression effect is unknown, but it is believed to be related to its effect of reversibly depleting monoamines. At least three neuronal protein classes regulate the effects of dopamine on voluntary and involuntary movement.22 The two presynaptic proteins are vesicular monoamine transporter subtype 2 (VMAT2) and dopamine transporter (DAT). Postsynaptically, dopamine activity is regulated by G-protein–linked dopamine receptors (eg, the D2 receptor). Tetrabenazine reduces the uptake of monoamines (including dopamine) into synaptic vesicles by reversibly binding to VMAT2, resulting in degradation of dopamine within axon terminals by monoamine oxidases.23 By blocking dopamine transport, tetrabenazine depletes dopamine with greater selectivity than it does other monoamines.24

The dosage of tetrabenazine for the treatment of motor disorders, particularly chorea, was established in the Huntington Study Group (HSG) clinical trial.25 In the HSG trial, a starting dose of 12.5 mg on day 1 was increased to 12.5 mg twice daily on days 2 to 7, and then by 12.5 mg/day at weekly intervals until the desired clinical effect, intolerable adverse effects, or a maximum dose of 100 mg/day was reached. Daily dosages of 37.5 mg or more are administered in three divided doses. Adverse events (reported in 70% of patients who received placebo and 91% of patients who received tetrabenazine) include sedation or somnolence, insomnia, and fatigue.21 These findings may be carried over to patients with tics.

Botulinum toxin may also help to control tics—especially dystonic tics. The premonitory symptoms of TS are usually unaffected by botulinum toxin.26 The adverse effect profile for patients with TS is similar to that of patients with dystonia or facial dyskinesia, and may include soreness, transient weakness, ptosis (if injected for eye blinking), and mild transient dysphagia (if injected into the larynx).27

MANAGING COMORBID CONDITIONS

Approximately 30% of patients with TS also have OCD.28 Treatment options include selective serotonin reuptake inhibitors at standard doses, and the tricyclic antidepressant clomipramine (25 mg once or twice daily, or 75 mg/day in sustained-release form). Trazodone, a serotonin antagonist and reuptake inhibitor that is associated with a lower incidence of anticholinergic effects, may be initiated at a dose of 50 mg/day and slowly increased to 150 to 400 mg/day depending on clinical response.

As many as 60% of patients with TS may also have ADHD.28 Methylphenidate is helpful for the treatment of ADHD and does not exacerbate tics, but it is a restricted medication. The recommended dose is 20 mg once daily, titrated upward as needed based on response. Atomoxetine carries a warning regarding increased risk of suicide. It has also been associated with an increased risk of sexual dysfunction and behavioral changes, including aggressive behaviors, agitation, and irratibility.29

 

 

DEEP BRAIN STIMULATION

Deep brain stimulation (DBS) has been shown to improve TS in single-case studies and in small series, although the long-term benefit is unclear. Potential targets of stimulation include midline thalamic centromedian-parafascicular (CM-PF) nuclei, the ventralis oralis complex of the thalamus, motor and limbic globus pallidus pars interna (GPi), and the anterior limb of the internal capsule.30 In particular, stimulation of the sensorimotor GPi may ameliorate hyperkinetic states.

One report described the results of DBS implantation in a 15-year-old boy with TS who had not responded to several pharmacologic treatment options.31 Six months after implantation, the patient exhibited markedly improved tic severity as measured using the Yale Global Tic Severity Scale, including a 76% reduction in motor tic severity, 68% reduction in vocal tics, and a complete resolution of impairment.31

Published consensus criteria for the selection of suitable candidates for DBS include age greater than 25 years, chronic and severe tics with severe functional impairment for at least 12 months, tics that are frequent and noticeable in most situations most of the time, failure of conventional medical therapy, medical stability for 6 months, and willingness to participate in ongoing psychologic interventions.32 Exclusion criteria include the presence of another medical condition that could explain the tics, an unstable medical condition, being considered likely to benefit from psychologic interventions, psychosocial factors that may complicate the recovery process or make it difficult to assess outcome, and unwillingness to participate in ongoing treatment for psychosocial problems or risk factors. Other factors that should be considered include comorbidities, the variability in tic severity over time, the involvement of a multidisciplinary treatment team, results of a thorough neuropsychologic assessment, expertise of the surgical team, and access to imaging facilities for presurgical mapping and postsurgical evaluation.

SUMMARY AND CONCLUSIONS

Tourette syndrome is not uncommon among the adult population of a typical neurology practice, and should not be considered exclusively a pediatric diagnosis. Several treatment options are available, including behavioral approaches and several medications. Treatment should focus on the most disabling symptoms. Neuropsychologic assessment and psychiatric support may be necessary for some patients. The same comorbidities that are encountered in children are usually evident in adult patients as well. In medically refractory cases, DBS surgery may be helpful.

Tourette syndrome (TS) is part of a spectrum of tic disorders. Tics are sudden, rapid, stereotyped, repetitive, nonrhythmic movements or vocalizations affecting discrete muscle groups, and are preceded by a sensory component. Patients in whom tic suppression is attempted report the experience of a sensation of inner pressure that must be released. This eventually results in the performance of motor movement or vocal sounds. TS is a disorder of childhood onset that is characterized by multiple motor and vocal tics. In some cases, there are features of obsessive compulsive disorder (OCD), attention deficit hyperactivity disorder (ADHD), or other behavioral manifestations such as coprolalia, echopraxia, palilalia, and self-injury.1,2 The spectrum of tic disorders includes:

  • Transient tics of childhood (tic duration less than 12 months)
  • Chronic motor or vocal tics (lasting more than 12 months), and
  • TS (variable motor and vocal tics lasting more than 12 months).

Many children meet the diagnostic criteria for TS between the ages of 6 and 9 years, but symptoms may improve by adulthood. The eventual loss of tics over time reflects the maturation of brain systems that control ballistic action.3

The tics that accompany TS may be defined as simple or complex, and as motor or vocal. Simple motor tics involve only a few muscles, such as eye blinking, shoulder shrugging, or facial grimacing. Complex motor tics involve multiple groups of muscles that are recruited in orchestrated bouts (eg, hand gestures, jumping, touching, or pressing), and may include copropraxia (a sudden tic-like vulgar, sexual, or obscene gesture) or echopraxia (involuntary, spontaneous imitation of someone else’s movements). Simple vocal tics are meaningless sounds such as throat clearing, grunting, sniffing, snorting, and chirping. Complex vocal tics involve speech and language such as sudden, spontaneous expression of single words or phrases, or speech blocking.4

Tics may be acquired as a consequence of other disorders, including head trauma, encephalitis, stroke, carbon monoxide poisoning, Creutzfeldt-Jakob disease, neurosyphilis, hypoglycemia, or Sydenham chorea.5 Genetic disorders such as Huntington disease may be associated with tics. Tics may also occur with certain chromosomal abnormalities or be associated with some neuropsychiatric disorders. Finally, tics may be caused by a large number of medications or illicit drugs, including cocaine, amphetamines, antipsychotics, and antidepressants.

The prevalence of all types of tics in childhood is approximately 6% to 12%, although the prevalence of chronic vocal tics is approximately 1 to 10 per 1,000 children and adolescents.6 TS is especially common among autistic children and in those with Asperger syndrome and other autistic spectrum disorders. A survey of patients at Cleveland Clinic Florida found that tics and TS accounted for 8% of all patients with movement disorders. Of patients with tics or TS who were older than 18 years, 70% were male.

PATHOPHYSIOLOGY OF TOURETTE SYNDROME: ROLE OF THE BASAL GANGLIA

Although the pathophysiology of TS is not completely understood, abnormal function of the basal ganglia is thought to be a central component of the disorder. The basal ganglia normally act to facilitate voluntary movements while suppressing competing involuntary ones. Abnormalities of basal ganglia activity are important in several disorders of motor function.7 Output neurons from the basal ganglia inhibit thalamic motor nuclei and midbrain neurons of the extrapyramidal motor system, and act to inhibit motor pattern generators in the cerebral cortex and brainstem. Hyperkinetic disorders, including tics, chorea, and dystonia, are thought to result at least in part from impaired inhibition of unwanted motor activity from the basal ganglia to downstream motor centers.7

Family heritability studies provide strong support that TS is a genetic disorder. For example, the concordance rate is 86% for monozygotic twins versus 20% for dizygotic twins.8 Chromosomes linked to TS include 2p32.2 and 13q31.1.9,10 Interactions between genetics and environment are also thought to play a significant role. The concept of pediatric autoimmune neuropsychiatric disorders associated with streptococcal (PANDAS) infections has been proposed to explain an apparent temporal association between streptococcal infections and exacerbation of tics. According to this model, molecular mimicry between streptococcal antigens and endogenous brain antigens results in an autoimmune attack.11 However, the identification of specific antibodies against basal ganglia cells remains controversial.12

MANAGEMENT OVERVIEW

Accurate diagnosis of TS is essential, and includes a complete history and neurologic examination. The tic phenomenology (complex vs simple) should be characterized, and the patient should be carefully questioned to identify the symptoms that are most bothersome (eg, motor or vocal tics, OCD, or ADHD). Pharmacotherapy should be reserved for problems that are functionally disabling and not remediable by nonpharmacologic interventions.

Treatment may also be required for other neuropsychiatric symptoms. Anxiety and depression have been reported in 19% to 80% of patients with tics, and depression is strongly correlated with the duration and severity of tics.13,14 Episodic outburst (rage), self-injurious, OCD, antisocial, and oppositional behaviors are all more common among individuals with tic disorders.15 Personality disorders may be related to OCD, ADHD, or to family or economic issues. Tic disorders are also associated with an increased incidence of somatic complaints, as well as higher rates of academic difficulties, which may be related to ADHD or medications. Sleep disturbances affect an estimated 20% to 50% of patients, and may include difficulty initiating or maintaining sleep, restlessness, movement-related arousal, or parasomnia.16

Education is an important part of treatment, and may include the patient, family members, teachers or other school staff, and work colleagues. A number of behavioral or psychosocial approaches may help to improve tics, including conditioning techniques, relaxation training, biofeedback, habit reversal, awareness training, and hypnosis.17

 

 

PHARMACOLOGIC TREATMENT: THREE TIERS

Options for the pharmacologic treatment of tics and TS include dopamine blockers, dopamine depleters, benzodiazepines, central alpha-adrenergic blockers, and botulinum toxin. Pharmacotherapy options can be divided into three tiers (Table), with first-tier drugs considered first-choice treatments.

First-tier therapies

The alpha-adrenergic blockers clonidine and guanfacine are first-tier therapies. Treatment should be initiated at a low dose and escalated gradually according to response, which is determined by the severity, and not the presence, of tics. Clonidine may be administered at a dose of 0.025 mg two or three times daily or, for maintenance, 0.1 mg three times daily; another option is 0.1, 0.2, or 0.3 mg weekly by transdermal administration. Guanfacine may be administered at a dose of 1 mg once daily. Alpha-adrenergic blockers are useful for the treatment of mild tics, and are considered first-line therapy for tic suppression. Side effects may include dry mouth, somnolence, and, rarely, blood pressure fluctuations.

Agents that affect gamma-amino butyric acid (GABA) neurotransmission have been associated with improved symptoms of tic disorders.18 For example, both clonazepam and diazepam have been reported to reduce TS symptoms.18 Both of these benzodiazepines are associated with sedation, blunting of cognition, and exacerbation of depression, however.19,20

Second-tier therapies

Second-tier therapies, consisting of neuroleptics, induce a rapid treatment response. Haloperidol may be started at a dose of 0.25 mg once daily, with a maintenance dosage of 0.5 to 3.0 mg/day. Cognitive blunting or extrapyramidal side effects are rare in patients with TS, but the potential for these side effects should be thoroughly discussed with the patient or parent/guardian before treatment. Pimozide 0.5 mg (2 to 6 mg/day for maintenance) may be associated with tremor or parkinsonian symptoms (predominantly akinesia). Risperidone 0.25 mg/day (0.5 to 4 mg/day for maintenance), olanzapine 2.5 mg/day (5 to 10 mg/day for maintenance), and quetiapine 25 mg twice daily (100 to 300 mg/day for maintenance) are associated with potential adverse effects of extrapyramidal symptoms, weight gain, and diabetes.

Third-tier therapies

Dopamine agonists (reserpine and tetrabenazine) and botulinum toxin are third-tier therapies. Reserpine, although rarely used in current clinical practice, may be administered at doses of 0.1 to 0.25 mg/day, titrating upward on the basis of clinical response. Tetrabenazine may be administered at a starting dose of 12.5 mg/day, with higher doses as needed depending on the response to treatment. Adverse effects include hypotension, sedation, extrapyramidal symptoms (predominantly parkinsonism), and depression.21

The exact mechanism by which tetrabenazine produces this suppression effect is unknown, but it is believed to be related to its effect of reversibly depleting monoamines. At least three neuronal protein classes regulate the effects of dopamine on voluntary and involuntary movement.22 The two presynaptic proteins are vesicular monoamine transporter subtype 2 (VMAT2) and dopamine transporter (DAT). Postsynaptically, dopamine activity is regulated by G-protein–linked dopamine receptors (eg, the D2 receptor). Tetrabenazine reduces the uptake of monoamines (including dopamine) into synaptic vesicles by reversibly binding to VMAT2, resulting in degradation of dopamine within axon terminals by monoamine oxidases.23 By blocking dopamine transport, tetrabenazine depletes dopamine with greater selectivity than it does other monoamines.24

The dosage of tetrabenazine for the treatment of motor disorders, particularly chorea, was established in the Huntington Study Group (HSG) clinical trial.25 In the HSG trial, a starting dose of 12.5 mg on day 1 was increased to 12.5 mg twice daily on days 2 to 7, and then by 12.5 mg/day at weekly intervals until the desired clinical effect, intolerable adverse effects, or a maximum dose of 100 mg/day was reached. Daily dosages of 37.5 mg or more are administered in three divided doses. Adverse events (reported in 70% of patients who received placebo and 91% of patients who received tetrabenazine) include sedation or somnolence, insomnia, and fatigue.21 These findings may be carried over to patients with tics.

Botulinum toxin may also help to control tics—especially dystonic tics. The premonitory symptoms of TS are usually unaffected by botulinum toxin.26 The adverse effect profile for patients with TS is similar to that of patients with dystonia or facial dyskinesia, and may include soreness, transient weakness, ptosis (if injected for eye blinking), and mild transient dysphagia (if injected into the larynx).27

MANAGING COMORBID CONDITIONS

Approximately 30% of patients with TS also have OCD.28 Treatment options include selective serotonin reuptake inhibitors at standard doses, and the tricyclic antidepressant clomipramine (25 mg once or twice daily, or 75 mg/day in sustained-release form). Trazodone, a serotonin antagonist and reuptake inhibitor that is associated with a lower incidence of anticholinergic effects, may be initiated at a dose of 50 mg/day and slowly increased to 150 to 400 mg/day depending on clinical response.

As many as 60% of patients with TS may also have ADHD.28 Methylphenidate is helpful for the treatment of ADHD and does not exacerbate tics, but it is a restricted medication. The recommended dose is 20 mg once daily, titrated upward as needed based on response. Atomoxetine carries a warning regarding increased risk of suicide. It has also been associated with an increased risk of sexual dysfunction and behavioral changes, including aggressive behaviors, agitation, and irratibility.29

 

 

DEEP BRAIN STIMULATION

Deep brain stimulation (DBS) has been shown to improve TS in single-case studies and in small series, although the long-term benefit is unclear. Potential targets of stimulation include midline thalamic centromedian-parafascicular (CM-PF) nuclei, the ventralis oralis complex of the thalamus, motor and limbic globus pallidus pars interna (GPi), and the anterior limb of the internal capsule.30 In particular, stimulation of the sensorimotor GPi may ameliorate hyperkinetic states.

One report described the results of DBS implantation in a 15-year-old boy with TS who had not responded to several pharmacologic treatment options.31 Six months after implantation, the patient exhibited markedly improved tic severity as measured using the Yale Global Tic Severity Scale, including a 76% reduction in motor tic severity, 68% reduction in vocal tics, and a complete resolution of impairment.31

Published consensus criteria for the selection of suitable candidates for DBS include age greater than 25 years, chronic and severe tics with severe functional impairment for at least 12 months, tics that are frequent and noticeable in most situations most of the time, failure of conventional medical therapy, medical stability for 6 months, and willingness to participate in ongoing psychologic interventions.32 Exclusion criteria include the presence of another medical condition that could explain the tics, an unstable medical condition, being considered likely to benefit from psychologic interventions, psychosocial factors that may complicate the recovery process or make it difficult to assess outcome, and unwillingness to participate in ongoing treatment for psychosocial problems or risk factors. Other factors that should be considered include comorbidities, the variability in tic severity over time, the involvement of a multidisciplinary treatment team, results of a thorough neuropsychologic assessment, expertise of the surgical team, and access to imaging facilities for presurgical mapping and postsurgical evaluation.

SUMMARY AND CONCLUSIONS

Tourette syndrome is not uncommon among the adult population of a typical neurology practice, and should not be considered exclusively a pediatric diagnosis. Several treatment options are available, including behavioral approaches and several medications. Treatment should focus on the most disabling symptoms. Neuropsychologic assessment and psychiatric support may be necessary for some patients. The same comorbidities that are encountered in children are usually evident in adult patients as well. In medically refractory cases, DBS surgery may be helpful.

References
  1. Robertson MM. Annotation: Gilles de la Tourette syndrome—an update. J Child Psychol Psychiatry 1994; 35:597611.
  2. Robertson MM, Althoff RR, Hafez A, Pauls DL. Principal components analysis of a large cohort with Tourette syndrome. Br J Psychiatry 2008; 193:3136.
  3. Flaherty AW. Movement disorders. In: Stern TA, Rosenbaum JF, Fava M, Biederman J, Rauch SL, eds. Massachusetts General Hospital: Comprehensive Clinical Psychiatry E-Book. Philadelphia, PA: Mosby Elsevier; 2008.
  4. Müller N. Tourette’s syndrome: clinical features, pathophysiology, and therapeutic approaches. Dialogues Clin Neurosci 2007; 9:161171.
  5. Bagheri MM, Kerbeshian J, Burd L. Recognition and management of Tourette’s syndrome and tic disorders. Am Fam Physician 1999; 59:22632272,2274.
  6. Lombroso PJ, Scahill L. Tourette syndrome and obsessive–compulsive disorder [published online ahead of print October 15, 2007]. Brain Dev 2008; 30:231237. 10.1016/j.braindev.2007.09.001
  7. Mink JW. The basal ganglia and involuntary movements: impaired inhibition of competing motor patterns. Arch Neurol 2003; 60:13651368.
  8. Singer HS, Smith-Hicks C, Lieberman D. Tourette syndrome. In: LeDoux M, ed. Animal Models of Movement Disorders. Academic Press; 2005.
  9. Tourette Syndrome Association International Consortium for Genetics. Genome scan for Tourette disorder in affected-sibling-pair and multigenerational families. Am J Hum Genet 2007; 80:265272.
  10. Abelson JF, Kwan KY, O’Roak BJ, et al. Sequence variants in SLITRK1 are associated with Tourette’s syndrome. Science 2005; 310:317320.
  11. Kurlan R. Tourette’s syndrome and ‘PANDAS’: will the relation bear out? Pediatric autoimmune neuropsychiatric disorders associated with streptococcal infection. Neurology 1998; 50:15301534.
  12. Morris CM, Pardo-Villamizar C, Gause CD, Singer HS. Serum autoantibodies measured by immunofluorescence confirm a failure to differentiate PANDAS and Tourette syndrome from controls [published online ahead of print September 27, 2008]. J Neurol Sci 2009; 276:4548. 10.1016/j.jns.2008.08.032
  13. Comings BG, Comings DE. A controlled study of Tourette syndrome. V. Depression and mania. Am J Hum Genet 1987; 41:804821.
  14. Robertson MM, Williamson F, Eapen V. Depressive symptomatology in young people with Gilles de la Tourette syndrome—a comparison of self-report scales [published online ahead of print February 7, 2006]. J Affect Disord 2006; 91:265268. 10.1016/j.jad.2005.12.046
  15. Budman CL, Bruun RD, Park KS, Lesser M, Olson M. Explosive outbursts in children with Tourette’s disorder. J Am Acad Child Adolesc Psychiatry 2000; 39:12701276.
  16. Kostanecka-Endress T, Banaschewski T, Kinkelbur J, et al. Disturbed sleep in children with Tourette syndrome: a polysomnographic study. J Psychosom Res 2003; 55:2329.
  17. Peterson AL. Psychosocial management of tics and intentional repetitive behaviors associated with Tourette syndrome. In:Woods DW, Piacentini J, Walkup JT, eds. Treating Tourette Syndrome and Tic Disorders: A Guide for Practitioners. Guilford Press; 2007.
  18. Robertson MM. Tourette syndrome, associated conditions and the complexities of treatment. Brain 2000; 123:425462.
  19. Klonopin [package insert]. South San Francisco, CA: Genentech USA, Inc.; 2010.
  20. Valium [package insert]. Nutley, NJ: Roche Laboratories, Inc.; 2008.
  21. Xenazine [package insert]. Deerfield, IL: Lundbeck Inc.; 2011.
  22. Schmitz Y, Benoit-Marand M, Gonon F, Sulzer D. Presynaptic regulation of dopaminergic neurotransmission. J Neurochem 2003; 87:273289.
  23. Morrow T. Gene therapy offers HD patients relief from some symptoms. Tetrabenazine inhibits the transport of a molecule called vesicular monoamine transporter type 2 or VMAT2. Manag Care 2008; 17:4647.
  24. Pearson SJ, Reynolds GP. Depletion of monoamine transmitters by tetrabenazine in brain tissue in Huntington’s disease. Neuropharmacology 1988; 27:717719.
  25. Huntington Study Group. Tetrabenazine as antichorea therapy in Huntington disease: a randomized controlled trial. Neurology 2006; 66:366372.
  26. Jankovic J, Kurlan R. Tourette syndrome: evolving concepts [published online ahead of print April 11, 2011]. Mov Disord 2011; 26:11491156. 10.1002/mds.23618
  27. Swain JE, Leckman JF. Tourette syndrome and tic disorders: overview and practical guide to diagnosis and treatment. Psychiatry (Edgmont) 2005; 2:2636.
  28. Gorman DA, Thompson N, Plessen KJ, Robertson MM, Leckman JF, Peterson BS. Psychosocial outcome and psychiatric comorbidity in older adolescents with Tourette syndrome: controlled study. Br J Psychiatry 2010; 197:3644.
  29. Strattera [package insert]. Indianapolis, IN: Eli Lilly and Company; 2011.
  30. Ackermans L, Temel Y, Visser-Vandewalle V. Deep brain stimulation in Tourette’s syndrome. Neurotherapeutics 2008; 5:339344.
  31. Shahed J, Poysky J, Kenney C, Simpson R, Jankovic J. GPi deep brain stimulation for Tourette syndrome improves tics and psychiatric comorbidities. Neurology 2007; 68:159160.
  32. Mink JW, Walkup J, Frey KA, et al .Patient selection and assessment recommendations for deep brain stimulation in Tourette syndrome. Mov Disord 2006; 21:18311838.
References
  1. Robertson MM. Annotation: Gilles de la Tourette syndrome—an update. J Child Psychol Psychiatry 1994; 35:597611.
  2. Robertson MM, Althoff RR, Hafez A, Pauls DL. Principal components analysis of a large cohort with Tourette syndrome. Br J Psychiatry 2008; 193:3136.
  3. Flaherty AW. Movement disorders. In: Stern TA, Rosenbaum JF, Fava M, Biederman J, Rauch SL, eds. Massachusetts General Hospital: Comprehensive Clinical Psychiatry E-Book. Philadelphia, PA: Mosby Elsevier; 2008.
  4. Müller N. Tourette’s syndrome: clinical features, pathophysiology, and therapeutic approaches. Dialogues Clin Neurosci 2007; 9:161171.
  5. Bagheri MM, Kerbeshian J, Burd L. Recognition and management of Tourette’s syndrome and tic disorders. Am Fam Physician 1999; 59:22632272,2274.
  6. Lombroso PJ, Scahill L. Tourette syndrome and obsessive–compulsive disorder [published online ahead of print October 15, 2007]. Brain Dev 2008; 30:231237. 10.1016/j.braindev.2007.09.001
  7. Mink JW. The basal ganglia and involuntary movements: impaired inhibition of competing motor patterns. Arch Neurol 2003; 60:13651368.
  8. Singer HS, Smith-Hicks C, Lieberman D. Tourette syndrome. In: LeDoux M, ed. Animal Models of Movement Disorders. Academic Press; 2005.
  9. Tourette Syndrome Association International Consortium for Genetics. Genome scan for Tourette disorder in affected-sibling-pair and multigenerational families. Am J Hum Genet 2007; 80:265272.
  10. Abelson JF, Kwan KY, O’Roak BJ, et al. Sequence variants in SLITRK1 are associated with Tourette’s syndrome. Science 2005; 310:317320.
  11. Kurlan R. Tourette’s syndrome and ‘PANDAS’: will the relation bear out? Pediatric autoimmune neuropsychiatric disorders associated with streptococcal infection. Neurology 1998; 50:15301534.
  12. Morris CM, Pardo-Villamizar C, Gause CD, Singer HS. Serum autoantibodies measured by immunofluorescence confirm a failure to differentiate PANDAS and Tourette syndrome from controls [published online ahead of print September 27, 2008]. J Neurol Sci 2009; 276:4548. 10.1016/j.jns.2008.08.032
  13. Comings BG, Comings DE. A controlled study of Tourette syndrome. V. Depression and mania. Am J Hum Genet 1987; 41:804821.
  14. Robertson MM, Williamson F, Eapen V. Depressive symptomatology in young people with Gilles de la Tourette syndrome—a comparison of self-report scales [published online ahead of print February 7, 2006]. J Affect Disord 2006; 91:265268. 10.1016/j.jad.2005.12.046
  15. Budman CL, Bruun RD, Park KS, Lesser M, Olson M. Explosive outbursts in children with Tourette’s disorder. J Am Acad Child Adolesc Psychiatry 2000; 39:12701276.
  16. Kostanecka-Endress T, Banaschewski T, Kinkelbur J, et al. Disturbed sleep in children with Tourette syndrome: a polysomnographic study. J Psychosom Res 2003; 55:2329.
  17. Peterson AL. Psychosocial management of tics and intentional repetitive behaviors associated with Tourette syndrome. In:Woods DW, Piacentini J, Walkup JT, eds. Treating Tourette Syndrome and Tic Disorders: A Guide for Practitioners. Guilford Press; 2007.
  18. Robertson MM. Tourette syndrome, associated conditions and the complexities of treatment. Brain 2000; 123:425462.
  19. Klonopin [package insert]. South San Francisco, CA: Genentech USA, Inc.; 2010.
  20. Valium [package insert]. Nutley, NJ: Roche Laboratories, Inc.; 2008.
  21. Xenazine [package insert]. Deerfield, IL: Lundbeck Inc.; 2011.
  22. Schmitz Y, Benoit-Marand M, Gonon F, Sulzer D. Presynaptic regulation of dopaminergic neurotransmission. J Neurochem 2003; 87:273289.
  23. Morrow T. Gene therapy offers HD patients relief from some symptoms. Tetrabenazine inhibits the transport of a molecule called vesicular monoamine transporter type 2 or VMAT2. Manag Care 2008; 17:4647.
  24. Pearson SJ, Reynolds GP. Depletion of monoamine transmitters by tetrabenazine in brain tissue in Huntington’s disease. Neuropharmacology 1988; 27:717719.
  25. Huntington Study Group. Tetrabenazine as antichorea therapy in Huntington disease: a randomized controlled trial. Neurology 2006; 66:366372.
  26. Jankovic J, Kurlan R. Tourette syndrome: evolving concepts [published online ahead of print April 11, 2011]. Mov Disord 2011; 26:11491156. 10.1002/mds.23618
  27. Swain JE, Leckman JF. Tourette syndrome and tic disorders: overview and practical guide to diagnosis and treatment. Psychiatry (Edgmont) 2005; 2:2636.
  28. Gorman DA, Thompson N, Plessen KJ, Robertson MM, Leckman JF, Peterson BS. Psychosocial outcome and psychiatric comorbidity in older adolescents with Tourette syndrome: controlled study. Br J Psychiatry 2010; 197:3644.
  29. Strattera [package insert]. Indianapolis, IN: Eli Lilly and Company; 2011.
  30. Ackermans L, Temel Y, Visser-Vandewalle V. Deep brain stimulation in Tourette’s syndrome. Neurotherapeutics 2008; 5:339344.
  31. Shahed J, Poysky J, Kenney C, Simpson R, Jankovic J. GPi deep brain stimulation for Tourette syndrome improves tics and psychiatric comorbidities. Neurology 2007; 68:159160.
  32. Mink JW, Walkup J, Frey KA, et al .Patient selection and assessment recommendations for deep brain stimulation in Tourette syndrome. Mov Disord 2006; 21:18311838.
Page Number
S35-S39
Page Number
S35-S39
Publications
Publications
Article Type
Display Headline
Tics and Tourette syndrome: An adult perspective
Display Headline
Tics and Tourette syndrome: An adult perspective
Citation Override
Cleveland Clinic Journal of Medicine 2012 July;79(suppl 2):S35-S39
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media