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

Quality, frailty, and common sense

Article Type
Changed
Mon, 11/13/2017 - 10:43
Display Headline
Quality, frailty, and common sense

Providing quality care for the frail elderly patient taxes the skills of the medical artisan. It requires not only knowledge and vigilance regarding appropriate drug prescribing, but also unrushed active listening, creativity, and common sense—in other words, the full palette of the art of medicine. Frail elderly patients are generally excluded from clinical trials. Hence, their management is dictated as much by our clinical experience as by evidence. Treating their congestive heart failure can be particularly challenging.

Congestive heart failure, as noted in the review by Samala et al in this issue of the Journal, is more prevalent in the elderly. Particularly in the frail elderly, managing severe congestive heart failure poses ethical, socioeconomic, and medical challenges. The presence of even subtle cognitive impairment requires detailed dialogue with family and caregivers about medications and about symptoms that warrant a trip to the emergency room. Patients on a fixed income may not be able to afford their medications and thus may use them sporadically. And the preprepared foods they often eat are laden with sodium.

The symptoms of congestive heart failure may easily go unrecognized or be attributed to other common problems. Sorting out the reasons for exertional fatigue, especially a generalized sense of fatigue, can be particularly vexing. Anemia and sarcopenia can directly cause exertional fatigue or “weakness” but may also exacerbate heart failure and cause similar symptoms. Pharmacologic and dietary causes for volume overload must be sought. Even intermittent use of over-the-counter nonsteroidal anti-inflammatory drugs can be problematic.

Severe congestive heart failure is a lethal disease. Current quality guidelines for its treatment emphasize the use of multiple drugs and devices. Yet vasoactive drugs may not be well tolerated in frail patients, who are particularly vulnerable to orthostatic hypotension and cerebral hypoperfusion. Digoxin, of marginal benefit in younger patients without tachyarrhythmias, has an even more tenuous risk-benefit ratio in the frail elderly. Beta-blockers may cause fatigue and depression, and even low-dose diuretics can exacerbate symptoms of bladder dysfunction. Previously implanted defibrillators may be inconsistent with the patient’s current end-of-life desires.

Ideal management of the genuinely frail elderly patient with severe congestive heart failure is not always a matter of ventricular assist devices, biventricular pacers, or angiotensin-converting enzyme inhibitors. At some point, referral to palliative care resources, guided by informed input from the patient, family members, and caregivers, may be the most appropriate high-quality care that we can (and should) offer.

Article PDF
Author and Disclosure Information

Brian F. Mandell, MD, PhD
Editor in Chief

Issue
Cleveland Clinic Journal of Medicine - 78(12)
Publications
Topics
Page Number
777
Sections
Author and Disclosure Information

Brian F. Mandell, MD, PhD
Editor in Chief

Author and Disclosure Information

Brian F. Mandell, MD, PhD
Editor in Chief

Article PDF
Article PDF
Related Articles

Providing quality care for the frail elderly patient taxes the skills of the medical artisan. It requires not only knowledge and vigilance regarding appropriate drug prescribing, but also unrushed active listening, creativity, and common sense—in other words, the full palette of the art of medicine. Frail elderly patients are generally excluded from clinical trials. Hence, their management is dictated as much by our clinical experience as by evidence. Treating their congestive heart failure can be particularly challenging.

Congestive heart failure, as noted in the review by Samala et al in this issue of the Journal, is more prevalent in the elderly. Particularly in the frail elderly, managing severe congestive heart failure poses ethical, socioeconomic, and medical challenges. The presence of even subtle cognitive impairment requires detailed dialogue with family and caregivers about medications and about symptoms that warrant a trip to the emergency room. Patients on a fixed income may not be able to afford their medications and thus may use them sporadically. And the preprepared foods they often eat are laden with sodium.

The symptoms of congestive heart failure may easily go unrecognized or be attributed to other common problems. Sorting out the reasons for exertional fatigue, especially a generalized sense of fatigue, can be particularly vexing. Anemia and sarcopenia can directly cause exertional fatigue or “weakness” but may also exacerbate heart failure and cause similar symptoms. Pharmacologic and dietary causes for volume overload must be sought. Even intermittent use of over-the-counter nonsteroidal anti-inflammatory drugs can be problematic.

Severe congestive heart failure is a lethal disease. Current quality guidelines for its treatment emphasize the use of multiple drugs and devices. Yet vasoactive drugs may not be well tolerated in frail patients, who are particularly vulnerable to orthostatic hypotension and cerebral hypoperfusion. Digoxin, of marginal benefit in younger patients without tachyarrhythmias, has an even more tenuous risk-benefit ratio in the frail elderly. Beta-blockers may cause fatigue and depression, and even low-dose diuretics can exacerbate symptoms of bladder dysfunction. Previously implanted defibrillators may be inconsistent with the patient’s current end-of-life desires.

Ideal management of the genuinely frail elderly patient with severe congestive heart failure is not always a matter of ventricular assist devices, biventricular pacers, or angiotensin-converting enzyme inhibitors. At some point, referral to palliative care resources, guided by informed input from the patient, family members, and caregivers, may be the most appropriate high-quality care that we can (and should) offer.

Providing quality care for the frail elderly patient taxes the skills of the medical artisan. It requires not only knowledge and vigilance regarding appropriate drug prescribing, but also unrushed active listening, creativity, and common sense—in other words, the full palette of the art of medicine. Frail elderly patients are generally excluded from clinical trials. Hence, their management is dictated as much by our clinical experience as by evidence. Treating their congestive heart failure can be particularly challenging.

Congestive heart failure, as noted in the review by Samala et al in this issue of the Journal, is more prevalent in the elderly. Particularly in the frail elderly, managing severe congestive heart failure poses ethical, socioeconomic, and medical challenges. The presence of even subtle cognitive impairment requires detailed dialogue with family and caregivers about medications and about symptoms that warrant a trip to the emergency room. Patients on a fixed income may not be able to afford their medications and thus may use them sporadically. And the preprepared foods they often eat are laden with sodium.

The symptoms of congestive heart failure may easily go unrecognized or be attributed to other common problems. Sorting out the reasons for exertional fatigue, especially a generalized sense of fatigue, can be particularly vexing. Anemia and sarcopenia can directly cause exertional fatigue or “weakness” but may also exacerbate heart failure and cause similar symptoms. Pharmacologic and dietary causes for volume overload must be sought. Even intermittent use of over-the-counter nonsteroidal anti-inflammatory drugs can be problematic.

Severe congestive heart failure is a lethal disease. Current quality guidelines for its treatment emphasize the use of multiple drugs and devices. Yet vasoactive drugs may not be well tolerated in frail patients, who are particularly vulnerable to orthostatic hypotension and cerebral hypoperfusion. Digoxin, of marginal benefit in younger patients without tachyarrhythmias, has an even more tenuous risk-benefit ratio in the frail elderly. Beta-blockers may cause fatigue and depression, and even low-dose diuretics can exacerbate symptoms of bladder dysfunction. Previously implanted defibrillators may be inconsistent with the patient’s current end-of-life desires.

Ideal management of the genuinely frail elderly patient with severe congestive heart failure is not always a matter of ventricular assist devices, biventricular pacers, or angiotensin-converting enzyme inhibitors. At some point, referral to palliative care resources, guided by informed input from the patient, family members, and caregivers, may be the most appropriate high-quality care that we can (and should) offer.

Issue
Cleveland Clinic Journal of Medicine - 78(12)
Issue
Cleveland Clinic Journal of Medicine - 78(12)
Page Number
777
Page Number
777
Publications
Publications
Topics
Article Type
Display Headline
Quality, frailty, and common sense
Display Headline
Quality, frailty, and common sense
Sections
Disallow All Ads
Alternative CME
Article PDF Media

Heart failure in frail, older patients: We can do ‘MORE’

Article Type
Changed
Mon, 11/13/2017 - 15:09
Display Headline
Heart failure in frail, older patients: We can do ‘MORE’

Mr. R. is an 85-year-old with congestive heart failure; the last time his ejection fraction was measured it was 30%. He also has hypertension, coronary artery disease (for which he underwent triple-vessel coronary artery bypass grafting), osteoarthritis, hyperlipidemia, and chronic obstructive pulmonary disease. He currently takes lisinopril (Zestril), carvedilol (Coreg), aspirin, clopidogrel (Plavix), digoxin, simvastatin (Zocor), furosemide (Lasix), an albuterol inhaler (Proventil), and over-the-counter naproxen (Naprosyn), the last two taken as needed.

Accompanied by his daughter, Mr. R. comes to see his primary care physician for a routine follow-up visit. He says he feels fine and has no shortness of breath or chest pain, but he feels light-headed at times, especially when he gets out of bed. He also mentions that he is bothered with having to get up three to four times at night to urinate.

On further questioning, he relates that he uses a cane to walk around the house and gets short of breath when walking from his bed to the bathroom and from one room to the next. He can feed himself, but he needs assistance with bathing and getting dressed.

Mr. R. admits that he has been feeling lonely since his wife died about a year ago. He now lives with his daughter and her family, and they all get along well. His daughter mentions that over the last 6 months he has not been eating well, that he appears to have lost interest in doing some of the things that he used to enjoy, and that he has lost weight. She adds that he has fallen twice in the last month.

On physical examination, Mr. R. is without distress but appears weak. He answers all questions appropriately, although his affect is flat and his daughter fills in some of the details.

Supine, his blood pressure is 160/90 mm Hg and his heart rate is 75; immediately after standing up he feels dizzy and his blood pressure drops to 120/60 mm Hg with a heart rate of 110. Three months ago he weighed 155 pounds (70.3 kg); today he weighs 145 pounds (65.9 kg).

His neck veins are not distended. On chest auscultation, bibasilar coarse crackles are heard, as well as a systolic murmur (grade 2 on a scale of 6), loudest in the second intercostal space at the right parasternal border. No peripheral edema is detected. His Mini-Mental State Exam score is 22 out of 30.

What changes, if any, should be made in Mr. R.’s management? What advice should the primary care physician give Mr. R. and his daughter about the course of his heart failure?

THE IMPORTANCE OF COMPLETE CARE

Mr. R. has multiple convoluted medical issues that plague many elderly patients with heart failure. To provide optimal care to patients like him, physicians need to draw on knowledge from the fields of internal medicine, geriatrics, and cardiology.

In this paper, we discuss how diagnosing and managing heart failure is different in elderly patients. We emphasize the importance of complete care of frail elderly patients, highlighting the pharmacologic and nonpharmacologic interventions that are available. Finally, we will return to Mr. R. and discuss a comprehensive plan for him.

HEART FAILURE, FRAILTY, DISABILITY ARE ALL CONNECTED

The ability to bounce back from physical insults, chiefly medical illnesses, sharply declines in old age. As various stressors accumulate, physical deterioration becomes inevitable. While some older adults can avoid going down this path of morbidity, in an increasing number of frail elderly patients, congestive heart failure inescapably assumes a complicated course.

Frailty is a state of increased vulnerability to stressors due to age-related declines in physiologic reserve.1 Two elements intimately related to frailty are comorbidity and disability.

Fried et al2 analyzed data from more than 5,000 older men and women in the Cardiovascular Health Study and concluded that comorbidity (ie, having two or more chronic diseases) is a risk factor for frailty, which in turn results in disability, falls, hospitalizations, and death.

Figure 1.

The relationship between congestive heart failure and frailty is complex. Not only does heart failure itself result in frailty, but its multiple therapies can put additional stress on a frail patient. In addition, the heart failure and its treatments can negatively affect coexisting disorders (Figure 1).

BY THE NUMBERS

Heart failure is largely a disorder of the elderly, and as the US population ages, heart failure is rising in prevalence to epidemic numbers.3 The median age of patients admitted to the hospital because of heart failure is 75,4 and patients age 65 and older account for more than 75% of heart failure hospitalizations.5 Every year, in every 1,000 people over age 65, nearly 10 new cases of heart failure are diagnosed.6

Before age 70, men are affected more than women, but the opposite is true at age 70 and beyond. The reason for this reversal is that women live longer and have a better prognosis, as the cause of heart failure in most women is diastolic dysfunction secondary to hypertension rather than systolic dysfunction due to coronary artery disease, as in most men.7

Heart failure is costly and generally has a poor prognosis. The total cost of treating it reached a staggering $37.2 billion in 2009, and it was the leading cause of Medicare hospital admissions.6 Heart failure is the primary cause or a contributory cause of death in about 290,000 patients each year, and the rate of death at 1 year is an astonishing 1 in 5.6 The median survival time after diagnosis is 2.3 to 3.6 years in patients ages 67 to 74, and it is considerably shorter—1.1 to 1.6 years—in patients age 85 and older.8

 

 

THE BROKEN HEART

In 2005, the American College of Cardiology and the American Heart Association defined congestive heart failure as “a complex clinical syndrome that can result from any structural or functional cardiac disorder that impairs the ability of the ventricle to fill with or eject blood.”9 This characterization captures the intricate nature of the disease: its spectrum of symptoms, its many causes (eg, coronary artery disease, hypertension, nonischemic or idiopathic cardiomyopathy, and valvular heart disease), and the dual pathophysiologic features of systolic and diastolic impairment.

Systolic vs diastolic failure

Of the various ways of classifying heart failure, the most important is systolic vs diastolic.

The hallmark of systolic heart failure is a decreased left ventricular ejection fraction, and it is characterized by a large thin-walled ventricle that is weak and unable to eject enough blood to generate a normal cardiac output.

In contrast, the ejection fraction is normal or nearly normal in diastolic heart failure, but the end-diastolic volume is decreased because the ventricle is hypertrophied and thick-walled. The resultant chamber has become small and stiff and does not have enough volume for sufficient cardiac output.

QUIRKS IN THE HISTORY AND PHYSICAL EXAMINATION

The combination of inactivity and coexisting illnesses in a frail older adult may obscure some of the usual clinical manifestations of heart failure. While shortness of breath on mild exertion, easy fatigability, and leg swelling are common in younger heart failure patients, these symptoms may be due to normal aging in a much older patient. Let us consider some important aspects of the common signs and symptoms associated with heart failure.

Dyspnea on exertion is one of the earliest and most prominent symptoms. The usual question asked of patients to elicit whether this key manifestation is present is, “Do you get short of breath after walking a block?” However, this question may not be appropriate for a frail elderly person whose activity is restricted by comorbidities such as severe arthritis, coronary artery disease, or peripheral arterial disease. For a patient like this, ask instead if he or she gets short of breath after milder forms of exertion, such as making the bed, walking to the bathroom, or changing clothes.10 Also, keep in mind that dyspnea on exertion may be due to other conditions, such as renal failure, lung disease, depression, anemia, or deconditioning.

Orthopnea and paroxysmal nocturnal dyspnea may not be volunteered or elicited if a patient is sleeping in a chair or a recliner.

Leg swelling is less specific in older adults than in younger patients because chronic venous insufficiency is common in older people.

Weight gain almost always accompanies symptomatic heart failure but may also be due to increased appetite secondary to depression.

A change in mental status is common in elderly people with heart failure, especially those with vascular dementia with extensive cerebrovascular atherosclerosis or those who have latent Alzheimer disease.10

Cough, a symptom of a multitude of disorders, may be an early or the only manifestation of heart failure.

Pulmonary crackles are typically detected in most heart failure patients, but they may not be as characteristic in older adults, as they may also be noted in bronchitis, pneumonia, and other chronic lung diseases.

Additional symptoms to watch for include fatigue, syncope, angina, nocturia, and oliguria.

The bottom line is to integrate individual findings with other elements of the history and physical examination in diagnosing heart failure and tracking its progression.

CLINCHING THE DIAGNOSIS

Congestive heart failure is essentially a clinical diagnosis best established even before ordering tests, especially during times and situations in which these tests are not always readily available, such as outside office hours and in a long-term care setting.

A reliable and thorough history and physical examination is the most important component of the diagnostic process.

An echocardiogram is obtained next to measure the ejection fraction, which has both prognostic and therapeutic significance. Echocardiography can also uncover potential contributory cardiac structural abnormalities.

A chest radiograph is also typically obtained to look for pulmonary congestion, but in older adults its interpretation may be skewed by chronic lung disease or spinal deformities such as scoliosis and kyphosis.

The B-type natriuretic peptide (BNP) level is a popular blood test. BNP is commonly elevated in patients with heart failure. However, an elevated level in older adults should always be evaluated within the context of other clinical findings, as it can also result from advancing age and diseases other than heart failure, such as coronary artery disease, chronic pulmonary disease, pulmonary embolism, and renal insufficiency.11,12

PHARMACOTHERAPY PEARLS

Drug treatment for heart failure has evolved rapidly. Robust and sophisticated clinical trials have led to guidelines that call for specific medications. Unfortunately, older patients, particularly the very old and frail, have been poorly represented in these studies.9 Nonetheless, the type and choice of drugs for the young and old are similar.

Take into account age-associated changes in pharmacokinetics

Age-associated changes in pharmacokinetics must be taken into account when prescribing drugs for heart failure.13

Oral absorption of cardiovascular drugs is not significantly affected by the various changes that occur in older adults (eg, reduced gastric acid production, gastric emptying rate, gastrointestinal blood flow, and mobility). However, reductions in both lean body mass and total body water that come with aging result in lower volumes of distribution and higher plasma concentrations of hydrophilic drugs, most notably angiotensin-converting enzyme (ACE) inhibitors and digoxin. In contrast, the plasma concentrations of lipophilic drugs such as beta-blockers and central alpha-agonists tend to decrease as the proportion of body fat increases in older adults.

As the plasma albumin level diminishes with age, the free-drug concentration of salicylates and warfarin (Coumadin), which are extensively albumin-bound, may increase.

The serum concentrations of cardiovascular drugs metabolized in the liver—eg, propranolol (Inderal), lidocaine, labetalol (Trandate), verapamil (Calan), diltiazem (Cardizem), nitrates, and warfarin—may be elevated due to reduced hepatic blood flow, mass, volume, and overall metabolic capacity.

Declines in renal blood flow, glomerular filtration, and tubular function may cause accumulation of drugs that are excreted through the kidneys.

Beware of toxicities

The drug regimen for heart failure usually consists of multiple agents, including a beta-blocker or digoxin, and all have potential adverse effects. “Start low and go slow” must be the guiding principle. Drug-drug and drug-disease interactions must be anticipated, especially when drugs of multiple classes are used in a frail patient with coexisting illnesses. The prescriber’s vigilance for toxicities must be heightened.

Table 1 lists some of the drugs used in treating heart failure, common adverse affects to watch for, and recommendations for their use.

 

 

WIELDING THE SCALPEL

A tenet of heart failure management is to correct the underlying cardiac structural abnormality. This often calls for invasive intervention along with optimization of drug therapy.

For example:

  • Diseased coronary arteries may be amenable to revascularization, either by percutaneous coronary intervention or by the much more involved coronary artery bypass grafting, with the aim of enhancing cardiac function.
  • Valves can be repaired or replaced in patients with valvular heart disease.
  • A pacemaker can be implanted to remedy sick sinus syndrome, especially with concurrent use of heart-rate-lowering agents such as beta-blockers.
  • Placement of an implantable cardioverter-defibrillator has been found to be effective in preventing death due to ventricular tachyarrhythmias in patients with an ejection fraction of less than 30%.9
  • Cardiac resynchronization with a biventricular pacemaker may increase the ejection fraction and cardiac output by eliminating dyssynchronous contraction of the left and right ventricles.14

In frail older adults, consideration of these invasive therapies must be individualized. While procedures such as percutaneous coronary intervention and pacemaker placement may not be as physically taxing as bypass grafting or valve replacement, the potential for surgical complications must be seriously considered, particularly if the patient has diminished physiologic reserve. Case-to-case consideration is also crucial in cardioverter-defibrillator insertion, as the survival benefit may be diminished in older adults, who likely have coexisting illnesses that predispose them to die of a noncardiac cause.15,16

The bottom line is to contemplate multiple factors—severity of the heart failure, comorbidities, baseline functional status, and social support—when assessing the appropriateness of an invasive intervention.

BEYOND DRUGS AND DEVICES: WE CAN DO ‘MORE’

Much of the spotlight has been on the various drugs and devices used to treat heart failure, but of equal importance for frail elderly patients are complementary approaches that can be used to ease disease progression and boost the quality of life. The acronym MORE highlights these strategies.

M: Multidisciplinary management programs

Heart failure disease-management programs are designed to provide comprehensive multidisciplinary care across different settings (ie, home, outpatient, and inpatient) to high-risk patients who often have multiple medical, social, and behavioral issues.9 Interventions usually include intensive patient education, encouraging patients to be more aggressive participants in their care, closely monitoring patients through telephone follow-up or home nursing, carefully reviewing medications to improve adherence to evidence-based guidelines, and multidisciplinary care with nurse case management directed by a physician.

Studies have shown that management programs, which were largely nurse-directed and targeted at older adults and patients with advanced disease, can improve quality of life and functional status, decrease hospitalizations for both heart failure and other causes, and decrease medical costs.17–19

O: Other diseases

Fundamental to the care of the frail is to pay close attention to comorbidities, such as anemia, arthritis, and depression. Table 2 enumerates diseases that are common in the elderly and that may interact with heart failure.20–23

R: Restrictions

Specific limitations in the intake of certain dietary elements are a valuable adjunct in heart failure management.

Sodium intake should be restricted to less than 3 g/day by not adding salt to meals and by avoiding salt-rich foods (eg, canned and processed foods).24 During times of distressing volume overload, a tighter sodium limit of 2 g/day is necessary, and diuretics may be less effective if this restriction is not implemented.

Fluid restriction depends on the patient’s clinical status.25 While it is not necessary to limit fluid intake in the absence of retention, a limit of 2 L/day is recommended if edema is detected. If volume overload is severe, the limit should be 1 L/day.

Alcohol is a myocardial depressant that reduces the left ventricular ejection fraction.26 Abstinence is a must for patients with alcohol-induced heart failure; otherwise, a limit of 1 drink (8 oz of beer, 4 oz of wine, or 1 oz of hard liquor) per day is suggested.24

Calories and fat intake are both important to watch, particularly in patients with obesity, hyperlipidemia, hypertension, or coronary artery disease.

 

 

E: End-of-life issues

Usual causes of death in patients with heart failure include sudden cardiac death, arrhythmias, hypotension, end-organ hypoperfusion, and metabolic derangement.27,28

Given the life-limiting nature of the disease in frail older adults, it is very important for clinicians to discuss end-of-life matters with patients and their families as early as possible. Needed are effective communication skills that foster respect, empathy, and mutual understanding.

Advance directives. The primary task is to encourage patients to develop advance health directives. These are legal documents that represent patients’ preferences about interventions available toward the end of life such as do-not-resuscitate orders, appointment of surrogate decision-makers, and use of life-sustaining interventions (eg, a feeding tube, dialysis, blood transfusions). Establishing these directives early on will help ease the transition from one mode of care to another (eg, from acute care to hospice care), prevent pointless use of resources (eg, emergency room visits, hospital admissions), and ensure that the patient’s wishes are carried out.

Palliative measures that aim to alleviate suffering and promote quality of life and dignity are available for patients with severe symptoms. For varying degrees of dyspnea, diuretics, nitrates, morphine, and positive inotropic agents such as dobutamine (Dobutrex) and milrinone (Primacor) can be tried. Thoracentesis is done in patients with extensive pleural effusion. Fatigue and anorexia are due to a combination of factors, namely, decreased cardiac output, increased neurohormone levels, deconditioning, depression, decreased sleep, and anxiety.29 Opioids, caffeine, exercise, oxygen, fluid and salt restriction, and correction of anemia and depression may help ease these symptoms.

Apart from addressing the above issues, health care professionals should also provide emotional and spiritual support to the patient and family.

For patients with an implantable cardioverter-defibrillator, deactivation is an important matter that needs to be addressed. Deactivation can be carried out with certainty once the goal of care has shifted away from curative efforts and either the patient or a surrogate decision-maker has made the informed decision to turn the device off. Berger30 raised three points that the clinician and decision-maker can discuss in trying to achieve a resolution during times of doubt and indecision:

  • The patient may no longer value continued survival
  • The device may no longer offer the prospect of increased survival
  • The device may impede active dying.

The idea of hospice care should be gradually and gently explored to ensure a prompt and seamless transition when the time comes. The patient and family need to know that the goal of hospice care is to ensure comfort and that they can benefit the most by enrolling early during the course of the terminal illness.

The Medicare hospice benefit is granted to patients who have been certified by two physicians to have a life expectancy of 6 months or less if their terminal illness runs its natural course. The criteria for determining that heart failure is terminal are:

  • New York Heart Association class III (symptomatic with less than ordinary activities) or IV (symptomatic at rest)
  • Left ventricular ejection fraction less than or equal to 20%
  • Persistent symptoms despite optimal medical management
  • Inability to tolerate optional management due to hypotension with or without renal failure.31

WHAT CAN WE DO FOR MR. R.?

Mr. R. has systolic heart failure stemming from coronary artery disease, and his symptoms put him in New York Heart Association class III. He is well managed with drugs of different appropriate classes: an ACE inhibitor, a beta-blocker, digoxin, an aldosterone antagonist, and a diuretic. His other drugs all have well-defined indications.

Since he does not have fluid overload, his furosemide can be stopped, and this change will likely relieve his orthostatic hypotension and nocturia. His systolic blood pressure target can be liberalized to 150 mm Hg or less, as tighter control might exacerbate orthostatic hypotension. This change, along with having him start using a walker instead of a cane, will hopefully prevent future falls. Furthermore, his naproxen should be discontinued, as it can worsen heart failure.

Mr. R. has symptoms of depression and thus needs to be started on an antidepressant and encouraged to engage in social activities as much as he can tolerate. These interventions may also help with his mild dementia, which is evidenced by a Mini-Mental State Exam score of 22. He will not benefit from sodium and fat restriction, as he has actually been losing weight.

To keep Mr. R.’s cognitive impairment and overall decline in function from compromising his compliance with his treatment, he will need a substantial amount of assistance, which his daughter alone may not be able to provide. To tackle this concern, a discussion about participating in a heart failure management program can be started with Mr. R. and his family.

More importantly, his advanced directives, including delegating a surrogate decision-maker and deciding on do-not-resuscitate status, have to be clarified. Finally, it would be prudent to introduce the concept of hospice care to the patient and his daughter while he is still coherent and able to state his preferences.

References
  1. Walston J, Hadley EC, Ferruci L, et al. Research agenda for frailty in older adults: toward a better understanding of physiology and etiology: summary from the American Geriatrics Society/National Institute on Aging Research Conference on Frailty in Older Adults. J Am Geriatr Soc 2006; 54:9911001.
  2. Fried LP, Tangen CM, Walston J, et al. Frailty in older adults: evidence for a phenotype. J Gerontol A Biol Sci Med Sci 2001; 56:M146M156.
  3. Schocken DD, Arrieta MI, Leaverton PE, Ross EA. Prevalence and mortality rate of congestive heart failure in the United States. J Am Coll Cardiol 1992; 20:301306.
  4. Popovic JR, 1999 National Hospital Discharge Survey: annual summary with detailed diagnosis and procedure data. National Center for Health Statistics. Vital Health Stat 2001; 13:1206.
  5. DeFrances CJ, Hall MJ, Podgornik MN. 2003 National Hospital Discharge Survey. Advance data from vital and health statistics; no. 359. Hyattsville (MD): National Center for Health Statistics, 2005.
  6. American Heart Association. Heart disease and stroke statistics—2009 update: a report From the American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Circulation 2009; 119:e21e181.
  7. Levy D, Larson MG, Vasan RS, et al. The progression from hypertension to congestive heart failure. JAMA 1996; 275:15571562.
  8. Croft JB, Giles WH, Pollard RA, et al. Heart failure survival among older adults in the United States: a poor prognosis for an emerging epidemic in the Medicare population. Arch Intern Med 1999; 159:505510.
  9. Hunt SA, Abraham WT, Chin MH, et al. ACC/AHA 2005 guideline update for the diagnosis and management of chronic heart failure in the adult: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines (Writing Committee to Update the 2001 Guidelines for the Evaluation and Management of Heart Failure). Circulation 2005; 112:e154e235.
  10. Ahmed A. Clinical manifestations, diagnostic assessment, and etiology of heart failure in older adults. Clin Geriatr Med 2007; 23:1130.
  11. Redfield MM, Rodeheffer RJ, Jacobsen SJ, et al. Plasma brain natriuretic peptide concentration: impact of age and gender. J Am Coll Cardiol 2002; 40:976982.
  12. Wang TJ, Larson MG, Levy D, et al. Impact of age and sex on plasma natriuretic peptide levels in healthy adults. Am J Cardiol 2002; 90:254258.
  13. Aronow WS, Frishman WH, Cheng-Lai A. Cardiovascular drug therapy in the elderly. Cardiol Rev 2007; 15:195215.
  14. Bakker P, Meijburg H, de Bries J, et al. Biventricular pacing in end-stage heart failure improves functional capacity and left ventricular function. J Interv Card Electrophysiol 2000; 4:395404.
  15. Healey JS, Hallstrom AP, Kuck KH, et al. Role of the implantable defibrillator among elderly patients with a history of life-threatening ventricular arrhythmias. Eur Heart J 2007; 28:17461749.
  16. Lee DS, Tu JV, Austin PC, et al. Effect of cardiac and noncardiac conditions on survival after defibrillator implantation. J Am Coll Cardiol 2007; 49:24082415.
  17. Rich MW, Beckham V, Wittenberg C, et al. A multidisciplinary intervention to prevent the readmission of elderly patients with congestive heart failure. N Engl J Med 1995; 333:11901195.
  18. Fonarow GC, Stevenson LW, Walden JA, et al. Impact of a comprehensive heart failure management program on hospital readmission and functional status of patients with advanced heart failure. J Am Coll Cardiol 1997; 30:725732.
  19. McAlister F, Stewart S, Ferrua S, McMurray JJ. Multidisciplinary strategies for the management of heart failure patients at high risk for admission: a systematic review of randomized trials. J Am Coll Cardiol 2004; 44:810819.
  20. Horwich TB, Fonarow GC, Hamilton MA, et al. Anemia is associated with worse symptoms, greater impairment in functional capacity and a significant increase in mortality in patients with advanced heart failure. J Am Coll Cardiol 2002; 39:17801786.
  21. Al-Ahmad A, Rand WM, Manjunath G, et al. Reduced kidney function and anemia as risk factors for mortality in patients with left ventricular dysfunction. J Am Coll Cardiol 2001; 38:955962.
  22. Singh SN, Fisher SG, Deedwania PC, et al. Pulmonary effect of amiodarone in patients with heart failure: the Congestive Heart Failure-Survival Trial of Antiarrhythmic Therapy (CHF-STAT) Investigators (Veterans Affairs Cooperative Study No. 320). J Am Coll Cardiol 1997; 30:514517.
  23. Cohen MB, Mather PJ. A review of the association between congestive heart failure and cognitive impairment. Am J Geriatr Cardiol 2007; 16:171174.
  24. Dracup K, Baker DW, Dunbar SB, et al. Management of heart failure. II. Counseling, education, and lifestyle modifications. JAMA 1994; 272:14421446.
  25. Lenihan DJ, Uretsky BF. Non-pharmacologic treatment of heart failure in the elderly. Clin Geriatr Med 2000; 16:477488.
  26. Regan TJ. Alcohol and the cardiovascular system. JAMA 1990; 264:377381.
  27. Teuteberg JJ, Lewis EF, Nohria A, et al. Characteristics of patients who die with heart failure and a low ejection fraction in the new millennium. J Card Fail 2006; 12:4753.
  28. Derfler MC, Jacob M, Wolf RE, et al. Mode of death from congestive heart failure: implications for clinical management. Am J Geriatr Cardiol 2004; 13:299304.
  29. Evangelista LS, Moser DK, Westlake C, et al. Correlates of fatigue in patients with heart failure. Prog Cardiovasc Nurs 2008; 23:1217.
  30. Berger JT. The ethics of deactivating implanted cardioverter defibrillators. Ann Intern Med 2005; 142:631634.
  31. Stuart B, Connor S, Kinzbrunner BM, et al. Medical guidelines for determining prognosis in selected non-cancer diseases, 2nd ed. Arlington VA, National Hospice Organization; 1996.
Article PDF
Author and Disclosure Information

Renato V. Samala, MD, FACP
Department of Hospice and Palliative Medicine, Taussig Cancer Institute, Cleveland Clinic

Viviana Navas, MD
Department of Cardiovascular Medicine, Cleveland Clinic Florida, Weston

Emily Saluke, BS
St. George’s University School of Medicine, Great River, NY

Jerry O. Ciocon, MD
Department of Geriatrics, Cleveland Clinic Florida, Weston

Address: Renato Samala, MD, FACP, Taussig Cancer Institute, R35, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail rvsamala@yahoo.com

Issue
Cleveland Clinic Journal of Medicine - 78(12)
Publications
Topics
Page Number
837-845
Sections
Author and Disclosure Information

Renato V. Samala, MD, FACP
Department of Hospice and Palliative Medicine, Taussig Cancer Institute, Cleveland Clinic

Viviana Navas, MD
Department of Cardiovascular Medicine, Cleveland Clinic Florida, Weston

Emily Saluke, BS
St. George’s University School of Medicine, Great River, NY

Jerry O. Ciocon, MD
Department of Geriatrics, Cleveland Clinic Florida, Weston

Address: Renato Samala, MD, FACP, Taussig Cancer Institute, R35, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail rvsamala@yahoo.com

Author and Disclosure Information

Renato V. Samala, MD, FACP
Department of Hospice and Palliative Medicine, Taussig Cancer Institute, Cleveland Clinic

Viviana Navas, MD
Department of Cardiovascular Medicine, Cleveland Clinic Florida, Weston

Emily Saluke, BS
St. George’s University School of Medicine, Great River, NY

Jerry O. Ciocon, MD
Department of Geriatrics, Cleveland Clinic Florida, Weston

Address: Renato Samala, MD, FACP, Taussig Cancer Institute, R35, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail rvsamala@yahoo.com

Article PDF
Article PDF
Related Articles

Mr. R. is an 85-year-old with congestive heart failure; the last time his ejection fraction was measured it was 30%. He also has hypertension, coronary artery disease (for which he underwent triple-vessel coronary artery bypass grafting), osteoarthritis, hyperlipidemia, and chronic obstructive pulmonary disease. He currently takes lisinopril (Zestril), carvedilol (Coreg), aspirin, clopidogrel (Plavix), digoxin, simvastatin (Zocor), furosemide (Lasix), an albuterol inhaler (Proventil), and over-the-counter naproxen (Naprosyn), the last two taken as needed.

Accompanied by his daughter, Mr. R. comes to see his primary care physician for a routine follow-up visit. He says he feels fine and has no shortness of breath or chest pain, but he feels light-headed at times, especially when he gets out of bed. He also mentions that he is bothered with having to get up three to four times at night to urinate.

On further questioning, he relates that he uses a cane to walk around the house and gets short of breath when walking from his bed to the bathroom and from one room to the next. He can feed himself, but he needs assistance with bathing and getting dressed.

Mr. R. admits that he has been feeling lonely since his wife died about a year ago. He now lives with his daughter and her family, and they all get along well. His daughter mentions that over the last 6 months he has not been eating well, that he appears to have lost interest in doing some of the things that he used to enjoy, and that he has lost weight. She adds that he has fallen twice in the last month.

On physical examination, Mr. R. is without distress but appears weak. He answers all questions appropriately, although his affect is flat and his daughter fills in some of the details.

Supine, his blood pressure is 160/90 mm Hg and his heart rate is 75; immediately after standing up he feels dizzy and his blood pressure drops to 120/60 mm Hg with a heart rate of 110. Three months ago he weighed 155 pounds (70.3 kg); today he weighs 145 pounds (65.9 kg).

His neck veins are not distended. On chest auscultation, bibasilar coarse crackles are heard, as well as a systolic murmur (grade 2 on a scale of 6), loudest in the second intercostal space at the right parasternal border. No peripheral edema is detected. His Mini-Mental State Exam score is 22 out of 30.

What changes, if any, should be made in Mr. R.’s management? What advice should the primary care physician give Mr. R. and his daughter about the course of his heart failure?

THE IMPORTANCE OF COMPLETE CARE

Mr. R. has multiple convoluted medical issues that plague many elderly patients with heart failure. To provide optimal care to patients like him, physicians need to draw on knowledge from the fields of internal medicine, geriatrics, and cardiology.

In this paper, we discuss how diagnosing and managing heart failure is different in elderly patients. We emphasize the importance of complete care of frail elderly patients, highlighting the pharmacologic and nonpharmacologic interventions that are available. Finally, we will return to Mr. R. and discuss a comprehensive plan for him.

HEART FAILURE, FRAILTY, DISABILITY ARE ALL CONNECTED

The ability to bounce back from physical insults, chiefly medical illnesses, sharply declines in old age. As various stressors accumulate, physical deterioration becomes inevitable. While some older adults can avoid going down this path of morbidity, in an increasing number of frail elderly patients, congestive heart failure inescapably assumes a complicated course.

Frailty is a state of increased vulnerability to stressors due to age-related declines in physiologic reserve.1 Two elements intimately related to frailty are comorbidity and disability.

Fried et al2 analyzed data from more than 5,000 older men and women in the Cardiovascular Health Study and concluded that comorbidity (ie, having two or more chronic diseases) is a risk factor for frailty, which in turn results in disability, falls, hospitalizations, and death.

Figure 1.

The relationship between congestive heart failure and frailty is complex. Not only does heart failure itself result in frailty, but its multiple therapies can put additional stress on a frail patient. In addition, the heart failure and its treatments can negatively affect coexisting disorders (Figure 1).

BY THE NUMBERS

Heart failure is largely a disorder of the elderly, and as the US population ages, heart failure is rising in prevalence to epidemic numbers.3 The median age of patients admitted to the hospital because of heart failure is 75,4 and patients age 65 and older account for more than 75% of heart failure hospitalizations.5 Every year, in every 1,000 people over age 65, nearly 10 new cases of heart failure are diagnosed.6

Before age 70, men are affected more than women, but the opposite is true at age 70 and beyond. The reason for this reversal is that women live longer and have a better prognosis, as the cause of heart failure in most women is diastolic dysfunction secondary to hypertension rather than systolic dysfunction due to coronary artery disease, as in most men.7

Heart failure is costly and generally has a poor prognosis. The total cost of treating it reached a staggering $37.2 billion in 2009, and it was the leading cause of Medicare hospital admissions.6 Heart failure is the primary cause or a contributory cause of death in about 290,000 patients each year, and the rate of death at 1 year is an astonishing 1 in 5.6 The median survival time after diagnosis is 2.3 to 3.6 years in patients ages 67 to 74, and it is considerably shorter—1.1 to 1.6 years—in patients age 85 and older.8

 

 

THE BROKEN HEART

In 2005, the American College of Cardiology and the American Heart Association defined congestive heart failure as “a complex clinical syndrome that can result from any structural or functional cardiac disorder that impairs the ability of the ventricle to fill with or eject blood.”9 This characterization captures the intricate nature of the disease: its spectrum of symptoms, its many causes (eg, coronary artery disease, hypertension, nonischemic or idiopathic cardiomyopathy, and valvular heart disease), and the dual pathophysiologic features of systolic and diastolic impairment.

Systolic vs diastolic failure

Of the various ways of classifying heart failure, the most important is systolic vs diastolic.

The hallmark of systolic heart failure is a decreased left ventricular ejection fraction, and it is characterized by a large thin-walled ventricle that is weak and unable to eject enough blood to generate a normal cardiac output.

In contrast, the ejection fraction is normal or nearly normal in diastolic heart failure, but the end-diastolic volume is decreased because the ventricle is hypertrophied and thick-walled. The resultant chamber has become small and stiff and does not have enough volume for sufficient cardiac output.

QUIRKS IN THE HISTORY AND PHYSICAL EXAMINATION

The combination of inactivity and coexisting illnesses in a frail older adult may obscure some of the usual clinical manifestations of heart failure. While shortness of breath on mild exertion, easy fatigability, and leg swelling are common in younger heart failure patients, these symptoms may be due to normal aging in a much older patient. Let us consider some important aspects of the common signs and symptoms associated with heart failure.

Dyspnea on exertion is one of the earliest and most prominent symptoms. The usual question asked of patients to elicit whether this key manifestation is present is, “Do you get short of breath after walking a block?” However, this question may not be appropriate for a frail elderly person whose activity is restricted by comorbidities such as severe arthritis, coronary artery disease, or peripheral arterial disease. For a patient like this, ask instead if he or she gets short of breath after milder forms of exertion, such as making the bed, walking to the bathroom, or changing clothes.10 Also, keep in mind that dyspnea on exertion may be due to other conditions, such as renal failure, lung disease, depression, anemia, or deconditioning.

Orthopnea and paroxysmal nocturnal dyspnea may not be volunteered or elicited if a patient is sleeping in a chair or a recliner.

Leg swelling is less specific in older adults than in younger patients because chronic venous insufficiency is common in older people.

Weight gain almost always accompanies symptomatic heart failure but may also be due to increased appetite secondary to depression.

A change in mental status is common in elderly people with heart failure, especially those with vascular dementia with extensive cerebrovascular atherosclerosis or those who have latent Alzheimer disease.10

Cough, a symptom of a multitude of disorders, may be an early or the only manifestation of heart failure.

Pulmonary crackles are typically detected in most heart failure patients, but they may not be as characteristic in older adults, as they may also be noted in bronchitis, pneumonia, and other chronic lung diseases.

Additional symptoms to watch for include fatigue, syncope, angina, nocturia, and oliguria.

The bottom line is to integrate individual findings with other elements of the history and physical examination in diagnosing heart failure and tracking its progression.

CLINCHING THE DIAGNOSIS

Congestive heart failure is essentially a clinical diagnosis best established even before ordering tests, especially during times and situations in which these tests are not always readily available, such as outside office hours and in a long-term care setting.

A reliable and thorough history and physical examination is the most important component of the diagnostic process.

An echocardiogram is obtained next to measure the ejection fraction, which has both prognostic and therapeutic significance. Echocardiography can also uncover potential contributory cardiac structural abnormalities.

A chest radiograph is also typically obtained to look for pulmonary congestion, but in older adults its interpretation may be skewed by chronic lung disease or spinal deformities such as scoliosis and kyphosis.

The B-type natriuretic peptide (BNP) level is a popular blood test. BNP is commonly elevated in patients with heart failure. However, an elevated level in older adults should always be evaluated within the context of other clinical findings, as it can also result from advancing age and diseases other than heart failure, such as coronary artery disease, chronic pulmonary disease, pulmonary embolism, and renal insufficiency.11,12

PHARMACOTHERAPY PEARLS

Drug treatment for heart failure has evolved rapidly. Robust and sophisticated clinical trials have led to guidelines that call for specific medications. Unfortunately, older patients, particularly the very old and frail, have been poorly represented in these studies.9 Nonetheless, the type and choice of drugs for the young and old are similar.

Take into account age-associated changes in pharmacokinetics

Age-associated changes in pharmacokinetics must be taken into account when prescribing drugs for heart failure.13

Oral absorption of cardiovascular drugs is not significantly affected by the various changes that occur in older adults (eg, reduced gastric acid production, gastric emptying rate, gastrointestinal blood flow, and mobility). However, reductions in both lean body mass and total body water that come with aging result in lower volumes of distribution and higher plasma concentrations of hydrophilic drugs, most notably angiotensin-converting enzyme (ACE) inhibitors and digoxin. In contrast, the plasma concentrations of lipophilic drugs such as beta-blockers and central alpha-agonists tend to decrease as the proportion of body fat increases in older adults.

As the plasma albumin level diminishes with age, the free-drug concentration of salicylates and warfarin (Coumadin), which are extensively albumin-bound, may increase.

The serum concentrations of cardiovascular drugs metabolized in the liver—eg, propranolol (Inderal), lidocaine, labetalol (Trandate), verapamil (Calan), diltiazem (Cardizem), nitrates, and warfarin—may be elevated due to reduced hepatic blood flow, mass, volume, and overall metabolic capacity.

Declines in renal blood flow, glomerular filtration, and tubular function may cause accumulation of drugs that are excreted through the kidneys.

Beware of toxicities

The drug regimen for heart failure usually consists of multiple agents, including a beta-blocker or digoxin, and all have potential adverse effects. “Start low and go slow” must be the guiding principle. Drug-drug and drug-disease interactions must be anticipated, especially when drugs of multiple classes are used in a frail patient with coexisting illnesses. The prescriber’s vigilance for toxicities must be heightened.

Table 1 lists some of the drugs used in treating heart failure, common adverse affects to watch for, and recommendations for their use.

 

 

WIELDING THE SCALPEL

A tenet of heart failure management is to correct the underlying cardiac structural abnormality. This often calls for invasive intervention along with optimization of drug therapy.

For example:

  • Diseased coronary arteries may be amenable to revascularization, either by percutaneous coronary intervention or by the much more involved coronary artery bypass grafting, with the aim of enhancing cardiac function.
  • Valves can be repaired or replaced in patients with valvular heart disease.
  • A pacemaker can be implanted to remedy sick sinus syndrome, especially with concurrent use of heart-rate-lowering agents such as beta-blockers.
  • Placement of an implantable cardioverter-defibrillator has been found to be effective in preventing death due to ventricular tachyarrhythmias in patients with an ejection fraction of less than 30%.9
  • Cardiac resynchronization with a biventricular pacemaker may increase the ejection fraction and cardiac output by eliminating dyssynchronous contraction of the left and right ventricles.14

In frail older adults, consideration of these invasive therapies must be individualized. While procedures such as percutaneous coronary intervention and pacemaker placement may not be as physically taxing as bypass grafting or valve replacement, the potential for surgical complications must be seriously considered, particularly if the patient has diminished physiologic reserve. Case-to-case consideration is also crucial in cardioverter-defibrillator insertion, as the survival benefit may be diminished in older adults, who likely have coexisting illnesses that predispose them to die of a noncardiac cause.15,16

The bottom line is to contemplate multiple factors—severity of the heart failure, comorbidities, baseline functional status, and social support—when assessing the appropriateness of an invasive intervention.

BEYOND DRUGS AND DEVICES: WE CAN DO ‘MORE’

Much of the spotlight has been on the various drugs and devices used to treat heart failure, but of equal importance for frail elderly patients are complementary approaches that can be used to ease disease progression and boost the quality of life. The acronym MORE highlights these strategies.

M: Multidisciplinary management programs

Heart failure disease-management programs are designed to provide comprehensive multidisciplinary care across different settings (ie, home, outpatient, and inpatient) to high-risk patients who often have multiple medical, social, and behavioral issues.9 Interventions usually include intensive patient education, encouraging patients to be more aggressive participants in their care, closely monitoring patients through telephone follow-up or home nursing, carefully reviewing medications to improve adherence to evidence-based guidelines, and multidisciplinary care with nurse case management directed by a physician.

Studies have shown that management programs, which were largely nurse-directed and targeted at older adults and patients with advanced disease, can improve quality of life and functional status, decrease hospitalizations for both heart failure and other causes, and decrease medical costs.17–19

O: Other diseases

Fundamental to the care of the frail is to pay close attention to comorbidities, such as anemia, arthritis, and depression. Table 2 enumerates diseases that are common in the elderly and that may interact with heart failure.20–23

R: Restrictions

Specific limitations in the intake of certain dietary elements are a valuable adjunct in heart failure management.

Sodium intake should be restricted to less than 3 g/day by not adding salt to meals and by avoiding salt-rich foods (eg, canned and processed foods).24 During times of distressing volume overload, a tighter sodium limit of 2 g/day is necessary, and diuretics may be less effective if this restriction is not implemented.

Fluid restriction depends on the patient’s clinical status.25 While it is not necessary to limit fluid intake in the absence of retention, a limit of 2 L/day is recommended if edema is detected. If volume overload is severe, the limit should be 1 L/day.

Alcohol is a myocardial depressant that reduces the left ventricular ejection fraction.26 Abstinence is a must for patients with alcohol-induced heart failure; otherwise, a limit of 1 drink (8 oz of beer, 4 oz of wine, or 1 oz of hard liquor) per day is suggested.24

Calories and fat intake are both important to watch, particularly in patients with obesity, hyperlipidemia, hypertension, or coronary artery disease.

 

 

E: End-of-life issues

Usual causes of death in patients with heart failure include sudden cardiac death, arrhythmias, hypotension, end-organ hypoperfusion, and metabolic derangement.27,28

Given the life-limiting nature of the disease in frail older adults, it is very important for clinicians to discuss end-of-life matters with patients and their families as early as possible. Needed are effective communication skills that foster respect, empathy, and mutual understanding.

Advance directives. The primary task is to encourage patients to develop advance health directives. These are legal documents that represent patients’ preferences about interventions available toward the end of life such as do-not-resuscitate orders, appointment of surrogate decision-makers, and use of life-sustaining interventions (eg, a feeding tube, dialysis, blood transfusions). Establishing these directives early on will help ease the transition from one mode of care to another (eg, from acute care to hospice care), prevent pointless use of resources (eg, emergency room visits, hospital admissions), and ensure that the patient’s wishes are carried out.

Palliative measures that aim to alleviate suffering and promote quality of life and dignity are available for patients with severe symptoms. For varying degrees of dyspnea, diuretics, nitrates, morphine, and positive inotropic agents such as dobutamine (Dobutrex) and milrinone (Primacor) can be tried. Thoracentesis is done in patients with extensive pleural effusion. Fatigue and anorexia are due to a combination of factors, namely, decreased cardiac output, increased neurohormone levels, deconditioning, depression, decreased sleep, and anxiety.29 Opioids, caffeine, exercise, oxygen, fluid and salt restriction, and correction of anemia and depression may help ease these symptoms.

Apart from addressing the above issues, health care professionals should also provide emotional and spiritual support to the patient and family.

For patients with an implantable cardioverter-defibrillator, deactivation is an important matter that needs to be addressed. Deactivation can be carried out with certainty once the goal of care has shifted away from curative efforts and either the patient or a surrogate decision-maker has made the informed decision to turn the device off. Berger30 raised three points that the clinician and decision-maker can discuss in trying to achieve a resolution during times of doubt and indecision:

  • The patient may no longer value continued survival
  • The device may no longer offer the prospect of increased survival
  • The device may impede active dying.

The idea of hospice care should be gradually and gently explored to ensure a prompt and seamless transition when the time comes. The patient and family need to know that the goal of hospice care is to ensure comfort and that they can benefit the most by enrolling early during the course of the terminal illness.

The Medicare hospice benefit is granted to patients who have been certified by two physicians to have a life expectancy of 6 months or less if their terminal illness runs its natural course. The criteria for determining that heart failure is terminal are:

  • New York Heart Association class III (symptomatic with less than ordinary activities) or IV (symptomatic at rest)
  • Left ventricular ejection fraction less than or equal to 20%
  • Persistent symptoms despite optimal medical management
  • Inability to tolerate optional management due to hypotension with or without renal failure.31

WHAT CAN WE DO FOR MR. R.?

Mr. R. has systolic heart failure stemming from coronary artery disease, and his symptoms put him in New York Heart Association class III. He is well managed with drugs of different appropriate classes: an ACE inhibitor, a beta-blocker, digoxin, an aldosterone antagonist, and a diuretic. His other drugs all have well-defined indications.

Since he does not have fluid overload, his furosemide can be stopped, and this change will likely relieve his orthostatic hypotension and nocturia. His systolic blood pressure target can be liberalized to 150 mm Hg or less, as tighter control might exacerbate orthostatic hypotension. This change, along with having him start using a walker instead of a cane, will hopefully prevent future falls. Furthermore, his naproxen should be discontinued, as it can worsen heart failure.

Mr. R. has symptoms of depression and thus needs to be started on an antidepressant and encouraged to engage in social activities as much as he can tolerate. These interventions may also help with his mild dementia, which is evidenced by a Mini-Mental State Exam score of 22. He will not benefit from sodium and fat restriction, as he has actually been losing weight.

To keep Mr. R.’s cognitive impairment and overall decline in function from compromising his compliance with his treatment, he will need a substantial amount of assistance, which his daughter alone may not be able to provide. To tackle this concern, a discussion about participating in a heart failure management program can be started with Mr. R. and his family.

More importantly, his advanced directives, including delegating a surrogate decision-maker and deciding on do-not-resuscitate status, have to be clarified. Finally, it would be prudent to introduce the concept of hospice care to the patient and his daughter while he is still coherent and able to state his preferences.

Mr. R. is an 85-year-old with congestive heart failure; the last time his ejection fraction was measured it was 30%. He also has hypertension, coronary artery disease (for which he underwent triple-vessel coronary artery bypass grafting), osteoarthritis, hyperlipidemia, and chronic obstructive pulmonary disease. He currently takes lisinopril (Zestril), carvedilol (Coreg), aspirin, clopidogrel (Plavix), digoxin, simvastatin (Zocor), furosemide (Lasix), an albuterol inhaler (Proventil), and over-the-counter naproxen (Naprosyn), the last two taken as needed.

Accompanied by his daughter, Mr. R. comes to see his primary care physician for a routine follow-up visit. He says he feels fine and has no shortness of breath or chest pain, but he feels light-headed at times, especially when he gets out of bed. He also mentions that he is bothered with having to get up three to four times at night to urinate.

On further questioning, he relates that he uses a cane to walk around the house and gets short of breath when walking from his bed to the bathroom and from one room to the next. He can feed himself, but he needs assistance with bathing and getting dressed.

Mr. R. admits that he has been feeling lonely since his wife died about a year ago. He now lives with his daughter and her family, and they all get along well. His daughter mentions that over the last 6 months he has not been eating well, that he appears to have lost interest in doing some of the things that he used to enjoy, and that he has lost weight. She adds that he has fallen twice in the last month.

On physical examination, Mr. R. is without distress but appears weak. He answers all questions appropriately, although his affect is flat and his daughter fills in some of the details.

Supine, his blood pressure is 160/90 mm Hg and his heart rate is 75; immediately after standing up he feels dizzy and his blood pressure drops to 120/60 mm Hg with a heart rate of 110. Three months ago he weighed 155 pounds (70.3 kg); today he weighs 145 pounds (65.9 kg).

His neck veins are not distended. On chest auscultation, bibasilar coarse crackles are heard, as well as a systolic murmur (grade 2 on a scale of 6), loudest in the second intercostal space at the right parasternal border. No peripheral edema is detected. His Mini-Mental State Exam score is 22 out of 30.

What changes, if any, should be made in Mr. R.’s management? What advice should the primary care physician give Mr. R. and his daughter about the course of his heart failure?

THE IMPORTANCE OF COMPLETE CARE

Mr. R. has multiple convoluted medical issues that plague many elderly patients with heart failure. To provide optimal care to patients like him, physicians need to draw on knowledge from the fields of internal medicine, geriatrics, and cardiology.

In this paper, we discuss how diagnosing and managing heart failure is different in elderly patients. We emphasize the importance of complete care of frail elderly patients, highlighting the pharmacologic and nonpharmacologic interventions that are available. Finally, we will return to Mr. R. and discuss a comprehensive plan for him.

HEART FAILURE, FRAILTY, DISABILITY ARE ALL CONNECTED

The ability to bounce back from physical insults, chiefly medical illnesses, sharply declines in old age. As various stressors accumulate, physical deterioration becomes inevitable. While some older adults can avoid going down this path of morbidity, in an increasing number of frail elderly patients, congestive heart failure inescapably assumes a complicated course.

Frailty is a state of increased vulnerability to stressors due to age-related declines in physiologic reserve.1 Two elements intimately related to frailty are comorbidity and disability.

Fried et al2 analyzed data from more than 5,000 older men and women in the Cardiovascular Health Study and concluded that comorbidity (ie, having two or more chronic diseases) is a risk factor for frailty, which in turn results in disability, falls, hospitalizations, and death.

Figure 1.

The relationship between congestive heart failure and frailty is complex. Not only does heart failure itself result in frailty, but its multiple therapies can put additional stress on a frail patient. In addition, the heart failure and its treatments can negatively affect coexisting disorders (Figure 1).

BY THE NUMBERS

Heart failure is largely a disorder of the elderly, and as the US population ages, heart failure is rising in prevalence to epidemic numbers.3 The median age of patients admitted to the hospital because of heart failure is 75,4 and patients age 65 and older account for more than 75% of heart failure hospitalizations.5 Every year, in every 1,000 people over age 65, nearly 10 new cases of heart failure are diagnosed.6

Before age 70, men are affected more than women, but the opposite is true at age 70 and beyond. The reason for this reversal is that women live longer and have a better prognosis, as the cause of heart failure in most women is diastolic dysfunction secondary to hypertension rather than systolic dysfunction due to coronary artery disease, as in most men.7

Heart failure is costly and generally has a poor prognosis. The total cost of treating it reached a staggering $37.2 billion in 2009, and it was the leading cause of Medicare hospital admissions.6 Heart failure is the primary cause or a contributory cause of death in about 290,000 patients each year, and the rate of death at 1 year is an astonishing 1 in 5.6 The median survival time after diagnosis is 2.3 to 3.6 years in patients ages 67 to 74, and it is considerably shorter—1.1 to 1.6 years—in patients age 85 and older.8

 

 

THE BROKEN HEART

In 2005, the American College of Cardiology and the American Heart Association defined congestive heart failure as “a complex clinical syndrome that can result from any structural or functional cardiac disorder that impairs the ability of the ventricle to fill with or eject blood.”9 This characterization captures the intricate nature of the disease: its spectrum of symptoms, its many causes (eg, coronary artery disease, hypertension, nonischemic or idiopathic cardiomyopathy, and valvular heart disease), and the dual pathophysiologic features of systolic and diastolic impairment.

Systolic vs diastolic failure

Of the various ways of classifying heart failure, the most important is systolic vs diastolic.

The hallmark of systolic heart failure is a decreased left ventricular ejection fraction, and it is characterized by a large thin-walled ventricle that is weak and unable to eject enough blood to generate a normal cardiac output.

In contrast, the ejection fraction is normal or nearly normal in diastolic heart failure, but the end-diastolic volume is decreased because the ventricle is hypertrophied and thick-walled. The resultant chamber has become small and stiff and does not have enough volume for sufficient cardiac output.

QUIRKS IN THE HISTORY AND PHYSICAL EXAMINATION

The combination of inactivity and coexisting illnesses in a frail older adult may obscure some of the usual clinical manifestations of heart failure. While shortness of breath on mild exertion, easy fatigability, and leg swelling are common in younger heart failure patients, these symptoms may be due to normal aging in a much older patient. Let us consider some important aspects of the common signs and symptoms associated with heart failure.

Dyspnea on exertion is one of the earliest and most prominent symptoms. The usual question asked of patients to elicit whether this key manifestation is present is, “Do you get short of breath after walking a block?” However, this question may not be appropriate for a frail elderly person whose activity is restricted by comorbidities such as severe arthritis, coronary artery disease, or peripheral arterial disease. For a patient like this, ask instead if he or she gets short of breath after milder forms of exertion, such as making the bed, walking to the bathroom, or changing clothes.10 Also, keep in mind that dyspnea on exertion may be due to other conditions, such as renal failure, lung disease, depression, anemia, or deconditioning.

Orthopnea and paroxysmal nocturnal dyspnea may not be volunteered or elicited if a patient is sleeping in a chair or a recliner.

Leg swelling is less specific in older adults than in younger patients because chronic venous insufficiency is common in older people.

Weight gain almost always accompanies symptomatic heart failure but may also be due to increased appetite secondary to depression.

A change in mental status is common in elderly people with heart failure, especially those with vascular dementia with extensive cerebrovascular atherosclerosis or those who have latent Alzheimer disease.10

Cough, a symptom of a multitude of disorders, may be an early or the only manifestation of heart failure.

Pulmonary crackles are typically detected in most heart failure patients, but they may not be as characteristic in older adults, as they may also be noted in bronchitis, pneumonia, and other chronic lung diseases.

Additional symptoms to watch for include fatigue, syncope, angina, nocturia, and oliguria.

The bottom line is to integrate individual findings with other elements of the history and physical examination in diagnosing heart failure and tracking its progression.

CLINCHING THE DIAGNOSIS

Congestive heart failure is essentially a clinical diagnosis best established even before ordering tests, especially during times and situations in which these tests are not always readily available, such as outside office hours and in a long-term care setting.

A reliable and thorough history and physical examination is the most important component of the diagnostic process.

An echocardiogram is obtained next to measure the ejection fraction, which has both prognostic and therapeutic significance. Echocardiography can also uncover potential contributory cardiac structural abnormalities.

A chest radiograph is also typically obtained to look for pulmonary congestion, but in older adults its interpretation may be skewed by chronic lung disease or spinal deformities such as scoliosis and kyphosis.

The B-type natriuretic peptide (BNP) level is a popular blood test. BNP is commonly elevated in patients with heart failure. However, an elevated level in older adults should always be evaluated within the context of other clinical findings, as it can also result from advancing age and diseases other than heart failure, such as coronary artery disease, chronic pulmonary disease, pulmonary embolism, and renal insufficiency.11,12

PHARMACOTHERAPY PEARLS

Drug treatment for heart failure has evolved rapidly. Robust and sophisticated clinical trials have led to guidelines that call for specific medications. Unfortunately, older patients, particularly the very old and frail, have been poorly represented in these studies.9 Nonetheless, the type and choice of drugs for the young and old are similar.

Take into account age-associated changes in pharmacokinetics

Age-associated changes in pharmacokinetics must be taken into account when prescribing drugs for heart failure.13

Oral absorption of cardiovascular drugs is not significantly affected by the various changes that occur in older adults (eg, reduced gastric acid production, gastric emptying rate, gastrointestinal blood flow, and mobility). However, reductions in both lean body mass and total body water that come with aging result in lower volumes of distribution and higher plasma concentrations of hydrophilic drugs, most notably angiotensin-converting enzyme (ACE) inhibitors and digoxin. In contrast, the plasma concentrations of lipophilic drugs such as beta-blockers and central alpha-agonists tend to decrease as the proportion of body fat increases in older adults.

As the plasma albumin level diminishes with age, the free-drug concentration of salicylates and warfarin (Coumadin), which are extensively albumin-bound, may increase.

The serum concentrations of cardiovascular drugs metabolized in the liver—eg, propranolol (Inderal), lidocaine, labetalol (Trandate), verapamil (Calan), diltiazem (Cardizem), nitrates, and warfarin—may be elevated due to reduced hepatic blood flow, mass, volume, and overall metabolic capacity.

Declines in renal blood flow, glomerular filtration, and tubular function may cause accumulation of drugs that are excreted through the kidneys.

Beware of toxicities

The drug regimen for heart failure usually consists of multiple agents, including a beta-blocker or digoxin, and all have potential adverse effects. “Start low and go slow” must be the guiding principle. Drug-drug and drug-disease interactions must be anticipated, especially when drugs of multiple classes are used in a frail patient with coexisting illnesses. The prescriber’s vigilance for toxicities must be heightened.

Table 1 lists some of the drugs used in treating heart failure, common adverse affects to watch for, and recommendations for their use.

 

 

WIELDING THE SCALPEL

A tenet of heart failure management is to correct the underlying cardiac structural abnormality. This often calls for invasive intervention along with optimization of drug therapy.

For example:

  • Diseased coronary arteries may be amenable to revascularization, either by percutaneous coronary intervention or by the much more involved coronary artery bypass grafting, with the aim of enhancing cardiac function.
  • Valves can be repaired or replaced in patients with valvular heart disease.
  • A pacemaker can be implanted to remedy sick sinus syndrome, especially with concurrent use of heart-rate-lowering agents such as beta-blockers.
  • Placement of an implantable cardioverter-defibrillator has been found to be effective in preventing death due to ventricular tachyarrhythmias in patients with an ejection fraction of less than 30%.9
  • Cardiac resynchronization with a biventricular pacemaker may increase the ejection fraction and cardiac output by eliminating dyssynchronous contraction of the left and right ventricles.14

In frail older adults, consideration of these invasive therapies must be individualized. While procedures such as percutaneous coronary intervention and pacemaker placement may not be as physically taxing as bypass grafting or valve replacement, the potential for surgical complications must be seriously considered, particularly if the patient has diminished physiologic reserve. Case-to-case consideration is also crucial in cardioverter-defibrillator insertion, as the survival benefit may be diminished in older adults, who likely have coexisting illnesses that predispose them to die of a noncardiac cause.15,16

The bottom line is to contemplate multiple factors—severity of the heart failure, comorbidities, baseline functional status, and social support—when assessing the appropriateness of an invasive intervention.

BEYOND DRUGS AND DEVICES: WE CAN DO ‘MORE’

Much of the spotlight has been on the various drugs and devices used to treat heart failure, but of equal importance for frail elderly patients are complementary approaches that can be used to ease disease progression and boost the quality of life. The acronym MORE highlights these strategies.

M: Multidisciplinary management programs

Heart failure disease-management programs are designed to provide comprehensive multidisciplinary care across different settings (ie, home, outpatient, and inpatient) to high-risk patients who often have multiple medical, social, and behavioral issues.9 Interventions usually include intensive patient education, encouraging patients to be more aggressive participants in their care, closely monitoring patients through telephone follow-up or home nursing, carefully reviewing medications to improve adherence to evidence-based guidelines, and multidisciplinary care with nurse case management directed by a physician.

Studies have shown that management programs, which were largely nurse-directed and targeted at older adults and patients with advanced disease, can improve quality of life and functional status, decrease hospitalizations for both heart failure and other causes, and decrease medical costs.17–19

O: Other diseases

Fundamental to the care of the frail is to pay close attention to comorbidities, such as anemia, arthritis, and depression. Table 2 enumerates diseases that are common in the elderly and that may interact with heart failure.20–23

R: Restrictions

Specific limitations in the intake of certain dietary elements are a valuable adjunct in heart failure management.

Sodium intake should be restricted to less than 3 g/day by not adding salt to meals and by avoiding salt-rich foods (eg, canned and processed foods).24 During times of distressing volume overload, a tighter sodium limit of 2 g/day is necessary, and diuretics may be less effective if this restriction is not implemented.

Fluid restriction depends on the patient’s clinical status.25 While it is not necessary to limit fluid intake in the absence of retention, a limit of 2 L/day is recommended if edema is detected. If volume overload is severe, the limit should be 1 L/day.

Alcohol is a myocardial depressant that reduces the left ventricular ejection fraction.26 Abstinence is a must for patients with alcohol-induced heart failure; otherwise, a limit of 1 drink (8 oz of beer, 4 oz of wine, or 1 oz of hard liquor) per day is suggested.24

Calories and fat intake are both important to watch, particularly in patients with obesity, hyperlipidemia, hypertension, or coronary artery disease.

 

 

E: End-of-life issues

Usual causes of death in patients with heart failure include sudden cardiac death, arrhythmias, hypotension, end-organ hypoperfusion, and metabolic derangement.27,28

Given the life-limiting nature of the disease in frail older adults, it is very important for clinicians to discuss end-of-life matters with patients and their families as early as possible. Needed are effective communication skills that foster respect, empathy, and mutual understanding.

Advance directives. The primary task is to encourage patients to develop advance health directives. These are legal documents that represent patients’ preferences about interventions available toward the end of life such as do-not-resuscitate orders, appointment of surrogate decision-makers, and use of life-sustaining interventions (eg, a feeding tube, dialysis, blood transfusions). Establishing these directives early on will help ease the transition from one mode of care to another (eg, from acute care to hospice care), prevent pointless use of resources (eg, emergency room visits, hospital admissions), and ensure that the patient’s wishes are carried out.

Palliative measures that aim to alleviate suffering and promote quality of life and dignity are available for patients with severe symptoms. For varying degrees of dyspnea, diuretics, nitrates, morphine, and positive inotropic agents such as dobutamine (Dobutrex) and milrinone (Primacor) can be tried. Thoracentesis is done in patients with extensive pleural effusion. Fatigue and anorexia are due to a combination of factors, namely, decreased cardiac output, increased neurohormone levels, deconditioning, depression, decreased sleep, and anxiety.29 Opioids, caffeine, exercise, oxygen, fluid and salt restriction, and correction of anemia and depression may help ease these symptoms.

Apart from addressing the above issues, health care professionals should also provide emotional and spiritual support to the patient and family.

For patients with an implantable cardioverter-defibrillator, deactivation is an important matter that needs to be addressed. Deactivation can be carried out with certainty once the goal of care has shifted away from curative efforts and either the patient or a surrogate decision-maker has made the informed decision to turn the device off. Berger30 raised three points that the clinician and decision-maker can discuss in trying to achieve a resolution during times of doubt and indecision:

  • The patient may no longer value continued survival
  • The device may no longer offer the prospect of increased survival
  • The device may impede active dying.

The idea of hospice care should be gradually and gently explored to ensure a prompt and seamless transition when the time comes. The patient and family need to know that the goal of hospice care is to ensure comfort and that they can benefit the most by enrolling early during the course of the terminal illness.

The Medicare hospice benefit is granted to patients who have been certified by two physicians to have a life expectancy of 6 months or less if their terminal illness runs its natural course. The criteria for determining that heart failure is terminal are:

  • New York Heart Association class III (symptomatic with less than ordinary activities) or IV (symptomatic at rest)
  • Left ventricular ejection fraction less than or equal to 20%
  • Persistent symptoms despite optimal medical management
  • Inability to tolerate optional management due to hypotension with or without renal failure.31

WHAT CAN WE DO FOR MR. R.?

Mr. R. has systolic heart failure stemming from coronary artery disease, and his symptoms put him in New York Heart Association class III. He is well managed with drugs of different appropriate classes: an ACE inhibitor, a beta-blocker, digoxin, an aldosterone antagonist, and a diuretic. His other drugs all have well-defined indications.

Since he does not have fluid overload, his furosemide can be stopped, and this change will likely relieve his orthostatic hypotension and nocturia. His systolic blood pressure target can be liberalized to 150 mm Hg or less, as tighter control might exacerbate orthostatic hypotension. This change, along with having him start using a walker instead of a cane, will hopefully prevent future falls. Furthermore, his naproxen should be discontinued, as it can worsen heart failure.

Mr. R. has symptoms of depression and thus needs to be started on an antidepressant and encouraged to engage in social activities as much as he can tolerate. These interventions may also help with his mild dementia, which is evidenced by a Mini-Mental State Exam score of 22. He will not benefit from sodium and fat restriction, as he has actually been losing weight.

To keep Mr. R.’s cognitive impairment and overall decline in function from compromising his compliance with his treatment, he will need a substantial amount of assistance, which his daughter alone may not be able to provide. To tackle this concern, a discussion about participating in a heart failure management program can be started with Mr. R. and his family.

More importantly, his advanced directives, including delegating a surrogate decision-maker and deciding on do-not-resuscitate status, have to be clarified. Finally, it would be prudent to introduce the concept of hospice care to the patient and his daughter while he is still coherent and able to state his preferences.

References
  1. Walston J, Hadley EC, Ferruci L, et al. Research agenda for frailty in older adults: toward a better understanding of physiology and etiology: summary from the American Geriatrics Society/National Institute on Aging Research Conference on Frailty in Older Adults. J Am Geriatr Soc 2006; 54:9911001.
  2. Fried LP, Tangen CM, Walston J, et al. Frailty in older adults: evidence for a phenotype. J Gerontol A Biol Sci Med Sci 2001; 56:M146M156.
  3. Schocken DD, Arrieta MI, Leaverton PE, Ross EA. Prevalence and mortality rate of congestive heart failure in the United States. J Am Coll Cardiol 1992; 20:301306.
  4. Popovic JR, 1999 National Hospital Discharge Survey: annual summary with detailed diagnosis and procedure data. National Center for Health Statistics. Vital Health Stat 2001; 13:1206.
  5. DeFrances CJ, Hall MJ, Podgornik MN. 2003 National Hospital Discharge Survey. Advance data from vital and health statistics; no. 359. Hyattsville (MD): National Center for Health Statistics, 2005.
  6. American Heart Association. Heart disease and stroke statistics—2009 update: a report From the American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Circulation 2009; 119:e21e181.
  7. Levy D, Larson MG, Vasan RS, et al. The progression from hypertension to congestive heart failure. JAMA 1996; 275:15571562.
  8. Croft JB, Giles WH, Pollard RA, et al. Heart failure survival among older adults in the United States: a poor prognosis for an emerging epidemic in the Medicare population. Arch Intern Med 1999; 159:505510.
  9. Hunt SA, Abraham WT, Chin MH, et al. ACC/AHA 2005 guideline update for the diagnosis and management of chronic heart failure in the adult: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines (Writing Committee to Update the 2001 Guidelines for the Evaluation and Management of Heart Failure). Circulation 2005; 112:e154e235.
  10. Ahmed A. Clinical manifestations, diagnostic assessment, and etiology of heart failure in older adults. Clin Geriatr Med 2007; 23:1130.
  11. Redfield MM, Rodeheffer RJ, Jacobsen SJ, et al. Plasma brain natriuretic peptide concentration: impact of age and gender. J Am Coll Cardiol 2002; 40:976982.
  12. Wang TJ, Larson MG, Levy D, et al. Impact of age and sex on plasma natriuretic peptide levels in healthy adults. Am J Cardiol 2002; 90:254258.
  13. Aronow WS, Frishman WH, Cheng-Lai A. Cardiovascular drug therapy in the elderly. Cardiol Rev 2007; 15:195215.
  14. Bakker P, Meijburg H, de Bries J, et al. Biventricular pacing in end-stage heart failure improves functional capacity and left ventricular function. J Interv Card Electrophysiol 2000; 4:395404.
  15. Healey JS, Hallstrom AP, Kuck KH, et al. Role of the implantable defibrillator among elderly patients with a history of life-threatening ventricular arrhythmias. Eur Heart J 2007; 28:17461749.
  16. Lee DS, Tu JV, Austin PC, et al. Effect of cardiac and noncardiac conditions on survival after defibrillator implantation. J Am Coll Cardiol 2007; 49:24082415.
  17. Rich MW, Beckham V, Wittenberg C, et al. A multidisciplinary intervention to prevent the readmission of elderly patients with congestive heart failure. N Engl J Med 1995; 333:11901195.
  18. Fonarow GC, Stevenson LW, Walden JA, et al. Impact of a comprehensive heart failure management program on hospital readmission and functional status of patients with advanced heart failure. J Am Coll Cardiol 1997; 30:725732.
  19. McAlister F, Stewart S, Ferrua S, McMurray JJ. Multidisciplinary strategies for the management of heart failure patients at high risk for admission: a systematic review of randomized trials. J Am Coll Cardiol 2004; 44:810819.
  20. Horwich TB, Fonarow GC, Hamilton MA, et al. Anemia is associated with worse symptoms, greater impairment in functional capacity and a significant increase in mortality in patients with advanced heart failure. J Am Coll Cardiol 2002; 39:17801786.
  21. Al-Ahmad A, Rand WM, Manjunath G, et al. Reduced kidney function and anemia as risk factors for mortality in patients with left ventricular dysfunction. J Am Coll Cardiol 2001; 38:955962.
  22. Singh SN, Fisher SG, Deedwania PC, et al. Pulmonary effect of amiodarone in patients with heart failure: the Congestive Heart Failure-Survival Trial of Antiarrhythmic Therapy (CHF-STAT) Investigators (Veterans Affairs Cooperative Study No. 320). J Am Coll Cardiol 1997; 30:514517.
  23. Cohen MB, Mather PJ. A review of the association between congestive heart failure and cognitive impairment. Am J Geriatr Cardiol 2007; 16:171174.
  24. Dracup K, Baker DW, Dunbar SB, et al. Management of heart failure. II. Counseling, education, and lifestyle modifications. JAMA 1994; 272:14421446.
  25. Lenihan DJ, Uretsky BF. Non-pharmacologic treatment of heart failure in the elderly. Clin Geriatr Med 2000; 16:477488.
  26. Regan TJ. Alcohol and the cardiovascular system. JAMA 1990; 264:377381.
  27. Teuteberg JJ, Lewis EF, Nohria A, et al. Characteristics of patients who die with heart failure and a low ejection fraction in the new millennium. J Card Fail 2006; 12:4753.
  28. Derfler MC, Jacob M, Wolf RE, et al. Mode of death from congestive heart failure: implications for clinical management. Am J Geriatr Cardiol 2004; 13:299304.
  29. Evangelista LS, Moser DK, Westlake C, et al. Correlates of fatigue in patients with heart failure. Prog Cardiovasc Nurs 2008; 23:1217.
  30. Berger JT. The ethics of deactivating implanted cardioverter defibrillators. Ann Intern Med 2005; 142:631634.
  31. Stuart B, Connor S, Kinzbrunner BM, et al. Medical guidelines for determining prognosis in selected non-cancer diseases, 2nd ed. Arlington VA, National Hospice Organization; 1996.
References
  1. Walston J, Hadley EC, Ferruci L, et al. Research agenda for frailty in older adults: toward a better understanding of physiology and etiology: summary from the American Geriatrics Society/National Institute on Aging Research Conference on Frailty in Older Adults. J Am Geriatr Soc 2006; 54:9911001.
  2. Fried LP, Tangen CM, Walston J, et al. Frailty in older adults: evidence for a phenotype. J Gerontol A Biol Sci Med Sci 2001; 56:M146M156.
  3. Schocken DD, Arrieta MI, Leaverton PE, Ross EA. Prevalence and mortality rate of congestive heart failure in the United States. J Am Coll Cardiol 1992; 20:301306.
  4. Popovic JR, 1999 National Hospital Discharge Survey: annual summary with detailed diagnosis and procedure data. National Center for Health Statistics. Vital Health Stat 2001; 13:1206.
  5. DeFrances CJ, Hall MJ, Podgornik MN. 2003 National Hospital Discharge Survey. Advance data from vital and health statistics; no. 359. Hyattsville (MD): National Center for Health Statistics, 2005.
  6. American Heart Association. Heart disease and stroke statistics—2009 update: a report From the American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Circulation 2009; 119:e21e181.
  7. Levy D, Larson MG, Vasan RS, et al. The progression from hypertension to congestive heart failure. JAMA 1996; 275:15571562.
  8. Croft JB, Giles WH, Pollard RA, et al. Heart failure survival among older adults in the United States: a poor prognosis for an emerging epidemic in the Medicare population. Arch Intern Med 1999; 159:505510.
  9. Hunt SA, Abraham WT, Chin MH, et al. ACC/AHA 2005 guideline update for the diagnosis and management of chronic heart failure in the adult: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines (Writing Committee to Update the 2001 Guidelines for the Evaluation and Management of Heart Failure). Circulation 2005; 112:e154e235.
  10. Ahmed A. Clinical manifestations, diagnostic assessment, and etiology of heart failure in older adults. Clin Geriatr Med 2007; 23:1130.
  11. Redfield MM, Rodeheffer RJ, Jacobsen SJ, et al. Plasma brain natriuretic peptide concentration: impact of age and gender. J Am Coll Cardiol 2002; 40:976982.
  12. Wang TJ, Larson MG, Levy D, et al. Impact of age and sex on plasma natriuretic peptide levels in healthy adults. Am J Cardiol 2002; 90:254258.
  13. Aronow WS, Frishman WH, Cheng-Lai A. Cardiovascular drug therapy in the elderly. Cardiol Rev 2007; 15:195215.
  14. Bakker P, Meijburg H, de Bries J, et al. Biventricular pacing in end-stage heart failure improves functional capacity and left ventricular function. J Interv Card Electrophysiol 2000; 4:395404.
  15. Healey JS, Hallstrom AP, Kuck KH, et al. Role of the implantable defibrillator among elderly patients with a history of life-threatening ventricular arrhythmias. Eur Heart J 2007; 28:17461749.
  16. Lee DS, Tu JV, Austin PC, et al. Effect of cardiac and noncardiac conditions on survival after defibrillator implantation. J Am Coll Cardiol 2007; 49:24082415.
  17. Rich MW, Beckham V, Wittenberg C, et al. A multidisciplinary intervention to prevent the readmission of elderly patients with congestive heart failure. N Engl J Med 1995; 333:11901195.
  18. Fonarow GC, Stevenson LW, Walden JA, et al. Impact of a comprehensive heart failure management program on hospital readmission and functional status of patients with advanced heart failure. J Am Coll Cardiol 1997; 30:725732.
  19. McAlister F, Stewart S, Ferrua S, McMurray JJ. Multidisciplinary strategies for the management of heart failure patients at high risk for admission: a systematic review of randomized trials. J Am Coll Cardiol 2004; 44:810819.
  20. Horwich TB, Fonarow GC, Hamilton MA, et al. Anemia is associated with worse symptoms, greater impairment in functional capacity and a significant increase in mortality in patients with advanced heart failure. J Am Coll Cardiol 2002; 39:17801786.
  21. Al-Ahmad A, Rand WM, Manjunath G, et al. Reduced kidney function and anemia as risk factors for mortality in patients with left ventricular dysfunction. J Am Coll Cardiol 2001; 38:955962.
  22. Singh SN, Fisher SG, Deedwania PC, et al. Pulmonary effect of amiodarone in patients with heart failure: the Congestive Heart Failure-Survival Trial of Antiarrhythmic Therapy (CHF-STAT) Investigators (Veterans Affairs Cooperative Study No. 320). J Am Coll Cardiol 1997; 30:514517.
  23. Cohen MB, Mather PJ. A review of the association between congestive heart failure and cognitive impairment. Am J Geriatr Cardiol 2007; 16:171174.
  24. Dracup K, Baker DW, Dunbar SB, et al. Management of heart failure. II. Counseling, education, and lifestyle modifications. JAMA 1994; 272:14421446.
  25. Lenihan DJ, Uretsky BF. Non-pharmacologic treatment of heart failure in the elderly. Clin Geriatr Med 2000; 16:477488.
  26. Regan TJ. Alcohol and the cardiovascular system. JAMA 1990; 264:377381.
  27. Teuteberg JJ, Lewis EF, Nohria A, et al. Characteristics of patients who die with heart failure and a low ejection fraction in the new millennium. J Card Fail 2006; 12:4753.
  28. Derfler MC, Jacob M, Wolf RE, et al. Mode of death from congestive heart failure: implications for clinical management. Am J Geriatr Cardiol 2004; 13:299304.
  29. Evangelista LS, Moser DK, Westlake C, et al. Correlates of fatigue in patients with heart failure. Prog Cardiovasc Nurs 2008; 23:1217.
  30. Berger JT. The ethics of deactivating implanted cardioverter defibrillators. Ann Intern Med 2005; 142:631634.
  31. Stuart B, Connor S, Kinzbrunner BM, et al. Medical guidelines for determining prognosis in selected non-cancer diseases, 2nd ed. Arlington VA, National Hospice Organization; 1996.
Issue
Cleveland Clinic Journal of Medicine - 78(12)
Issue
Cleveland Clinic Journal of Medicine - 78(12)
Page Number
837-845
Page Number
837-845
Publications
Publications
Topics
Article Type
Display Headline
Heart failure in frail, older patients: We can do ‘MORE’
Display Headline
Heart failure in frail, older patients: We can do ‘MORE’
Sections
Inside the Article

KEY POINTS

  • Not only does heart failure itself result in frailty, but its treatment can also put additional stress on an already frail patient. In addition, the illness and its treatments can negatively affect coexisting disorders.
  • Common signs and symptoms of heart failure are less specific in older adults, and atypical symptoms may predominate.
  • Age-associated changes in pharmacokinetics must be taken into account when prescribing drugs for heart failure.
  • Effective communication among health professionals, patients, and families is necessary.
  • Given the life-limiting nature of heart failure in frail older adults, it is critical for clinicians to discuss end-of-life issues with patients and their families as soon as possible.
Disallow All Ads
Alternative CME
Article PDF Media

Progressive Multifocal Leukoencephalopathy in the Biologic Era: Implications for Practice

Article Type
Changed
Wed, 04/10/2019 - 11:28
Display Headline
Progressive Multifocal Leukoencephalopathy in the Biologic Era: Implications for Practice
Article PDF
Issue
Cleveland Clinic Journal of Medicine - 78(11)
Publications
Topics
Page Number
S1-S41
Sections
Article PDF
Article PDF
Issue
Cleveland Clinic Journal of Medicine - 78(11)
Issue
Cleveland Clinic Journal of Medicine - 78(11)
Page Number
S1-S41
Page Number
S1-S41
Publications
Publications
Topics
Article Type
Display Headline
Progressive Multifocal Leukoencephalopathy in the Biologic Era: Implications for Practice
Display Headline
Progressive Multifocal Leukoencephalopathy in the Biologic Era: Implications for Practice
Sections
Citation Override
Cleveland Clinic Journal of Medicine 2011 November;78(11 suppl 2):S1-S41
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

Introduction: Progressive multifocal leukoencephalopathy in the biologic era

Article Type
Changed
Tue, 10/02/2018 - 08:47
Display Headline
Introduction: Progressive multifocal leukoencephalopathy in the biologic era

The epidemiology of progressive multifocal leukoencephalopathy (PML) has evolved in recent years. Until the availability of natalizumab, PML was seen primarily by physicians who treated patients with human immunodeficiency virus (HIV) infection. This situation changed in 2005 when PML was first associated with natalizumab therapy in patients with multiple sclerosis (MS). This discovery focused attention and investigative resources on the relationship between PML and natalizumab, and subsequently on other biologically based therapies. In the years since, we have learned about the pathogenesis of PML and developed concepts of risk mitigation.

Increasingly selective, potent, and innovative biologically based immune-based therapeutics have led to enhanced therapeutic options, but also to more surprises, with PML being observed in unexpected patient populations. The MS community and clinicians who routinely use biologic therapies are alert to this disease and its implications for patients. PML remains rare enough, however, that community-based clinicians may be less attuned to the impact and risks of these therapies.

This Cleveland Clinic Journal of Medicine supplement addresses the issues of awareness, recognition, and management of PML for clinicians whose patients may be at risk, including those in the fields of infectious disease, neurology, oncology, and rheumatology. The supplement provides an overview of the pathogenesis and clinical picture of PML, its evolving epidemiology, and the current approaches to its management.

The articles and their accompanying discussions are based on a roundtable held at Cleveland Clinic on January 31, 2011. The roundtable’s expert faculty contributed insights from several different perspectives, including laboratory research on JC virus–induced demyelinization in PML; front-line experience starting in the early 1980s when PML was understood to be an HIV-related disease and continuing to its current status as a potential complication of biologic therapy; and management of a large pharmaceutical safety program that monitors and facilitates reporting of potential fatal drug side effects.

Readers of these articles will acquire understanding of the history and clinical picture of PML, appreciation of the influence of biologic therapies in several specialties, and enhanced awareness of when to consider PML and what actions to take when the diagnosis is a possibility.

Article PDF
Author and Disclosure Information

Leonard Calabrese, DO
Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University; R.J. Fasenmyer Chair of Clinical Immunology, Department of Rheumatic and Immunologic Diseases, Orthopaedic & Rheumatologic Institute, Cleveland Clinic, Cleveland, OH

Publications
Page Number
S2
Author and Disclosure Information

Leonard Calabrese, DO
Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University; R.J. Fasenmyer Chair of Clinical Immunology, Department of Rheumatic and Immunologic Diseases, Orthopaedic & Rheumatologic Institute, Cleveland Clinic, Cleveland, OH

Author and Disclosure Information

Leonard Calabrese, DO
Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University; R.J. Fasenmyer Chair of Clinical Immunology, Department of Rheumatic and Immunologic Diseases, Orthopaedic & Rheumatologic Institute, Cleveland Clinic, Cleveland, OH

Article PDF
Article PDF

The epidemiology of progressive multifocal leukoencephalopathy (PML) has evolved in recent years. Until the availability of natalizumab, PML was seen primarily by physicians who treated patients with human immunodeficiency virus (HIV) infection. This situation changed in 2005 when PML was first associated with natalizumab therapy in patients with multiple sclerosis (MS). This discovery focused attention and investigative resources on the relationship between PML and natalizumab, and subsequently on other biologically based therapies. In the years since, we have learned about the pathogenesis of PML and developed concepts of risk mitigation.

Increasingly selective, potent, and innovative biologically based immune-based therapeutics have led to enhanced therapeutic options, but also to more surprises, with PML being observed in unexpected patient populations. The MS community and clinicians who routinely use biologic therapies are alert to this disease and its implications for patients. PML remains rare enough, however, that community-based clinicians may be less attuned to the impact and risks of these therapies.

This Cleveland Clinic Journal of Medicine supplement addresses the issues of awareness, recognition, and management of PML for clinicians whose patients may be at risk, including those in the fields of infectious disease, neurology, oncology, and rheumatology. The supplement provides an overview of the pathogenesis and clinical picture of PML, its evolving epidemiology, and the current approaches to its management.

The articles and their accompanying discussions are based on a roundtable held at Cleveland Clinic on January 31, 2011. The roundtable’s expert faculty contributed insights from several different perspectives, including laboratory research on JC virus–induced demyelinization in PML; front-line experience starting in the early 1980s when PML was understood to be an HIV-related disease and continuing to its current status as a potential complication of biologic therapy; and management of a large pharmaceutical safety program that monitors and facilitates reporting of potential fatal drug side effects.

Readers of these articles will acquire understanding of the history and clinical picture of PML, appreciation of the influence of biologic therapies in several specialties, and enhanced awareness of when to consider PML and what actions to take when the diagnosis is a possibility.

The epidemiology of progressive multifocal leukoencephalopathy (PML) has evolved in recent years. Until the availability of natalizumab, PML was seen primarily by physicians who treated patients with human immunodeficiency virus (HIV) infection. This situation changed in 2005 when PML was first associated with natalizumab therapy in patients with multiple sclerosis (MS). This discovery focused attention and investigative resources on the relationship between PML and natalizumab, and subsequently on other biologically based therapies. In the years since, we have learned about the pathogenesis of PML and developed concepts of risk mitigation.

Increasingly selective, potent, and innovative biologically based immune-based therapeutics have led to enhanced therapeutic options, but also to more surprises, with PML being observed in unexpected patient populations. The MS community and clinicians who routinely use biologic therapies are alert to this disease and its implications for patients. PML remains rare enough, however, that community-based clinicians may be less attuned to the impact and risks of these therapies.

This Cleveland Clinic Journal of Medicine supplement addresses the issues of awareness, recognition, and management of PML for clinicians whose patients may be at risk, including those in the fields of infectious disease, neurology, oncology, and rheumatology. The supplement provides an overview of the pathogenesis and clinical picture of PML, its evolving epidemiology, and the current approaches to its management.

The articles and their accompanying discussions are based on a roundtable held at Cleveland Clinic on January 31, 2011. The roundtable’s expert faculty contributed insights from several different perspectives, including laboratory research on JC virus–induced demyelinization in PML; front-line experience starting in the early 1980s when PML was understood to be an HIV-related disease and continuing to its current status as a potential complication of biologic therapy; and management of a large pharmaceutical safety program that monitors and facilitates reporting of potential fatal drug side effects.

Readers of these articles will acquire understanding of the history and clinical picture of PML, appreciation of the influence of biologic therapies in several specialties, and enhanced awareness of when to consider PML and what actions to take when the diagnosis is a possibility.

Page Number
S2
Page Number
S2
Publications
Publications
Article Type
Display Headline
Introduction: Progressive multifocal leukoencephalopathy in the biologic era
Display Headline
Introduction: Progressive multifocal leukoencephalopathy in the biologic era
Citation Override
Cleveland Clinic Journal of Medicine 2011 November;78(suppl 2):S2
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media

History and current concepts in the pathogenesis of PML

Article Type
Changed
Tue, 10/02/2018 - 08:47
Display Headline
History and current concepts in the pathogenesis of PML

The neuropathology of progressive multifocal leukoencephalopathy (PML) was first reported in 1958 following examination of brain tissue from two cases of chronic lymphocytic leukemia and one case of Hodgkin lymphoma.1 The classic triad of symptoms of PML—cognitive impairment, visual deficits, and motor dysfunction—had been observed previously but had not been formally described.2

Until PML was discovered in patients with autoimmune diseases treated with biologic therapies that do not directly suppress immunity, PML had been considered a very rare, virus-induced demyelinating disease of the white matter that occurred in immune-compromised patients. The incidence of PML rose sharply in the mid-1980s with the pandemic of human immunodeficiency virus (HIV)-1 infection and continues as an acquired immunodeficiency syndrome–defining illness at a rate of approximately 1% to 3% of HIV-1 seropositive individuals; more recently, it has been seen in approximately 1 in 850 natalizumab-treated individuals who have multiple sclerosis (MS). The incidence of PML in natalizumab-treated MS patients increases with dosing; among those who receive 24 or more doses, the incidence is 1 in 400.

The cause of PML was unknown until 1971, when viral particles were observed by electron microscopy in PML brain lesions and subsequently isolated at the University of Wisconsin, Madison, in cultures of human fetal brain tissue.3 The designation of JC virus (JCV) was derived from the initials of the patient whose brain tissue was used for culture and isolation. Variants in the noncoding region of the genome were then serially identified as Mad 1, Mad 2, and so on, representing the geographic location, Madison, Wisconsin, where the virus was identified.

The JCV, a polyomavirus, is a nonenveloped DNA virus with icosahedral structure containing double-stranded DNA genomes. The circular genome of JCV contains early and late transcription units, the latter of which encodes three virion structural proteins—VPl, VP2, and VP3. Humans generate antibodies directed against the amino terminal end of VP1 and perhaps VP2 and VP3.

JC VIRUS PATHOGENESIS

JCV pathogenesis is studied in cell cultures derived from human fetal brain tissue. In vitro, JCV robustly infects astrocytes, making it important to identify the culture’s cellular phenotypes. A cell line was developed that allows multiplication of JCV and, more recently, human multipotential progenitor cells were isolated and are being grown from the human developing brain at various gestational stages. The lineage pathways of these cells can be differentiated into astrocytes, oligodendrocytes, and neurons. Initiating infection in progenitor cells with JC virions made it possible to determine which cells were susceptible to infection. JCV susceptibility is evident in progenitor-derived astrocytes and glial cells, which reflects the pathologic process in PML brain tissue. Neuronal cells, by contrast, are not susceptible to infection.4

JC VIRUS CHARACTERISTICS: GLOBAL DISTRIBUTION, TRIAD OF SYMPTOMS

Subcortical multifocal white matter lesions are the classic feature of PML on neuroimaging. Seroepidemiology of JCV has revealed ubiquitous distribution, with 50% to 60% of adults aged 20 to 50 years demonstrating antibody to JCV.5 The percentage of the population with antibody increases with age, but may vary among geographic regions. Prevalence is lower among remote populations.

Although the initial site of JCV infection is not well characterized, we know that the primary infection is not in the brain. The JCV has a selective tropism for replication in glial cells in the human brain, but the absence of an animal model for PML has hindered our understanding of the JCV migration to the brain and the initiation and development of central nervous system infection.

Although humans carry JCV-specific antibodies, the clinical significance of these antibodies is unknown. Antibody levels rise during active infection, at times to very high titers, but offer no protection. T-cell–mediated immune responses directed to structural and nonstructural proteins are important in controlling infection.

A high index of suspicion for PML is warranted in individuals who demonstrate the classic triad of symptoms (cognitive impairment, visual deficits, and motor dysfunction) and in whom magnetic resonance imaging shows evidence of demyelinated plaque lesions; however, evidence of the presence of JCV DNA in pathologic tissue is necessary to confirm a diagnosis of PML.

The development of an in situ DNA hybridization assay using a biotinylated probe has facilitated identification of JCV DNA in the infected nuclei of the pathologic tissue. The presence of JCV DNA in cerebrospinal fluid (CSF) samples can be detected using a quantitative polymerase chain reaction assay, targeting the viral genome in the amino terminal end of the viral T protein.6 This T protein coding region was targeted because it does not crossreact, even with other human polyomaviruses, and it is intolerant of mutations. This assay is certified by the Clinical Laboratory Improvement Amendments, licensed by the National Institutes of Health; it is the most sensitive (to levels of 10 copies/mL sample) assay available.

JC VIRUS SUSCEPTIBILITY FACTORS

Despite the high prevalence of JCV infection, PML is rare, suggesting important barriers to its development. Although the receptor for JCV has been identified as alpha 2,6-linked sialic acid, the host range for productive infection is controlled by factors within the cell nucleus that bind to the viral promoter; this process initiates transcription of mRNA for the coordinated synthesis of viral proteins. Only certain cells have the necessary DNA binding proteins in high enough concentrations to allow lytic infection to take place, spreading by cell-to-cell contact. These cells include oligodendrocytes, the primary target for JCV, whose destruction leads to PML; astrocytes; and the CD34+ and CD19+ cells of the immune system. JCV can also be found in urine, at times in very high concentrations. It is present in the uroepithelial cells and multiplies without apparent pathologic consequences. Virus isolated from the urine has not been grown in cell culture systems in the laboratory setting.

Bone marrow CD34+ hematopoietic progenitor cells represent a potential pathway of JCV pathogenesis: in six people with PML, latent JCV DNA was demonstrated in pathologic tissue from lymph, spleen, or bone marrow biopsies taken months to years before the patient developed neurologic disease.7

Figure 1. A model for pathogenesis of JC virus (JCV) infection. The initial site of infection is assumed to be tonsillar stromal cells, from which JCV is disseminated by lymphocytes to bone marrow CD34 cells and uroepithelial cells in the kidney—presumed sites of latency. Reactivation occurs in the event of immunosuppression or immune modulation, after which it gains entry into the brain by way of peripheral B lymphocytes. Susceptible cells (oligodendrocytes, astrocytes, and progenitor cells) share a characteristic expression pattern of nuclear transcription factors required for synthesis of JCV early viral protein mRNA and replication of JCV DNA.
The initial site of JCV infection is not known, but it is believed that the virus infects stromal cells within the tonsils and then disseminates through peripheral blood (Figure 1).8 The CD34+ hematopoietic stem cells carry the virus in bone marrow, and the uroepithelial cell is the carrier in the kidney. The nucleotide arrangement of the regulatory region that drives infection is markedly different in the kidney than in the bone marrow or brain tissue of patients with PML. JCV isolated from the kidney is referred to as the archetype sequence, which, unlike PML in the brain, has no tandem repeats. No pathology is associated with JCV kidney infection, although about 30% of the population excrete JCV in the urine.9,10

Upon immunosuppression, reactivation of the virus occurs, with evidence of the virus found in CD10 and CD19/20 lymphocytes in the peripheral blood of some individuals. Blood-to-brain viral dissemination results in infection of oligodendrocytes, astrocytes, and progenitor cells.

 

 

Susceptibility is related to nucleotide sequences

Susceptibility to PML is associated with promoter/enhancer nucleotide sequences. The tandem repeat nucleotide structure has been found in the peripheral blood leukocytes and the CSF of patients with PML. Although the arrangement of nucleotide sequences in the viral regulatory region is highly variable among patients with PML, there are no alterations in the sequence within the origin of DNA replication. These highly conserved sequences contain regions for DNA-binding proteins that drive transcription, initiating the life cycle of the virus.

The nuclear transcription factor NF-1 is a cell-specific regulator of JCV promoter/enhancer activity. In humans, the NF-1 family of DNA-binding proteins is encoded by four discrete genes, one of which is NF-1 class X (NF-1X), a critical transcription factor that affects JCV cells. The human brain makes NF-1X in concentrations greater than the concentrations of other NF-1 transcription family members of DNA-binding proteins. NF-1X is located adjacent to and interacts with another family of transcription factors, activator protein-1, which has also been associated with JC viral activity.

Spi-B expression a factor in natalizumab-treated patients

Another transcription factor, Spi-B, binds to sequences present in the JCV promoter/enhancer. Spi-B is a regulator of JCV gene expression in susceptible cells and appears to play an important role in JCV activity. The expression of Spi-B is upregulated in patients with MS who are treated with the monoclonal antibody natalizumab, a population of patients in whom PML has been recently described.11–15

Reprinted, with permission, from The New England Journal of Medicine (Major EO. Reemergence of PML in natalizumab-treated patients: new cases, same concerns. N Engl J Med 2009; 361:1041–1043). © 2009 Massachusetts Medical Society. All rights reserved.
Figure 2. Development of progressive multifocal leukoencephalopathy in the setting of natalizumab therapy. Natalizumab forces the migration of hematopoietic stem cells and pre-B cells from the bone marrow by preventing their attachment to vascular-cell adhesion molecule (VCAM). The JC virus (JCV) can reside in the bone marrow in a latent state for extended periods and migrate to the peripheral circulation. JCV uses B cells and their DNA-binding proteins to initiate viral replication.

Natalizumab binds to the alpha-4 integrin molecule, preventing hematopoietic stem cells and developing B cells from attaching to a vascular-cell adhesion molecule and forcing them to migrate from the bone marrow (Figure 2).16 An ideal environment is created for JCV when the natalizumab-induced increase in CD34+ cells in the circulation is combined with upregulation of gene cells involved in B-cell maturation. JCV can reside in the bone marrow in a latent state and can use B cells and their DNA-binding proteins to initiate viral multiplication, eventually gaining entry into the brain to cause PML.

In addition to natalizumab, PML has been described in patients treated with efalizumab, another biologic agent that binds alpha-4 integrin molecules on the surface of T and B cells, preventing their entry into the brain, gut, and skin, and forcing migration of bone marrow CD34+ into peripheral circulation for long periods.9,17,18 Rituximab, another monoclonal antibody, binds the CD20 surface molecule on B cells, causing their depletion from the peripheral circulation through complement-mediated cytolysis.7

Risk factors for development of PML

Measurable risk factors for PML include:

  • Rising antibody titers
  • Evidence of viremia, especially persistent viremia associated with repeat sequences in the regulatory region of the viral genome
  • Ineffective T-cell (CD4 and CD8) responses
  • Molecular host factors (ie, Spi-B expression in B cells) that support JCV infection in potentially susceptible cells.

The presence of more than one of these risk factors is necessary for development of PML.

VIRAL LATENCY IN B LYMPHOCYTES IN BONE MARROW

A strong link between JCV infection in cells of the immune system and those of the nervous system points to the importance of the tissue origin of JCV latency. Bone marrow harbors CD34+ cells that migrate into the peripheral circulation and undergo differentiation to pre-B and mature B cells, augmenting JCV growth. The emergence of PML in patients treated with natalizumab, rituximab, efalizumab, and other immune-altering drugs underscores this observation.

As noted, the incidence of PML in natalizumab-treated patients with MS and Crohn disease rises as the number of doses increases. Analysis of blood samples collected from patients treated with natalizumab at baseline and again during treatment at months 1 to 12 and beyond 24 months demonstrates that the frequency of CD34+ cells in the peripheral circulation increases with the duration of therapy, adding credence to the theory that CD34+ cells act as a reservoir for latent virus. A higher frequency of CD34+ cells is associated with viremia.

The role of Spi-B in JC virus latency

Understanding the role of Spi-B during JCV latency and reactivation is increasingly important as the number of patients treated with immunomodulatory agents that can develop PML continues to rise. Spi-B is highly represented in the B cell and CD34+ cell fractions. Spi-B expression in B cells correlates with reactivation of JCV in immune cells in natalizumab-treated patients. In a sample of four patients with MS treated with natalizumab who developed PML, T-cell responses have been ineffective (absent or aberrant). Two patients had no detectable T-cell response to JCV; the other two demonstrated response, but their CD4 T-cell responses were dominated by interleukin-10–producing cells.

Longitudinal examination of CSF samples from 13 MS patients who were treated with natalizumab and subsequently developed PML revealed persistence of viral load even though all patients experienced immune reconstitution inflammatory syndrome and most had high levels of anti-JCV antibodies.19

 

 

SUMMARY

Despite the prevalence of JCV in the population, the development of PML is rare. Levels of JCV antibody rise during the course of active JCV infection, but they do not protect against infection. T-cell responses directed to structural and nonstructural proteins play a role in controlling infection. Latency of JCV is associated with specific cells of the immune system, and its reactivation can follow alteration of normal immune cell function—either immunosuppression or immunomodulation. Risk factors for the development of PML include rising antibody titers and ineffective T-cell (CD4 and CD8) responses.

DISCUSSION

Dr. Berger: Does natalizumab upregulate Spi-B in glial cells?

Dr. Major: We never tested this directly. From human brain cultures, we know that Spi-B is made in glial cells, not in neurons. We are considering the idea that wherever JCV binds, it takes advantage of certain types of DNA-binding proteins in the molecular regulation. If the binding takes place in an immune system cell, for example, Spi-B plays an important role.

Dr. Berger: Koralnik et al demonstrated JCV excretion in urine in MS patients after 12 months of treatment with natalizumab, and at 18 months, viremia in 60% of the patients.20 Yet, repeated studies of patients taking natalizumab have failed to demonstrate viremia or conversion of virus in the archetype. How do these findings correlate with your thoughts on the action of natalizumab in the pathogenesis of PML?

Dr. Major: We certainly know that natalizumab forces migration of hematopoietic stem cells and pre-B cells out of the marrow, but our findings have differed somewhat from those of Koralnik’s laboratory. For example, in the several hundred nucleotide sequences we have looked at in PML brain tissue, we have found the Mad 1 genotype once. We consider Mad 1 to be a potential laboratory contamination, so if we find Mad 1 we resequence the sample. We never clone because cloning can introduce alterations; we sequence directly from the clinical tissue. We can identify Mad 1 because our assay is very sensitive. In normal individuals, CD34+ cells compose approximately 0.01% of the peripheral circulation; in individuals treated with natalizumab, however, their composition is 0.1% to 0.3%. So if there is a potential for latent infection, we have an opportunity to find it in those cells. Its presence does not necessarily mean that the individual is going to develop PML, however; there are other controlling factors.

Dr. Rudick: Have you found the virus in B cells in healthy people?

Dr. Major: Yes we have, in about one-third. It is higher than what we would expect to see in the normal population.

Dr. Rudick: How can that finding be turned into something that’s clinically useful?

Dr. Major: If you’re trying to identify persons who are more susceptible to PML given underlying risk factors—treatment with natalizumab or rituximab, presence of HIV infection, or some other immune-altering condition—looking at one parameter isn’t going to help. Based on the available data, rising antibody titers signals an active infection, and viremia of any kind means probable latent infection. Because this is a small event in very few cells, you will not have the numbers of cells needed to identify susceptibility in a normal population. For now, we monitor patients at risk and, if we find viremia, we assess the cell population to determine whether a molecular factor like Spi-B is upregulated. We hope to develop an assay in which we can obtain one test tube of blood and report T-cell responses, molecular factors, antibody titer, and presence or absence of viremia. Such an assay would provide the data necessary to make a clinical decision.

References
  1. Astrom KE, Mancall EL, Richardson EP. Progressive multifocal leukoencephalopathy; a hitherto unrecognized complication of chronic lymphatic leukaemia and Hodgkin’s disease. Brain 1958; 81:93111.
  2. Hallervorden J. Eigennartige und nicht rubriziebare Prozesse. In:Bumke O, ed. Handbuch der Geiteskranheiten. Vol. 2. Die Anatomie der Psychosen. Berlin: Springer; 1930:10631107.
  3. Padgett BL, Walker DL, ZuRhein GM, Eckroade RJ, Dessel BH. Cultivation of a papova-like virus from human brain with progressive multifocal leucoencephalopathy. Lancet 1971; 1:12571260.
  4. Major EO, Amemiya K, Tornatore CS, Houff SA, Berger JR. Pathogenesis and molecular biology of progressive multifocal leukoencephalopathy, the JC virus-induced demyelinating disease of the human brain. Clin Microbiol Rev 1992; 5:4973.
  5. Walker D, Padgett B. The epidemiology of human polyomaviruses. In:Sever J, Madden D, eds. Polyomaviruses and Human Neurological Disease. New York, NY: Alan R. Liss, Inc.; 1983:99106.
  6. Ryschkewitsch C, Jensen P, Hou J, Fahle G, Fischer S, Major EO. Comparison of PCR-southern hybridization and quantitative realtime PCR for the detection of JC and BK viral nucleotide sequences in urine and cerebrospinal fluid. J Virol Methods 2004; 121:217221.
  7. Monaco MC, Jensen PN, Hou J, Durham LC, Major EO. Detection of JC virus DNA in human tonsil tissue: evidence for site of initial viral infection. J Virol 1998; 72:99189923.
  8. Major EO. Progressive multifocal leukoencephalopathy in patients on immunomodulatory therapies. Annu Rev Med 2010; 61:3547.
  9. Imperiale M, Major E. Polyomavirus. In:Knipe D, Howley P, eds. Field Virology. 5th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2007:22632298.
  10. Langer-Gould A, Atlas SW, Green AJ, Bollen AW, Pelletier D. Progressive multifocal leukoencephalopathy in a patient treated with natalizumab. N Engl J Med 2005; 353:375381.
  11. Van Assche G, Van Ranst M, Sciot R, et al. Progressive multifocal leukoencephalopathy after natalizumab therapy for Crohn’s disease. N Engl J Med 2005; 353:362368.
  12. Kleinschmidt-DeMasters BK, Tyler KL. Progressive multifocal leukoencephalopathy complicating treatment with natalizumab and interferon beta-1a for multiple sclerosis. N Engl J Med 2005; 353:369374.
  13. Bozic C, Belcher G, Kooijmans-Coutinho M, et al. Natalizumab utilization and safety in patients with relapsing multiple sclerosis: updated results from TOUCH™ and TYGRIS. Paper presented at: 60th Annual Meeting of the American Academy of Neurology; April 15, 2008; Chicago, IL.
  14. Kappos L, Bates D, Hartung HP, et al. Natalizumab treatment for multiple sclerosis: recommendations for patient selection and monitoring. Lancet Neurol 2007; 6:431441.
  15. Major EO. Reemergence of PML in natalizumab-treated patients—new cases, same concerns. N Engl J Med 2009; 361:10411043.
  16. Vugmeyster Y, Kikuchi T, Lowes MA, et al. Efalizumab (anti-CD11a)-induced increase in peripheral blood leukocytes in psoriasis patients is preferentially mediated by altered trafficking of memory CD8+ T cells into lesional skin. Clin Immunol 2004; 113:3846.
  17. Guttman-Yassky E, Vugmeyster Y, Lowes MA, et al. Blockade of CD11a by efalizumab in psoriasis patients induces a unique state of T-cell hyporesponsiveness. J Invest Dermatol 2008; 128:11821191.
  18. Carson KR, Evens AM, Richey EA, et al. Progressive multifocal leukoencephalopathy after rituximab therapy in HIV-negative patients: a report of 57 cases from the Research on Adverse Drug Events and Reports project. Blood 2009; 113:48344840.
  19. Ryschkewitsch CF, Jensen PN, Monaco MC, Major EO. JC virus persistence following progressive multifocal leukoencephalopathy in multiple sclerosis patients treated with natalizumab. Ann Neurol 2010; 68:384391.
  20. Koralnik IJ, Du Pasquier RA, Kuroda MJ, et al. Association of prolonged survival in HLA-A2+ progressive multifocal leuko encephalopathy patients with a CTL response specific for a commonly recognized JC virus epitope. J Immunol 2002; 168:499504.
Article PDF
Author and Disclosure Information

Eugene O. Major, PhD
Chief, Laboratory of Molecular Medicine and Neuroscience, AIDS Coordinator, Intramural Program, National Institute of Neurological Disorders and Stroke, Bethesda, MD

Correspondence: Eugene O. Major, PhD, Laboratory of Molecular Medicine and Neuroscience, NINDS, Building 10, Room 3B14, 10 Center Drive, MSC 1296, Bethesda, MD 20892-1296; majorg@ninds.nih.gov

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

This article was developed from an audio transcript of Dr. Major’s presentation at a roundtable convened at Cleveland Clinic on January 31, 2011. The transcript was edited by the Cleveland Clinic Journal of Medicine staff for clarity and conciseness, and was then reviewed, revised, and approved by Dr. Major.

Publications
Page Number
S3-S7
Author and Disclosure Information

Eugene O. Major, PhD
Chief, Laboratory of Molecular Medicine and Neuroscience, AIDS Coordinator, Intramural Program, National Institute of Neurological Disorders and Stroke, Bethesda, MD

Correspondence: Eugene O. Major, PhD, Laboratory of Molecular Medicine and Neuroscience, NINDS, Building 10, Room 3B14, 10 Center Drive, MSC 1296, Bethesda, MD 20892-1296; majorg@ninds.nih.gov

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

This article was developed from an audio transcript of Dr. Major’s presentation at a roundtable convened at Cleveland Clinic on January 31, 2011. The transcript was edited by the Cleveland Clinic Journal of Medicine staff for clarity and conciseness, and was then reviewed, revised, and approved by Dr. Major.

Author and Disclosure Information

Eugene O. Major, PhD
Chief, Laboratory of Molecular Medicine and Neuroscience, AIDS Coordinator, Intramural Program, National Institute of Neurological Disorders and Stroke, Bethesda, MD

Correspondence: Eugene O. Major, PhD, Laboratory of Molecular Medicine and Neuroscience, NINDS, Building 10, Room 3B14, 10 Center Drive, MSC 1296, Bethesda, MD 20892-1296; majorg@ninds.nih.gov

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

This article was developed from an audio transcript of Dr. Major’s presentation at a roundtable convened at Cleveland Clinic on January 31, 2011. The transcript was edited by the Cleveland Clinic Journal of Medicine staff for clarity and conciseness, and was then reviewed, revised, and approved by Dr. Major.

Article PDF
Article PDF

The neuropathology of progressive multifocal leukoencephalopathy (PML) was first reported in 1958 following examination of brain tissue from two cases of chronic lymphocytic leukemia and one case of Hodgkin lymphoma.1 The classic triad of symptoms of PML—cognitive impairment, visual deficits, and motor dysfunction—had been observed previously but had not been formally described.2

Until PML was discovered in patients with autoimmune diseases treated with biologic therapies that do not directly suppress immunity, PML had been considered a very rare, virus-induced demyelinating disease of the white matter that occurred in immune-compromised patients. The incidence of PML rose sharply in the mid-1980s with the pandemic of human immunodeficiency virus (HIV)-1 infection and continues as an acquired immunodeficiency syndrome–defining illness at a rate of approximately 1% to 3% of HIV-1 seropositive individuals; more recently, it has been seen in approximately 1 in 850 natalizumab-treated individuals who have multiple sclerosis (MS). The incidence of PML in natalizumab-treated MS patients increases with dosing; among those who receive 24 or more doses, the incidence is 1 in 400.

The cause of PML was unknown until 1971, when viral particles were observed by electron microscopy in PML brain lesions and subsequently isolated at the University of Wisconsin, Madison, in cultures of human fetal brain tissue.3 The designation of JC virus (JCV) was derived from the initials of the patient whose brain tissue was used for culture and isolation. Variants in the noncoding region of the genome were then serially identified as Mad 1, Mad 2, and so on, representing the geographic location, Madison, Wisconsin, where the virus was identified.

The JCV, a polyomavirus, is a nonenveloped DNA virus with icosahedral structure containing double-stranded DNA genomes. The circular genome of JCV contains early and late transcription units, the latter of which encodes three virion structural proteins—VPl, VP2, and VP3. Humans generate antibodies directed against the amino terminal end of VP1 and perhaps VP2 and VP3.

JC VIRUS PATHOGENESIS

JCV pathogenesis is studied in cell cultures derived from human fetal brain tissue. In vitro, JCV robustly infects astrocytes, making it important to identify the culture’s cellular phenotypes. A cell line was developed that allows multiplication of JCV and, more recently, human multipotential progenitor cells were isolated and are being grown from the human developing brain at various gestational stages. The lineage pathways of these cells can be differentiated into astrocytes, oligodendrocytes, and neurons. Initiating infection in progenitor cells with JC virions made it possible to determine which cells were susceptible to infection. JCV susceptibility is evident in progenitor-derived astrocytes and glial cells, which reflects the pathologic process in PML brain tissue. Neuronal cells, by contrast, are not susceptible to infection.4

JC VIRUS CHARACTERISTICS: GLOBAL DISTRIBUTION, TRIAD OF SYMPTOMS

Subcortical multifocal white matter lesions are the classic feature of PML on neuroimaging. Seroepidemiology of JCV has revealed ubiquitous distribution, with 50% to 60% of adults aged 20 to 50 years demonstrating antibody to JCV.5 The percentage of the population with antibody increases with age, but may vary among geographic regions. Prevalence is lower among remote populations.

Although the initial site of JCV infection is not well characterized, we know that the primary infection is not in the brain. The JCV has a selective tropism for replication in glial cells in the human brain, but the absence of an animal model for PML has hindered our understanding of the JCV migration to the brain and the initiation and development of central nervous system infection.

Although humans carry JCV-specific antibodies, the clinical significance of these antibodies is unknown. Antibody levels rise during active infection, at times to very high titers, but offer no protection. T-cell–mediated immune responses directed to structural and nonstructural proteins are important in controlling infection.

A high index of suspicion for PML is warranted in individuals who demonstrate the classic triad of symptoms (cognitive impairment, visual deficits, and motor dysfunction) and in whom magnetic resonance imaging shows evidence of demyelinated plaque lesions; however, evidence of the presence of JCV DNA in pathologic tissue is necessary to confirm a diagnosis of PML.

The development of an in situ DNA hybridization assay using a biotinylated probe has facilitated identification of JCV DNA in the infected nuclei of the pathologic tissue. The presence of JCV DNA in cerebrospinal fluid (CSF) samples can be detected using a quantitative polymerase chain reaction assay, targeting the viral genome in the amino terminal end of the viral T protein.6 This T protein coding region was targeted because it does not crossreact, even with other human polyomaviruses, and it is intolerant of mutations. This assay is certified by the Clinical Laboratory Improvement Amendments, licensed by the National Institutes of Health; it is the most sensitive (to levels of 10 copies/mL sample) assay available.

JC VIRUS SUSCEPTIBILITY FACTORS

Despite the high prevalence of JCV infection, PML is rare, suggesting important barriers to its development. Although the receptor for JCV has been identified as alpha 2,6-linked sialic acid, the host range for productive infection is controlled by factors within the cell nucleus that bind to the viral promoter; this process initiates transcription of mRNA for the coordinated synthesis of viral proteins. Only certain cells have the necessary DNA binding proteins in high enough concentrations to allow lytic infection to take place, spreading by cell-to-cell contact. These cells include oligodendrocytes, the primary target for JCV, whose destruction leads to PML; astrocytes; and the CD34+ and CD19+ cells of the immune system. JCV can also be found in urine, at times in very high concentrations. It is present in the uroepithelial cells and multiplies without apparent pathologic consequences. Virus isolated from the urine has not been grown in cell culture systems in the laboratory setting.

Bone marrow CD34+ hematopoietic progenitor cells represent a potential pathway of JCV pathogenesis: in six people with PML, latent JCV DNA was demonstrated in pathologic tissue from lymph, spleen, or bone marrow biopsies taken months to years before the patient developed neurologic disease.7

Figure 1. A model for pathogenesis of JC virus (JCV) infection. The initial site of infection is assumed to be tonsillar stromal cells, from which JCV is disseminated by lymphocytes to bone marrow CD34 cells and uroepithelial cells in the kidney—presumed sites of latency. Reactivation occurs in the event of immunosuppression or immune modulation, after which it gains entry into the brain by way of peripheral B lymphocytes. Susceptible cells (oligodendrocytes, astrocytes, and progenitor cells) share a characteristic expression pattern of nuclear transcription factors required for synthesis of JCV early viral protein mRNA and replication of JCV DNA.
The initial site of JCV infection is not known, but it is believed that the virus infects stromal cells within the tonsils and then disseminates through peripheral blood (Figure 1).8 The CD34+ hematopoietic stem cells carry the virus in bone marrow, and the uroepithelial cell is the carrier in the kidney. The nucleotide arrangement of the regulatory region that drives infection is markedly different in the kidney than in the bone marrow or brain tissue of patients with PML. JCV isolated from the kidney is referred to as the archetype sequence, which, unlike PML in the brain, has no tandem repeats. No pathology is associated with JCV kidney infection, although about 30% of the population excrete JCV in the urine.9,10

Upon immunosuppression, reactivation of the virus occurs, with evidence of the virus found in CD10 and CD19/20 lymphocytes in the peripheral blood of some individuals. Blood-to-brain viral dissemination results in infection of oligodendrocytes, astrocytes, and progenitor cells.

 

 

Susceptibility is related to nucleotide sequences

Susceptibility to PML is associated with promoter/enhancer nucleotide sequences. The tandem repeat nucleotide structure has been found in the peripheral blood leukocytes and the CSF of patients with PML. Although the arrangement of nucleotide sequences in the viral regulatory region is highly variable among patients with PML, there are no alterations in the sequence within the origin of DNA replication. These highly conserved sequences contain regions for DNA-binding proteins that drive transcription, initiating the life cycle of the virus.

The nuclear transcription factor NF-1 is a cell-specific regulator of JCV promoter/enhancer activity. In humans, the NF-1 family of DNA-binding proteins is encoded by four discrete genes, one of which is NF-1 class X (NF-1X), a critical transcription factor that affects JCV cells. The human brain makes NF-1X in concentrations greater than the concentrations of other NF-1 transcription family members of DNA-binding proteins. NF-1X is located adjacent to and interacts with another family of transcription factors, activator protein-1, which has also been associated with JC viral activity.

Spi-B expression a factor in natalizumab-treated patients

Another transcription factor, Spi-B, binds to sequences present in the JCV promoter/enhancer. Spi-B is a regulator of JCV gene expression in susceptible cells and appears to play an important role in JCV activity. The expression of Spi-B is upregulated in patients with MS who are treated with the monoclonal antibody natalizumab, a population of patients in whom PML has been recently described.11–15

Reprinted, with permission, from The New England Journal of Medicine (Major EO. Reemergence of PML in natalizumab-treated patients: new cases, same concerns. N Engl J Med 2009; 361:1041–1043). © 2009 Massachusetts Medical Society. All rights reserved.
Figure 2. Development of progressive multifocal leukoencephalopathy in the setting of natalizumab therapy. Natalizumab forces the migration of hematopoietic stem cells and pre-B cells from the bone marrow by preventing their attachment to vascular-cell adhesion molecule (VCAM). The JC virus (JCV) can reside in the bone marrow in a latent state for extended periods and migrate to the peripheral circulation. JCV uses B cells and their DNA-binding proteins to initiate viral replication.

Natalizumab binds to the alpha-4 integrin molecule, preventing hematopoietic stem cells and developing B cells from attaching to a vascular-cell adhesion molecule and forcing them to migrate from the bone marrow (Figure 2).16 An ideal environment is created for JCV when the natalizumab-induced increase in CD34+ cells in the circulation is combined with upregulation of gene cells involved in B-cell maturation. JCV can reside in the bone marrow in a latent state and can use B cells and their DNA-binding proteins to initiate viral multiplication, eventually gaining entry into the brain to cause PML.

In addition to natalizumab, PML has been described in patients treated with efalizumab, another biologic agent that binds alpha-4 integrin molecules on the surface of T and B cells, preventing their entry into the brain, gut, and skin, and forcing migration of bone marrow CD34+ into peripheral circulation for long periods.9,17,18 Rituximab, another monoclonal antibody, binds the CD20 surface molecule on B cells, causing their depletion from the peripheral circulation through complement-mediated cytolysis.7

Risk factors for development of PML

Measurable risk factors for PML include:

  • Rising antibody titers
  • Evidence of viremia, especially persistent viremia associated with repeat sequences in the regulatory region of the viral genome
  • Ineffective T-cell (CD4 and CD8) responses
  • Molecular host factors (ie, Spi-B expression in B cells) that support JCV infection in potentially susceptible cells.

The presence of more than one of these risk factors is necessary for development of PML.

VIRAL LATENCY IN B LYMPHOCYTES IN BONE MARROW

A strong link between JCV infection in cells of the immune system and those of the nervous system points to the importance of the tissue origin of JCV latency. Bone marrow harbors CD34+ cells that migrate into the peripheral circulation and undergo differentiation to pre-B and mature B cells, augmenting JCV growth. The emergence of PML in patients treated with natalizumab, rituximab, efalizumab, and other immune-altering drugs underscores this observation.

As noted, the incidence of PML in natalizumab-treated patients with MS and Crohn disease rises as the number of doses increases. Analysis of blood samples collected from patients treated with natalizumab at baseline and again during treatment at months 1 to 12 and beyond 24 months demonstrates that the frequency of CD34+ cells in the peripheral circulation increases with the duration of therapy, adding credence to the theory that CD34+ cells act as a reservoir for latent virus. A higher frequency of CD34+ cells is associated with viremia.

The role of Spi-B in JC virus latency

Understanding the role of Spi-B during JCV latency and reactivation is increasingly important as the number of patients treated with immunomodulatory agents that can develop PML continues to rise. Spi-B is highly represented in the B cell and CD34+ cell fractions. Spi-B expression in B cells correlates with reactivation of JCV in immune cells in natalizumab-treated patients. In a sample of four patients with MS treated with natalizumab who developed PML, T-cell responses have been ineffective (absent or aberrant). Two patients had no detectable T-cell response to JCV; the other two demonstrated response, but their CD4 T-cell responses were dominated by interleukin-10–producing cells.

Longitudinal examination of CSF samples from 13 MS patients who were treated with natalizumab and subsequently developed PML revealed persistence of viral load even though all patients experienced immune reconstitution inflammatory syndrome and most had high levels of anti-JCV antibodies.19

 

 

SUMMARY

Despite the prevalence of JCV in the population, the development of PML is rare. Levels of JCV antibody rise during the course of active JCV infection, but they do not protect against infection. T-cell responses directed to structural and nonstructural proteins play a role in controlling infection. Latency of JCV is associated with specific cells of the immune system, and its reactivation can follow alteration of normal immune cell function—either immunosuppression or immunomodulation. Risk factors for the development of PML include rising antibody titers and ineffective T-cell (CD4 and CD8) responses.

DISCUSSION

Dr. Berger: Does natalizumab upregulate Spi-B in glial cells?

Dr. Major: We never tested this directly. From human brain cultures, we know that Spi-B is made in glial cells, not in neurons. We are considering the idea that wherever JCV binds, it takes advantage of certain types of DNA-binding proteins in the molecular regulation. If the binding takes place in an immune system cell, for example, Spi-B plays an important role.

Dr. Berger: Koralnik et al demonstrated JCV excretion in urine in MS patients after 12 months of treatment with natalizumab, and at 18 months, viremia in 60% of the patients.20 Yet, repeated studies of patients taking natalizumab have failed to demonstrate viremia or conversion of virus in the archetype. How do these findings correlate with your thoughts on the action of natalizumab in the pathogenesis of PML?

Dr. Major: We certainly know that natalizumab forces migration of hematopoietic stem cells and pre-B cells out of the marrow, but our findings have differed somewhat from those of Koralnik’s laboratory. For example, in the several hundred nucleotide sequences we have looked at in PML brain tissue, we have found the Mad 1 genotype once. We consider Mad 1 to be a potential laboratory contamination, so if we find Mad 1 we resequence the sample. We never clone because cloning can introduce alterations; we sequence directly from the clinical tissue. We can identify Mad 1 because our assay is very sensitive. In normal individuals, CD34+ cells compose approximately 0.01% of the peripheral circulation; in individuals treated with natalizumab, however, their composition is 0.1% to 0.3%. So if there is a potential for latent infection, we have an opportunity to find it in those cells. Its presence does not necessarily mean that the individual is going to develop PML, however; there are other controlling factors.

Dr. Rudick: Have you found the virus in B cells in healthy people?

Dr. Major: Yes we have, in about one-third. It is higher than what we would expect to see in the normal population.

Dr. Rudick: How can that finding be turned into something that’s clinically useful?

Dr. Major: If you’re trying to identify persons who are more susceptible to PML given underlying risk factors—treatment with natalizumab or rituximab, presence of HIV infection, or some other immune-altering condition—looking at one parameter isn’t going to help. Based on the available data, rising antibody titers signals an active infection, and viremia of any kind means probable latent infection. Because this is a small event in very few cells, you will not have the numbers of cells needed to identify susceptibility in a normal population. For now, we monitor patients at risk and, if we find viremia, we assess the cell population to determine whether a molecular factor like Spi-B is upregulated. We hope to develop an assay in which we can obtain one test tube of blood and report T-cell responses, molecular factors, antibody titer, and presence or absence of viremia. Such an assay would provide the data necessary to make a clinical decision.

The neuropathology of progressive multifocal leukoencephalopathy (PML) was first reported in 1958 following examination of brain tissue from two cases of chronic lymphocytic leukemia and one case of Hodgkin lymphoma.1 The classic triad of symptoms of PML—cognitive impairment, visual deficits, and motor dysfunction—had been observed previously but had not been formally described.2

Until PML was discovered in patients with autoimmune diseases treated with biologic therapies that do not directly suppress immunity, PML had been considered a very rare, virus-induced demyelinating disease of the white matter that occurred in immune-compromised patients. The incidence of PML rose sharply in the mid-1980s with the pandemic of human immunodeficiency virus (HIV)-1 infection and continues as an acquired immunodeficiency syndrome–defining illness at a rate of approximately 1% to 3% of HIV-1 seropositive individuals; more recently, it has been seen in approximately 1 in 850 natalizumab-treated individuals who have multiple sclerosis (MS). The incidence of PML in natalizumab-treated MS patients increases with dosing; among those who receive 24 or more doses, the incidence is 1 in 400.

The cause of PML was unknown until 1971, when viral particles were observed by electron microscopy in PML brain lesions and subsequently isolated at the University of Wisconsin, Madison, in cultures of human fetal brain tissue.3 The designation of JC virus (JCV) was derived from the initials of the patient whose brain tissue was used for culture and isolation. Variants in the noncoding region of the genome were then serially identified as Mad 1, Mad 2, and so on, representing the geographic location, Madison, Wisconsin, where the virus was identified.

The JCV, a polyomavirus, is a nonenveloped DNA virus with icosahedral structure containing double-stranded DNA genomes. The circular genome of JCV contains early and late transcription units, the latter of which encodes three virion structural proteins—VPl, VP2, and VP3. Humans generate antibodies directed against the amino terminal end of VP1 and perhaps VP2 and VP3.

JC VIRUS PATHOGENESIS

JCV pathogenesis is studied in cell cultures derived from human fetal brain tissue. In vitro, JCV robustly infects astrocytes, making it important to identify the culture’s cellular phenotypes. A cell line was developed that allows multiplication of JCV and, more recently, human multipotential progenitor cells were isolated and are being grown from the human developing brain at various gestational stages. The lineage pathways of these cells can be differentiated into astrocytes, oligodendrocytes, and neurons. Initiating infection in progenitor cells with JC virions made it possible to determine which cells were susceptible to infection. JCV susceptibility is evident in progenitor-derived astrocytes and glial cells, which reflects the pathologic process in PML brain tissue. Neuronal cells, by contrast, are not susceptible to infection.4

JC VIRUS CHARACTERISTICS: GLOBAL DISTRIBUTION, TRIAD OF SYMPTOMS

Subcortical multifocal white matter lesions are the classic feature of PML on neuroimaging. Seroepidemiology of JCV has revealed ubiquitous distribution, with 50% to 60% of adults aged 20 to 50 years demonstrating antibody to JCV.5 The percentage of the population with antibody increases with age, but may vary among geographic regions. Prevalence is lower among remote populations.

Although the initial site of JCV infection is not well characterized, we know that the primary infection is not in the brain. The JCV has a selective tropism for replication in glial cells in the human brain, but the absence of an animal model for PML has hindered our understanding of the JCV migration to the brain and the initiation and development of central nervous system infection.

Although humans carry JCV-specific antibodies, the clinical significance of these antibodies is unknown. Antibody levels rise during active infection, at times to very high titers, but offer no protection. T-cell–mediated immune responses directed to structural and nonstructural proteins are important in controlling infection.

A high index of suspicion for PML is warranted in individuals who demonstrate the classic triad of symptoms (cognitive impairment, visual deficits, and motor dysfunction) and in whom magnetic resonance imaging shows evidence of demyelinated plaque lesions; however, evidence of the presence of JCV DNA in pathologic tissue is necessary to confirm a diagnosis of PML.

The development of an in situ DNA hybridization assay using a biotinylated probe has facilitated identification of JCV DNA in the infected nuclei of the pathologic tissue. The presence of JCV DNA in cerebrospinal fluid (CSF) samples can be detected using a quantitative polymerase chain reaction assay, targeting the viral genome in the amino terminal end of the viral T protein.6 This T protein coding region was targeted because it does not crossreact, even with other human polyomaviruses, and it is intolerant of mutations. This assay is certified by the Clinical Laboratory Improvement Amendments, licensed by the National Institutes of Health; it is the most sensitive (to levels of 10 copies/mL sample) assay available.

JC VIRUS SUSCEPTIBILITY FACTORS

Despite the high prevalence of JCV infection, PML is rare, suggesting important barriers to its development. Although the receptor for JCV has been identified as alpha 2,6-linked sialic acid, the host range for productive infection is controlled by factors within the cell nucleus that bind to the viral promoter; this process initiates transcription of mRNA for the coordinated synthesis of viral proteins. Only certain cells have the necessary DNA binding proteins in high enough concentrations to allow lytic infection to take place, spreading by cell-to-cell contact. These cells include oligodendrocytes, the primary target for JCV, whose destruction leads to PML; astrocytes; and the CD34+ and CD19+ cells of the immune system. JCV can also be found in urine, at times in very high concentrations. It is present in the uroepithelial cells and multiplies without apparent pathologic consequences. Virus isolated from the urine has not been grown in cell culture systems in the laboratory setting.

Bone marrow CD34+ hematopoietic progenitor cells represent a potential pathway of JCV pathogenesis: in six people with PML, latent JCV DNA was demonstrated in pathologic tissue from lymph, spleen, or bone marrow biopsies taken months to years before the patient developed neurologic disease.7

Figure 1. A model for pathogenesis of JC virus (JCV) infection. The initial site of infection is assumed to be tonsillar stromal cells, from which JCV is disseminated by lymphocytes to bone marrow CD34 cells and uroepithelial cells in the kidney—presumed sites of latency. Reactivation occurs in the event of immunosuppression or immune modulation, after which it gains entry into the brain by way of peripheral B lymphocytes. Susceptible cells (oligodendrocytes, astrocytes, and progenitor cells) share a characteristic expression pattern of nuclear transcription factors required for synthesis of JCV early viral protein mRNA and replication of JCV DNA.
The initial site of JCV infection is not known, but it is believed that the virus infects stromal cells within the tonsils and then disseminates through peripheral blood (Figure 1).8 The CD34+ hematopoietic stem cells carry the virus in bone marrow, and the uroepithelial cell is the carrier in the kidney. The nucleotide arrangement of the regulatory region that drives infection is markedly different in the kidney than in the bone marrow or brain tissue of patients with PML. JCV isolated from the kidney is referred to as the archetype sequence, which, unlike PML in the brain, has no tandem repeats. No pathology is associated with JCV kidney infection, although about 30% of the population excrete JCV in the urine.9,10

Upon immunosuppression, reactivation of the virus occurs, with evidence of the virus found in CD10 and CD19/20 lymphocytes in the peripheral blood of some individuals. Blood-to-brain viral dissemination results in infection of oligodendrocytes, astrocytes, and progenitor cells.

 

 

Susceptibility is related to nucleotide sequences

Susceptibility to PML is associated with promoter/enhancer nucleotide sequences. The tandem repeat nucleotide structure has been found in the peripheral blood leukocytes and the CSF of patients with PML. Although the arrangement of nucleotide sequences in the viral regulatory region is highly variable among patients with PML, there are no alterations in the sequence within the origin of DNA replication. These highly conserved sequences contain regions for DNA-binding proteins that drive transcription, initiating the life cycle of the virus.

The nuclear transcription factor NF-1 is a cell-specific regulator of JCV promoter/enhancer activity. In humans, the NF-1 family of DNA-binding proteins is encoded by four discrete genes, one of which is NF-1 class X (NF-1X), a critical transcription factor that affects JCV cells. The human brain makes NF-1X in concentrations greater than the concentrations of other NF-1 transcription family members of DNA-binding proteins. NF-1X is located adjacent to and interacts with another family of transcription factors, activator protein-1, which has also been associated with JC viral activity.

Spi-B expression a factor in natalizumab-treated patients

Another transcription factor, Spi-B, binds to sequences present in the JCV promoter/enhancer. Spi-B is a regulator of JCV gene expression in susceptible cells and appears to play an important role in JCV activity. The expression of Spi-B is upregulated in patients with MS who are treated with the monoclonal antibody natalizumab, a population of patients in whom PML has been recently described.11–15

Reprinted, with permission, from The New England Journal of Medicine (Major EO. Reemergence of PML in natalizumab-treated patients: new cases, same concerns. N Engl J Med 2009; 361:1041–1043). © 2009 Massachusetts Medical Society. All rights reserved.
Figure 2. Development of progressive multifocal leukoencephalopathy in the setting of natalizumab therapy. Natalizumab forces the migration of hematopoietic stem cells and pre-B cells from the bone marrow by preventing their attachment to vascular-cell adhesion molecule (VCAM). The JC virus (JCV) can reside in the bone marrow in a latent state for extended periods and migrate to the peripheral circulation. JCV uses B cells and their DNA-binding proteins to initiate viral replication.

Natalizumab binds to the alpha-4 integrin molecule, preventing hematopoietic stem cells and developing B cells from attaching to a vascular-cell adhesion molecule and forcing them to migrate from the bone marrow (Figure 2).16 An ideal environment is created for JCV when the natalizumab-induced increase in CD34+ cells in the circulation is combined with upregulation of gene cells involved in B-cell maturation. JCV can reside in the bone marrow in a latent state and can use B cells and their DNA-binding proteins to initiate viral multiplication, eventually gaining entry into the brain to cause PML.

In addition to natalizumab, PML has been described in patients treated with efalizumab, another biologic agent that binds alpha-4 integrin molecules on the surface of T and B cells, preventing their entry into the brain, gut, and skin, and forcing migration of bone marrow CD34+ into peripheral circulation for long periods.9,17,18 Rituximab, another monoclonal antibody, binds the CD20 surface molecule on B cells, causing their depletion from the peripheral circulation through complement-mediated cytolysis.7

Risk factors for development of PML

Measurable risk factors for PML include:

  • Rising antibody titers
  • Evidence of viremia, especially persistent viremia associated with repeat sequences in the regulatory region of the viral genome
  • Ineffective T-cell (CD4 and CD8) responses
  • Molecular host factors (ie, Spi-B expression in B cells) that support JCV infection in potentially susceptible cells.

The presence of more than one of these risk factors is necessary for development of PML.

VIRAL LATENCY IN B LYMPHOCYTES IN BONE MARROW

A strong link between JCV infection in cells of the immune system and those of the nervous system points to the importance of the tissue origin of JCV latency. Bone marrow harbors CD34+ cells that migrate into the peripheral circulation and undergo differentiation to pre-B and mature B cells, augmenting JCV growth. The emergence of PML in patients treated with natalizumab, rituximab, efalizumab, and other immune-altering drugs underscores this observation.

As noted, the incidence of PML in natalizumab-treated patients with MS and Crohn disease rises as the number of doses increases. Analysis of blood samples collected from patients treated with natalizumab at baseline and again during treatment at months 1 to 12 and beyond 24 months demonstrates that the frequency of CD34+ cells in the peripheral circulation increases with the duration of therapy, adding credence to the theory that CD34+ cells act as a reservoir for latent virus. A higher frequency of CD34+ cells is associated with viremia.

The role of Spi-B in JC virus latency

Understanding the role of Spi-B during JCV latency and reactivation is increasingly important as the number of patients treated with immunomodulatory agents that can develop PML continues to rise. Spi-B is highly represented in the B cell and CD34+ cell fractions. Spi-B expression in B cells correlates with reactivation of JCV in immune cells in natalizumab-treated patients. In a sample of four patients with MS treated with natalizumab who developed PML, T-cell responses have been ineffective (absent or aberrant). Two patients had no detectable T-cell response to JCV; the other two demonstrated response, but their CD4 T-cell responses were dominated by interleukin-10–producing cells.

Longitudinal examination of CSF samples from 13 MS patients who were treated with natalizumab and subsequently developed PML revealed persistence of viral load even though all patients experienced immune reconstitution inflammatory syndrome and most had high levels of anti-JCV antibodies.19

 

 

SUMMARY

Despite the prevalence of JCV in the population, the development of PML is rare. Levels of JCV antibody rise during the course of active JCV infection, but they do not protect against infection. T-cell responses directed to structural and nonstructural proteins play a role in controlling infection. Latency of JCV is associated with specific cells of the immune system, and its reactivation can follow alteration of normal immune cell function—either immunosuppression or immunomodulation. Risk factors for the development of PML include rising antibody titers and ineffective T-cell (CD4 and CD8) responses.

DISCUSSION

Dr. Berger: Does natalizumab upregulate Spi-B in glial cells?

Dr. Major: We never tested this directly. From human brain cultures, we know that Spi-B is made in glial cells, not in neurons. We are considering the idea that wherever JCV binds, it takes advantage of certain types of DNA-binding proteins in the molecular regulation. If the binding takes place in an immune system cell, for example, Spi-B plays an important role.

Dr. Berger: Koralnik et al demonstrated JCV excretion in urine in MS patients after 12 months of treatment with natalizumab, and at 18 months, viremia in 60% of the patients.20 Yet, repeated studies of patients taking natalizumab have failed to demonstrate viremia or conversion of virus in the archetype. How do these findings correlate with your thoughts on the action of natalizumab in the pathogenesis of PML?

Dr. Major: We certainly know that natalizumab forces migration of hematopoietic stem cells and pre-B cells out of the marrow, but our findings have differed somewhat from those of Koralnik’s laboratory. For example, in the several hundred nucleotide sequences we have looked at in PML brain tissue, we have found the Mad 1 genotype once. We consider Mad 1 to be a potential laboratory contamination, so if we find Mad 1 we resequence the sample. We never clone because cloning can introduce alterations; we sequence directly from the clinical tissue. We can identify Mad 1 because our assay is very sensitive. In normal individuals, CD34+ cells compose approximately 0.01% of the peripheral circulation; in individuals treated with natalizumab, however, their composition is 0.1% to 0.3%. So if there is a potential for latent infection, we have an opportunity to find it in those cells. Its presence does not necessarily mean that the individual is going to develop PML, however; there are other controlling factors.

Dr. Rudick: Have you found the virus in B cells in healthy people?

Dr. Major: Yes we have, in about one-third. It is higher than what we would expect to see in the normal population.

Dr. Rudick: How can that finding be turned into something that’s clinically useful?

Dr. Major: If you’re trying to identify persons who are more susceptible to PML given underlying risk factors—treatment with natalizumab or rituximab, presence of HIV infection, or some other immune-altering condition—looking at one parameter isn’t going to help. Based on the available data, rising antibody titers signals an active infection, and viremia of any kind means probable latent infection. Because this is a small event in very few cells, you will not have the numbers of cells needed to identify susceptibility in a normal population. For now, we monitor patients at risk and, if we find viremia, we assess the cell population to determine whether a molecular factor like Spi-B is upregulated. We hope to develop an assay in which we can obtain one test tube of blood and report T-cell responses, molecular factors, antibody titer, and presence or absence of viremia. Such an assay would provide the data necessary to make a clinical decision.

References
  1. Astrom KE, Mancall EL, Richardson EP. Progressive multifocal leukoencephalopathy; a hitherto unrecognized complication of chronic lymphatic leukaemia and Hodgkin’s disease. Brain 1958; 81:93111.
  2. Hallervorden J. Eigennartige und nicht rubriziebare Prozesse. In:Bumke O, ed. Handbuch der Geiteskranheiten. Vol. 2. Die Anatomie der Psychosen. Berlin: Springer; 1930:10631107.
  3. Padgett BL, Walker DL, ZuRhein GM, Eckroade RJ, Dessel BH. Cultivation of a papova-like virus from human brain with progressive multifocal leucoencephalopathy. Lancet 1971; 1:12571260.
  4. Major EO, Amemiya K, Tornatore CS, Houff SA, Berger JR. Pathogenesis and molecular biology of progressive multifocal leukoencephalopathy, the JC virus-induced demyelinating disease of the human brain. Clin Microbiol Rev 1992; 5:4973.
  5. Walker D, Padgett B. The epidemiology of human polyomaviruses. In:Sever J, Madden D, eds. Polyomaviruses and Human Neurological Disease. New York, NY: Alan R. Liss, Inc.; 1983:99106.
  6. Ryschkewitsch C, Jensen P, Hou J, Fahle G, Fischer S, Major EO. Comparison of PCR-southern hybridization and quantitative realtime PCR for the detection of JC and BK viral nucleotide sequences in urine and cerebrospinal fluid. J Virol Methods 2004; 121:217221.
  7. Monaco MC, Jensen PN, Hou J, Durham LC, Major EO. Detection of JC virus DNA in human tonsil tissue: evidence for site of initial viral infection. J Virol 1998; 72:99189923.
  8. Major EO. Progressive multifocal leukoencephalopathy in patients on immunomodulatory therapies. Annu Rev Med 2010; 61:3547.
  9. Imperiale M, Major E. Polyomavirus. In:Knipe D, Howley P, eds. Field Virology. 5th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2007:22632298.
  10. Langer-Gould A, Atlas SW, Green AJ, Bollen AW, Pelletier D. Progressive multifocal leukoencephalopathy in a patient treated with natalizumab. N Engl J Med 2005; 353:375381.
  11. Van Assche G, Van Ranst M, Sciot R, et al. Progressive multifocal leukoencephalopathy after natalizumab therapy for Crohn’s disease. N Engl J Med 2005; 353:362368.
  12. Kleinschmidt-DeMasters BK, Tyler KL. Progressive multifocal leukoencephalopathy complicating treatment with natalizumab and interferon beta-1a for multiple sclerosis. N Engl J Med 2005; 353:369374.
  13. Bozic C, Belcher G, Kooijmans-Coutinho M, et al. Natalizumab utilization and safety in patients with relapsing multiple sclerosis: updated results from TOUCH™ and TYGRIS. Paper presented at: 60th Annual Meeting of the American Academy of Neurology; April 15, 2008; Chicago, IL.
  14. Kappos L, Bates D, Hartung HP, et al. Natalizumab treatment for multiple sclerosis: recommendations for patient selection and monitoring. Lancet Neurol 2007; 6:431441.
  15. Major EO. Reemergence of PML in natalizumab-treated patients—new cases, same concerns. N Engl J Med 2009; 361:10411043.
  16. Vugmeyster Y, Kikuchi T, Lowes MA, et al. Efalizumab (anti-CD11a)-induced increase in peripheral blood leukocytes in psoriasis patients is preferentially mediated by altered trafficking of memory CD8+ T cells into lesional skin. Clin Immunol 2004; 113:3846.
  17. Guttman-Yassky E, Vugmeyster Y, Lowes MA, et al. Blockade of CD11a by efalizumab in psoriasis patients induces a unique state of T-cell hyporesponsiveness. J Invest Dermatol 2008; 128:11821191.
  18. Carson KR, Evens AM, Richey EA, et al. Progressive multifocal leukoencephalopathy after rituximab therapy in HIV-negative patients: a report of 57 cases from the Research on Adverse Drug Events and Reports project. Blood 2009; 113:48344840.
  19. Ryschkewitsch CF, Jensen PN, Monaco MC, Major EO. JC virus persistence following progressive multifocal leukoencephalopathy in multiple sclerosis patients treated with natalizumab. Ann Neurol 2010; 68:384391.
  20. Koralnik IJ, Du Pasquier RA, Kuroda MJ, et al. Association of prolonged survival in HLA-A2+ progressive multifocal leuko encephalopathy patients with a CTL response specific for a commonly recognized JC virus epitope. J Immunol 2002; 168:499504.
References
  1. Astrom KE, Mancall EL, Richardson EP. Progressive multifocal leukoencephalopathy; a hitherto unrecognized complication of chronic lymphatic leukaemia and Hodgkin’s disease. Brain 1958; 81:93111.
  2. Hallervorden J. Eigennartige und nicht rubriziebare Prozesse. In:Bumke O, ed. Handbuch der Geiteskranheiten. Vol. 2. Die Anatomie der Psychosen. Berlin: Springer; 1930:10631107.
  3. Padgett BL, Walker DL, ZuRhein GM, Eckroade RJ, Dessel BH. Cultivation of a papova-like virus from human brain with progressive multifocal leucoencephalopathy. Lancet 1971; 1:12571260.
  4. Major EO, Amemiya K, Tornatore CS, Houff SA, Berger JR. Pathogenesis and molecular biology of progressive multifocal leukoencephalopathy, the JC virus-induced demyelinating disease of the human brain. Clin Microbiol Rev 1992; 5:4973.
  5. Walker D, Padgett B. The epidemiology of human polyomaviruses. In:Sever J, Madden D, eds. Polyomaviruses and Human Neurological Disease. New York, NY: Alan R. Liss, Inc.; 1983:99106.
  6. Ryschkewitsch C, Jensen P, Hou J, Fahle G, Fischer S, Major EO. Comparison of PCR-southern hybridization and quantitative realtime PCR for the detection of JC and BK viral nucleotide sequences in urine and cerebrospinal fluid. J Virol Methods 2004; 121:217221.
  7. Monaco MC, Jensen PN, Hou J, Durham LC, Major EO. Detection of JC virus DNA in human tonsil tissue: evidence for site of initial viral infection. J Virol 1998; 72:99189923.
  8. Major EO. Progressive multifocal leukoencephalopathy in patients on immunomodulatory therapies. Annu Rev Med 2010; 61:3547.
  9. Imperiale M, Major E. Polyomavirus. In:Knipe D, Howley P, eds. Field Virology. 5th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2007:22632298.
  10. Langer-Gould A, Atlas SW, Green AJ, Bollen AW, Pelletier D. Progressive multifocal leukoencephalopathy in a patient treated with natalizumab. N Engl J Med 2005; 353:375381.
  11. Van Assche G, Van Ranst M, Sciot R, et al. Progressive multifocal leukoencephalopathy after natalizumab therapy for Crohn’s disease. N Engl J Med 2005; 353:362368.
  12. Kleinschmidt-DeMasters BK, Tyler KL. Progressive multifocal leukoencephalopathy complicating treatment with natalizumab and interferon beta-1a for multiple sclerosis. N Engl J Med 2005; 353:369374.
  13. Bozic C, Belcher G, Kooijmans-Coutinho M, et al. Natalizumab utilization and safety in patients with relapsing multiple sclerosis: updated results from TOUCH™ and TYGRIS. Paper presented at: 60th Annual Meeting of the American Academy of Neurology; April 15, 2008; Chicago, IL.
  14. Kappos L, Bates D, Hartung HP, et al. Natalizumab treatment for multiple sclerosis: recommendations for patient selection and monitoring. Lancet Neurol 2007; 6:431441.
  15. Major EO. Reemergence of PML in natalizumab-treated patients—new cases, same concerns. N Engl J Med 2009; 361:10411043.
  16. Vugmeyster Y, Kikuchi T, Lowes MA, et al. Efalizumab (anti-CD11a)-induced increase in peripheral blood leukocytes in psoriasis patients is preferentially mediated by altered trafficking of memory CD8+ T cells into lesional skin. Clin Immunol 2004; 113:3846.
  17. Guttman-Yassky E, Vugmeyster Y, Lowes MA, et al. Blockade of CD11a by efalizumab in psoriasis patients induces a unique state of T-cell hyporesponsiveness. J Invest Dermatol 2008; 128:11821191.
  18. Carson KR, Evens AM, Richey EA, et al. Progressive multifocal leukoencephalopathy after rituximab therapy in HIV-negative patients: a report of 57 cases from the Research on Adverse Drug Events and Reports project. Blood 2009; 113:48344840.
  19. Ryschkewitsch CF, Jensen PN, Monaco MC, Major EO. JC virus persistence following progressive multifocal leukoencephalopathy in multiple sclerosis patients treated with natalizumab. Ann Neurol 2010; 68:384391.
  20. Koralnik IJ, Du Pasquier RA, Kuroda MJ, et al. Association of prolonged survival in HLA-A2+ progressive multifocal leuko encephalopathy patients with a CTL response specific for a commonly recognized JC virus epitope. J Immunol 2002; 168:499504.
Page Number
S3-S7
Page Number
S3-S7
Publications
Publications
Article Type
Display Headline
History and current concepts in the pathogenesis of PML
Display Headline
History and current concepts in the pathogenesis of PML
Citation Override
Cleveland Clinic Journal of Medicine 2011 November;78(suppl 2):S3-S7
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media

The clinical features of PML

Article Type
Changed
Tue, 10/02/2018 - 09:22
Display Headline
The clinical features of PML

Progressive multifocal leukoencephalopathy (PML) was a rare disease until the era of human immunodeficiency virus (HIV) infection, when the number of cases of PML markedly increased. We are now entering a new era in which PML is being observed in patients treated with biologic agents for diseases not associated with development of PML.

This article reviews the epidemiology and symptoms that characterize PML, the identification of lesions on radiographic imaging that support the diagnosis, the value of laboratory studies and immunocytochemistry in the diagnosis, and clinical outcomes.

CHANGING EPIDEMIOLOGY OF PML

Figure. Although certain findings are considered classic in progressive multifocal leukoencephalopathy (PML) patients, the percentage of patients affected has shifted as the disorder has evolved from the pre–acquired immunodeficiency syndrome (AIDS) era to PML associated with biologic therapies such as natalizumab. Prior to the AIDS era, nearly one-half of patients with PML were affected by visual deficits. Among patients with AIDS-related PML, motor weakness is a feature for more than one-half, while cognitive impairment is the most common clinical presentation of natalizumab-related PML, affecting nearly one-half of patients.1,2,6,9
The presentation and epidemiology of PML have evolved over the last several decades. Prior to the acquired immunodeficiency syndrome (AIDS) era, nearly one-half of patients with PML were affected by visual deficits. Among patients with AIDS-related PML, motor weakness is a feature for more than one-half, while cognitive impairment is the most common clinical presentation in natalizumab-related PML, affecting nearly one-half of patients (Figure).

The pre-AIDS era

Lesions of subcortical white matter characterize PML and the patient’s clinical manifestations reflect their location. Brooks and Walker1 reviewed 69 pathologically confirmed and 40 virologically and pathologically confirmed cases of PML in the era before AIDS, and categorized the neurologic signs and symptoms at onset and during disease progression; the clinical picture had three significant findings:

  • Impaired vision: Defective vision, most commonly homonymous hemianopsia, was the most frequent presenting sign, present in 35% to 45% of cases. At the time of diagnosis, 6% to 8% of the patients were cortically blind because of bioccipital pathology.
  • Motor weakness: Motor weakness was the initial sign in 25% to 33% of patients. At the time of diagnosis, hemiparesis or hemiplegia was present in nearly all patients.
  • Changes in mentation: A change in mentation, including personality change, difficulty with memory, emotional lability, and frank dementia, was the presenting sign in approximately one-third of cases and eventually involved most patients.

AIDS-related PML

The epidemiology of PML changed with the AIDS pandemic. From 1958 to 1984, Brooks and Walker1 identified 230 cases of PML; in the period from 1981 to 1994, Berger and colleagues2 described 154 cases of AIDS-related PML that had been identified by the University of Miami Medical Center and the Broward County medical examiner’s office. The frequency of PML from 1991 through 1994 was 12-fold greater than the frequency 10 years earlier, from 1981 through 1984. Among the patients with AIDS-related PML, the most common initial symptoms were weakness (42%), speech abnormalities (40%), cognitive abnormalities (36%), gait abnormalities (29%), sensory loss (19%), and visual impairment (19%), followed by seizures, diplopia, and limb incoordination. The most common findings at the time of initial physical examination were weakness (54%), followed by gait abnormalities (20%), cognitive abnormalities (20%), dysarthria (24%), aphasia (19%), sensory loss (19%), visual impairment (17%), and oculomotor palsy (6%). For about 5% of patients with PML, it is the heralding manifestation of AIDS.

Although clinical features consistent with cerebral hemisphere lesions are most common, brainstem and cerebellar findings are also observed. Among these are ataxia, dysmetria, dysarthria, and oculomotor nerve palsies.2–4 Other signs and symptoms associated with PML include headache, vertigo, seizures, sensory deficits, parkinsonism, 5 aphasia, and neglect syndromes.1–4 In some cases, the coexistence of encephalitis with HIV infection could have accounted for some of the symptoms.

PML associated with monoclonal antibody therapy

Natalizumab is an alpha-4-beta-1 integrin inhibitor approved for the treatment of relapsing-remitting multiple sclerosis (MS); patients taking natalizumab represent the second largest group with PML (the largest group is patients with AIDS). Natalizumab-associated PML has some noteworthy features. The most common clinical presentations are cognitive disorders (48%), motor abnormalities (37%), language disturbances (31%), and visual defects (26%). Lesions are often monofocal rather than multifocal and the most common site of involvement is the frontal lobe.6 Among MS patients with natalizumab-associated PML, 30% to 40% have gadolinium-enhancing lesions on magnetic resonance imaging (MRI) at the time of diagnosis.

IMMUNE RECONSTITUTION INFLAMMATORY SYNDROME

The immune reconstitution inflammatory syndrome (IRIS) is a paradoxical clinical deterioration that typically occurs in the face of immunologic recovery (Table).7,8 Although not unique to PML or opportunistic infections, it is frequently observed in the setting of HIV infection and natalizumab-associated PML, and it may be concurrent with the diagnosis of PML.6,9

Among patients with HIV, predictors for the development of IRIS include antiretroviral naiveté, profoundly low CD4 lymphocyte counts (< 50 cells/mm3), a rapid decrease in HIV load, and the presence of active or subclinical opportunistic infections at the time of initiation of combined antiretroviral therapy. Tan and colleagues8 have reported the largest series to date. Of the 54 patients in their series, 36 developed PML and IRIS simultaneously, and 18 had worsening of preexisting PML. Although some investigators have recommended corticosteroid therapy for PML-IRIS, no controlled trials have been conducted and caution has been advised, particularly in patients without contrast enhancement on MRI or mass effect.

DIAGNOSTIC TESTING: NEUROIMAGING, CEREBROSPINAL FLUID ANALYSIS

Neuroimaging, including computed tomography (CT) and MRI, is a useful diagnostic tool for investigating a patient with PML. Cerebrospinal fluid (CSF) analysis for the presence of JC virus (JCV) may play a significant role, but it primarily serves to rule out other illnesses.

Computed tomography: lesion size may not reflect clinical status

On CT, demyelinating lesions appear as subcortical hypodensities, often with a propensity for parietooccipital areas that are confined to the white matter at the junction interface of the gray-white junction of the cortex.9–11 Lesions may be seen in the corpus callosum, thalamus, and basal ganglia,9 but changes in the size of lesions observed on CT do not necessarily reflect clinical progression.12 Prior to the availability of highly active antiretroviral therapy (HAART) for the treatment of AIDS, mass effect was exceptionally rare. However, the development of IRIS with PML, typically in AIDS patients following the use of HAART, may be associated with edema.13 Single-dose intravenous contrast and delayed, double-dose contrast CT scanning enhancement is observed in a minority of patients, typically fewer than 10%.8 This enhancement is generally faint and peripherally located.

Magnetic resonance imaging may show lesions before clinical disease

MRI is vastly more sensitive than CT in detecting the demyelinating lesions of PML.9,14 On rare occasions, MRI will clearly demonstrate pathology when CT is normal. In fact, MRI may show lesions in advance of clinically apparent disease.15 The characteristics of these lesions are hyperintensity on T2-weighted imaging, fluid-attenuated inversion recovery sequences, and hypointensity on T1-weighted image. Apparent diffusion coefficients (ADC) on MRI are typically normal to low in new lesions and at the advancing edge of lesions; the ADC was typically higher in the center of lesions.16

As observed on CT, approximately 10% of patients exhibit a faint rim of gadolinium enhancement.2,9 Enhancement is more common with PML-IRIS, and the distribution of lesions parallels what is seen pathologically. Enhancement PML lesions have altered signal characteristics compared with the surrounding white matter.9,17–19 In contrast, 15% of HIV-associated PML showed gadolinium enhancement on MRI at the time of diagnosis.6,9

Cerebrospinal fluid analysis

With the exception of polymerase chain reaction (PCR) for JCV, the primary utility of lumbar puncture in the setting of possible PML is to exclude the presence of other illnesses, including treatable infections.

CSF findings in patients with PML are nonspecific, with most patients demon strating a normal profile. A mild lymphocytic pleocytosis, which is rarely (if ever) more than 25 leukocytes/mL, occurs in 15% of patients. Total protein level is mildly elevated in approximately 20% to 30% of patients.

The CSF examination in HIV-infected patients with PML may reflect changes associated with HIV: low-grade lymphocytic pleocytosis (< 20 cells/mm3), mildly elevated protein (< 65 mg/dL), and elevated immunoglobulin G and oligoclonal bands. These abnormalities should not be attributed to PML.

 

 

DIAGNOSIS

The most reliable and accurate method for the diagnosis of PML remains brain biopsy that demonstrates the characteristic triad of histopathologic findings (demyelination, bizarre astrocytes, and enlarged oligodendrocyte nuclei) coupled with evidence of JCV infection. With respect to the latter, in situ hybridization or immunocytochemistry can be employed. In situ DNADNA hybridization is a method of annealing JCV DNA to complementary strands either in paraffin-embedded tissue or in frozen sections from biopsy samples.

In immunocytochemistry, antibodies to both T antigen and the common polyomavirus capsid antigen are used to detect cells undergoing productive viral infection. Cells that are positive by in situ hybridization are in a stage of active viral replication. Cells positive by immunocytochemistry that are expressing viral capsid antigens are in a stage of viral transcription and translation (ie, undergoing productive infection). In addition to their utility in confirming a diagnosis of PML, these techniques have demonstrated the presence of JCV in perivascular locations and at sites distant from foci of demyelination. Alternatively, PCR may be used to demonstrate JCV in brain tissue.

In the absence of biopsy, which few deem necessary today, a widely employed approach to diagnosis requires the demonstration of:

  • JCV in the CSF by PCR
  • Compatible clinical presentation
  • An MRI finding consistent with PML
  • No other alternative diagnosis.

With an ultrasensitive PCR technique, sensitivities should approach or may exceed 95%, but PCR sensitivity remains at 75% in some laboratories. Because the viral copy numbers in the CSF may be low, particularly in a patient treated with a monoclonal antibody such as natalizumab, the CSF PCR may be falsely negative.

If clinical suspicion of the disease remains high in the face of an initially negative CSF JCV, the CSF analysis should be repeated. CSF analysis for JCV is approximately 99% specific, but recent studies demonstrating low copy numbers of JCV in the CSF of patients with MS have raised concerns about potential pitfalls of this assay.20

PROGNOSIS

Until recently, PML was regarded as virtually universally fatal. The mean survival in the pre-AIDS era was approximately 6 months, and mortality was 80% within 9 months of disease onset. Rarely, patients had long survivals that ranged from 5 years to 19 years.

In the early years of the AIDS era, survival with PML did not appear to differ significantly from that observed in the pre-AIDS years. In the largest study of HIV-associated PML in the era prior to HAART, the median survival was 183 days.2 However, the majority of individuals were dead within 3 months of diagnosis. Only 8% to 10% of patients survived longer than 12 months, which has been regarded as “prolonged survival.” This long survival skewed the mean and median survival rates in this population.

Several factors have since been identified that correlate with prolonged survival in HIV-associated PML, including PML as the heralding manifestation of HIV, CD4 counts exceeding 300 cells per mm3, contrast enhancement of the lesions on radiographic imaging, low copy number or decreasing JCV titers in CSF,21–24 and the presence of JCV-specific cytotoxic T cells.25 A better prognosis has also been postulated for higher CSF levels of macrophage chemoattractant protein-126 and PML associated with JCV VP1 loop-specific polymorphisms.27

Prognosis of HIV-associated PML improves with immune system restoration

In the era of HAART, not only has the incidence of HIV-associated PML declined, but the prognosis of affected patients has improved as well. This development highlights the importance of restoration of the immune system in both disease prevention and survival. Some estimate that as many as 50% of HAART-treated patients with PML exhibit prolonged survival. In one study of 25 patients, the median survival was more than 46 weeks.28

Nonetheless, PML continues to have the worst prognosis of any AIDS-related cerebral disorder, with those having advanced immunosuppression being most susceptible to the disorder. For AIDS patients with PML, those who were HAART-naïve at the time of diagnosis appear to have better survival than treatment-experienced patients.29 Survival also correlates with reduced JCV load in the CSF30 and improved CD4 lymphocyte counts (CD4 counts > 100 cells/mm3).31

Prognosis of natalizumab-associated PML is different

The prognosis of natalizumab-associated PML differs from that of HIV-associated PML. In a series of 35 patients, 25 (71%) patients were alive on average 6 months after diagnosis.32 Prognosis was worse with a longer time to diagnosis and the presence of widespread disease.

Most deaths in patients taking natalizumab who developed PML have occurred during IRIS. Steroid treatment of IRIS appears to improve prognosis,8 but no scientifically rigorous study has been undertaken to demonstrate this recommendation.7 Among the survivors, neurologic deficit was mild in one-third, moderate in one-third, and severe in one-third of patients.

CONCLUSION: DISPELLING SOME MYTHS

Several assumptions about PML are not necessarily true. For example, although PML implies the presence of multifocal lesions as a characteristic of the disease, the lesions may be monofocal, especially with natalizumab-associated PML. The lesions of PML may show early gadolinium enhancement on neuroimaging. Although lesions typically are seen in subcortical white matter, cortical involvement also may be observed. Cerebellar granular cell degeneration may occur in association with PML or in isolation. Disease progression and death are not inevitable, even in the absence of treatment. The most important determinant for survival is restoration of the immune system.

DISCUSSION

Dr. Calabrese: Why are sensory deficits so common?

Dr. Berger: We don’t know. Because we see involvement in the parietal lobe, we would anticipate observing sensory deficits. I think that a lot of sensation occurs deep in the thalamic area, which is not often involved in PML. Also, we often don’t test for some of the deficits that may occur.

Dr. Rudick: Do you know of any cases of natalizumab-associated PML detected as an incidental finding on MRI, making a case for screening MRI in patients without clinical symptoms?

Dr. Berger: There have been a handful of cases, including one of the seminal cases of natalizumab-associated PML, in which MRI abnormalities were observed in advance of clinically recognized symptomatology.

Dr. Calabrese: The correlate question is, if a patient with a risk factor—be it HIV or treatment with a biologic agent—has a common neurocognitive sign or perhaps some subtle motor findings, does a normal MRI have 100% negative predictive value?

Dr. Berger: I have yet to see somebody with PML who has a normal MRI.

Dr. Simpson: What you may see are lesions that are not typical MRI lesions of white matter hypointensity. In some cases, as Dr. Berger mentioned in his summary, we’ll see cerebellar degeneration—atrophy—but not necessarily white matter lesions.

References
  1. Brooks BR, Walker DL. Progressive multifocal leukoencephalopathy. Neurol Clin 1984; 2:299313.
  2. Berger JR, Pall L, Lanska D, Whiteman M. Progressive multifocal leukoencephalopathy in patients with HIV infection. J Neurovirol 1998; 4:5968.
  3. Parr J, Horoupian DS, Winkelman AC. Cerebellar form of progressive multifocal leukoencephalopathy (PML). Can J Neurol Sci 1979; 6:123128.
  4. Jones HR, Hedley-Whyte ET, Freidberg SR, Kelleher JE, Krolikowski J. Primary cerebellopontine progressive multifocal leukoencephalopathy diagnosed premortem by cerebellar biopsy. Ann Neurol 1982; 11:199202.
  5. O’Riordan S, McGuigan C, Farrell M, Hutchinson M. Progressive multifocal leucoencephalopathy presenting with parkinsonism. J Neurol 2003; 250:13791381.
  6. Clifford DB, De Luca A, Simpson DM, Arendt G, Giovannoni G, Nath A. Natalizumab-associated progressive multifocal leukoencephalopathy in patients with multiple sclerosis: lessons from 28 cases. Lancet Neurol 2010; 9:438446.
  7. Berger JR. Steroids for PML-IRIS. A double-edged sword? Neurology 2009; 72:14541455.
  8. Tan K, Roda R, Ostrow L, McArthur J, Nath A. PML-IRIS in patients with HIV infection: clinical manifestations and treatment with steroids. Neurology 2009; 72:14581464.
  9. Whiteman ML, Post MJ, Berger JR, Tate LG, Bell MD, Limonte LP. Progressive multifocal leukoencephalopathy in 47 HIV-seropositive patients: neuroimaging with clinical and pathologic correlation. Radiology 1993; 187:233240.
  10. Conomy JP, Weinstein MA, Agamanolis D, Holt WS. Computed tomography in progressive multifocal leukoencephalopathy. AJR Am J Roentgenol 1976; 127:663665.
  11. Huckman MS. Computed tomography in the diagnosis of degenerative brain disease. Radiol Clin North Am 1982; 20:169183.
  12. Krupp LB, Lipton RB, Swerdlow ML, Leeds NE, Llena J. Progressive multifocal leukoencephalopathy: clinical and radiographic features. Ann Neurol 1985; 17:344349.
  13. Rushing EJ, Liappis A, Smirniotopoulos JD, et al. Immune reconstitution inflammatory syndrome of the brain: case illustrations of a challenging entity. J Neuropathol Exp Neurol 2008; 67:819827.
  14. Post MJ, Sheldon JJ, Hensley GT, et al. Central nervous system disease in acquired immunodeficiency syndrome: prospective correlation using CT, MR imaging, and pathologic studies. Radiology 1986; 158:141148.
  15. Langer-Gould A, Atlas SW, Green AJ, Bollen AW, Pelletier D. Progressive multifocal leukoencephalopathy in a patient treated with natalizumab. N Engl J Med 2005; 353:375381.
  16. Bergui M, Bradac GB, Oguz KK, et al. Progressive multifocal leukoencephalopathy: diffusion-weighted imaging and pathological correlations. Neuroradiology 2004; 46:2225.
  17. Guilleux MH, Steiner RE, Young IR. MR imaging in progressive multifocal leukoencephalopathy. AJNR Am J Neuroradiol 1986; 7:10331035.
  18. Levy JD, Cottingham KL, Campbell RJ, et al. Progressive multifocal leukoencephalopathy and magnetic resonance imaging. Ann Neurol 1986; 19:399401.
  19. Mark AS, Atlas SW. Progressive multifocal leukoencephalopathy in patients with AIDS: appearance on MR images. Radiology 1989; 173:517520.
  20. Iacobaeus E, Ryschkewitsch C, Gravell M, et al. Analysis of cerebrospinal fluid and cerebrospinal fluid cells from patients with multiple sclerosis for detection of JC virus DNA. Mult Scler 2009; 15:2835.
  21. Berger JR, Levy RM, Flomenhoft D, Dobbs M. Predictive factors for prolonged survival in acquired immunodeficiency syndrome-associated progressive multifocal leukoencephalopathy. Ann Neurol 1998; 44:341349.
  22. Taoufik Y, Gasnault J, Karaterki A, et al. Prognostic value of JC virus load in cerebrospinal fluid of patients with progressive multifocal leukoencephalopathy. J Infect Dis 1998; 178:18161820.
  23. Yiannoutsos CT, Major EO, Curfman B, et al. Relation of JC virus DNA in the cerebrospinal fluid to survival in acquired immunodeficiency syndrome patients with biopsy-proven progressive multifocal leukoencephalopathy. Ann Neurol 1999; 45:816821.
  24. Bossolasco S, Calori G, Moretti F, et al. Prognostic significance of JC virus DNA levels in cerebrospinal fluid of patients with HIV-associated progressive multifocal leukoencephalopathy. Clin Infect Dis 2005; 40:738744.
  25. Du Pasquier RA, Clark KW, Smith PS, et al. JCV-specific cellular immune response correlates with a favorable clinical outcome in HIV-infected individuals with progressive multifocal leukoencephalopathy. J Neurovirol 2001; 7:318322.
  26. Marzocchetti A, Cingolani A, Giambenedetto SD, et al. Macrophage chemoattractant protein-1 levels in cerebrospinal fluid correlate with containment of JC virus and prognosis of acquired immunodeficiency syndrome-associated progressive multifocal leukoencephalopathy. J Neurovirol 2005; 11:219224.
  27. Delbue S, Branchetti E, Bertolacci S, et al. JC virus VP1 loop-specific polymorphisms are associated with favorable prognosis for progressive multifocal leukoencephalopathy. J Neurovirol 2009; 15:5156.
  28. Clifford DB, Yiannoutsos C, Glicksman M, et al. HAART improves prognosis in HIV-associated progressive multifocal leukoencephalopathy. Neurology 1999; 52:623625.
  29. Wyen C, Hoffmann C, Schmeisser N, et al. Progressive multi focal leukoencephalopathy in patients on highly active antiretroviral therapy: survival and risk factors of death. J Acquir Immune Defic Syndr 2004; 37:12631268.
  30. De Luca A, Giancola ML, Ammassari A, et al. The effect of potent antiretroviral therapy and JC virus load in cerebrospinal fluid on clinical outcome of patients with AIDS-associated progressive multifocal leukoencephalopathy. J Infect Dis 2000; 182:10771083.
  31. Berenguer J, Miralles P, Arrizabalaga J, et al. Clinical course and prognostic factors of progressive multifocal leukoencephalopathy in patients treated with highly active antiretroviral therapy. Clin Infect Dis 2003; 36:10471052.
  32. Vermersch P, Kappos L, Gold R, et al. Clinical outcomes of natalizumab associated progressive multifocal leukoencephalopathy. Neurology 2011; 76:16971704.
Article PDF
Author and Disclosure Information

Joseph R. Berger, MD
Ruth L. Works Professor and Chairman, Department of Neurology, University of Kentucky College of Medicine, Lexington, KY

Correspondence: Joseph R. Berger, MD, Department of Neurology, University of Kentucky Medical Center, KY Clinic (Wing D) – L445, Lexington, KY 40536-0284; jrbneuro@email.uky.ed

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

This article was developed from an audio transcript of Dr. Berger’s presentation at a roundtable convened at Cleveland Clinic on January 31, 2011. The transcript was edited by the Cleveland Clinic Journal of Medicine staff for clarity and conciseness, and was then reviewed, revised, and approved by Dr. Berger.

Publications
Page Number
S8-S12
Author and Disclosure Information

Joseph R. Berger, MD
Ruth L. Works Professor and Chairman, Department of Neurology, University of Kentucky College of Medicine, Lexington, KY

Correspondence: Joseph R. Berger, MD, Department of Neurology, University of Kentucky Medical Center, KY Clinic (Wing D) – L445, Lexington, KY 40536-0284; jrbneuro@email.uky.ed

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

This article was developed from an audio transcript of Dr. Berger’s presentation at a roundtable convened at Cleveland Clinic on January 31, 2011. The transcript was edited by the Cleveland Clinic Journal of Medicine staff for clarity and conciseness, and was then reviewed, revised, and approved by Dr. Berger.

Author and Disclosure Information

Joseph R. Berger, MD
Ruth L. Works Professor and Chairman, Department of Neurology, University of Kentucky College of Medicine, Lexington, KY

Correspondence: Joseph R. Berger, MD, Department of Neurology, University of Kentucky Medical Center, KY Clinic (Wing D) – L445, Lexington, KY 40536-0284; jrbneuro@email.uky.ed

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

This article was developed from an audio transcript of Dr. Berger’s presentation at a roundtable convened at Cleveland Clinic on January 31, 2011. The transcript was edited by the Cleveland Clinic Journal of Medicine staff for clarity and conciseness, and was then reviewed, revised, and approved by Dr. Berger.

Article PDF
Article PDF

Progressive multifocal leukoencephalopathy (PML) was a rare disease until the era of human immunodeficiency virus (HIV) infection, when the number of cases of PML markedly increased. We are now entering a new era in which PML is being observed in patients treated with biologic agents for diseases not associated with development of PML.

This article reviews the epidemiology and symptoms that characterize PML, the identification of lesions on radiographic imaging that support the diagnosis, the value of laboratory studies and immunocytochemistry in the diagnosis, and clinical outcomes.

CHANGING EPIDEMIOLOGY OF PML

Figure. Although certain findings are considered classic in progressive multifocal leukoencephalopathy (PML) patients, the percentage of patients affected has shifted as the disorder has evolved from the pre–acquired immunodeficiency syndrome (AIDS) era to PML associated with biologic therapies such as natalizumab. Prior to the AIDS era, nearly one-half of patients with PML were affected by visual deficits. Among patients with AIDS-related PML, motor weakness is a feature for more than one-half, while cognitive impairment is the most common clinical presentation of natalizumab-related PML, affecting nearly one-half of patients.1,2,6,9
The presentation and epidemiology of PML have evolved over the last several decades. Prior to the acquired immunodeficiency syndrome (AIDS) era, nearly one-half of patients with PML were affected by visual deficits. Among patients with AIDS-related PML, motor weakness is a feature for more than one-half, while cognitive impairment is the most common clinical presentation in natalizumab-related PML, affecting nearly one-half of patients (Figure).

The pre-AIDS era

Lesions of subcortical white matter characterize PML and the patient’s clinical manifestations reflect their location. Brooks and Walker1 reviewed 69 pathologically confirmed and 40 virologically and pathologically confirmed cases of PML in the era before AIDS, and categorized the neurologic signs and symptoms at onset and during disease progression; the clinical picture had three significant findings:

  • Impaired vision: Defective vision, most commonly homonymous hemianopsia, was the most frequent presenting sign, present in 35% to 45% of cases. At the time of diagnosis, 6% to 8% of the patients were cortically blind because of bioccipital pathology.
  • Motor weakness: Motor weakness was the initial sign in 25% to 33% of patients. At the time of diagnosis, hemiparesis or hemiplegia was present in nearly all patients.
  • Changes in mentation: A change in mentation, including personality change, difficulty with memory, emotional lability, and frank dementia, was the presenting sign in approximately one-third of cases and eventually involved most patients.

AIDS-related PML

The epidemiology of PML changed with the AIDS pandemic. From 1958 to 1984, Brooks and Walker1 identified 230 cases of PML; in the period from 1981 to 1994, Berger and colleagues2 described 154 cases of AIDS-related PML that had been identified by the University of Miami Medical Center and the Broward County medical examiner’s office. The frequency of PML from 1991 through 1994 was 12-fold greater than the frequency 10 years earlier, from 1981 through 1984. Among the patients with AIDS-related PML, the most common initial symptoms were weakness (42%), speech abnormalities (40%), cognitive abnormalities (36%), gait abnormalities (29%), sensory loss (19%), and visual impairment (19%), followed by seizures, diplopia, and limb incoordination. The most common findings at the time of initial physical examination were weakness (54%), followed by gait abnormalities (20%), cognitive abnormalities (20%), dysarthria (24%), aphasia (19%), sensory loss (19%), visual impairment (17%), and oculomotor palsy (6%). For about 5% of patients with PML, it is the heralding manifestation of AIDS.

Although clinical features consistent with cerebral hemisphere lesions are most common, brainstem and cerebellar findings are also observed. Among these are ataxia, dysmetria, dysarthria, and oculomotor nerve palsies.2–4 Other signs and symptoms associated with PML include headache, vertigo, seizures, sensory deficits, parkinsonism, 5 aphasia, and neglect syndromes.1–4 In some cases, the coexistence of encephalitis with HIV infection could have accounted for some of the symptoms.

PML associated with monoclonal antibody therapy

Natalizumab is an alpha-4-beta-1 integrin inhibitor approved for the treatment of relapsing-remitting multiple sclerosis (MS); patients taking natalizumab represent the second largest group with PML (the largest group is patients with AIDS). Natalizumab-associated PML has some noteworthy features. The most common clinical presentations are cognitive disorders (48%), motor abnormalities (37%), language disturbances (31%), and visual defects (26%). Lesions are often monofocal rather than multifocal and the most common site of involvement is the frontal lobe.6 Among MS patients with natalizumab-associated PML, 30% to 40% have gadolinium-enhancing lesions on magnetic resonance imaging (MRI) at the time of diagnosis.

IMMUNE RECONSTITUTION INFLAMMATORY SYNDROME

The immune reconstitution inflammatory syndrome (IRIS) is a paradoxical clinical deterioration that typically occurs in the face of immunologic recovery (Table).7,8 Although not unique to PML or opportunistic infections, it is frequently observed in the setting of HIV infection and natalizumab-associated PML, and it may be concurrent with the diagnosis of PML.6,9

Among patients with HIV, predictors for the development of IRIS include antiretroviral naiveté, profoundly low CD4 lymphocyte counts (< 50 cells/mm3), a rapid decrease in HIV load, and the presence of active or subclinical opportunistic infections at the time of initiation of combined antiretroviral therapy. Tan and colleagues8 have reported the largest series to date. Of the 54 patients in their series, 36 developed PML and IRIS simultaneously, and 18 had worsening of preexisting PML. Although some investigators have recommended corticosteroid therapy for PML-IRIS, no controlled trials have been conducted and caution has been advised, particularly in patients without contrast enhancement on MRI or mass effect.

DIAGNOSTIC TESTING: NEUROIMAGING, CEREBROSPINAL FLUID ANALYSIS

Neuroimaging, including computed tomography (CT) and MRI, is a useful diagnostic tool for investigating a patient with PML. Cerebrospinal fluid (CSF) analysis for the presence of JC virus (JCV) may play a significant role, but it primarily serves to rule out other illnesses.

Computed tomography: lesion size may not reflect clinical status

On CT, demyelinating lesions appear as subcortical hypodensities, often with a propensity for parietooccipital areas that are confined to the white matter at the junction interface of the gray-white junction of the cortex.9–11 Lesions may be seen in the corpus callosum, thalamus, and basal ganglia,9 but changes in the size of lesions observed on CT do not necessarily reflect clinical progression.12 Prior to the availability of highly active antiretroviral therapy (HAART) for the treatment of AIDS, mass effect was exceptionally rare. However, the development of IRIS with PML, typically in AIDS patients following the use of HAART, may be associated with edema.13 Single-dose intravenous contrast and delayed, double-dose contrast CT scanning enhancement is observed in a minority of patients, typically fewer than 10%.8 This enhancement is generally faint and peripherally located.

Magnetic resonance imaging may show lesions before clinical disease

MRI is vastly more sensitive than CT in detecting the demyelinating lesions of PML.9,14 On rare occasions, MRI will clearly demonstrate pathology when CT is normal. In fact, MRI may show lesions in advance of clinically apparent disease.15 The characteristics of these lesions are hyperintensity on T2-weighted imaging, fluid-attenuated inversion recovery sequences, and hypointensity on T1-weighted image. Apparent diffusion coefficients (ADC) on MRI are typically normal to low in new lesions and at the advancing edge of lesions; the ADC was typically higher in the center of lesions.16

As observed on CT, approximately 10% of patients exhibit a faint rim of gadolinium enhancement.2,9 Enhancement is more common with PML-IRIS, and the distribution of lesions parallels what is seen pathologically. Enhancement PML lesions have altered signal characteristics compared with the surrounding white matter.9,17–19 In contrast, 15% of HIV-associated PML showed gadolinium enhancement on MRI at the time of diagnosis.6,9

Cerebrospinal fluid analysis

With the exception of polymerase chain reaction (PCR) for JCV, the primary utility of lumbar puncture in the setting of possible PML is to exclude the presence of other illnesses, including treatable infections.

CSF findings in patients with PML are nonspecific, with most patients demon strating a normal profile. A mild lymphocytic pleocytosis, which is rarely (if ever) more than 25 leukocytes/mL, occurs in 15% of patients. Total protein level is mildly elevated in approximately 20% to 30% of patients.

The CSF examination in HIV-infected patients with PML may reflect changes associated with HIV: low-grade lymphocytic pleocytosis (< 20 cells/mm3), mildly elevated protein (< 65 mg/dL), and elevated immunoglobulin G and oligoclonal bands. These abnormalities should not be attributed to PML.

 

 

DIAGNOSIS

The most reliable and accurate method for the diagnosis of PML remains brain biopsy that demonstrates the characteristic triad of histopathologic findings (demyelination, bizarre astrocytes, and enlarged oligodendrocyte nuclei) coupled with evidence of JCV infection. With respect to the latter, in situ hybridization or immunocytochemistry can be employed. In situ DNADNA hybridization is a method of annealing JCV DNA to complementary strands either in paraffin-embedded tissue or in frozen sections from biopsy samples.

In immunocytochemistry, antibodies to both T antigen and the common polyomavirus capsid antigen are used to detect cells undergoing productive viral infection. Cells that are positive by in situ hybridization are in a stage of active viral replication. Cells positive by immunocytochemistry that are expressing viral capsid antigens are in a stage of viral transcription and translation (ie, undergoing productive infection). In addition to their utility in confirming a diagnosis of PML, these techniques have demonstrated the presence of JCV in perivascular locations and at sites distant from foci of demyelination. Alternatively, PCR may be used to demonstrate JCV in brain tissue.

In the absence of biopsy, which few deem necessary today, a widely employed approach to diagnosis requires the demonstration of:

  • JCV in the CSF by PCR
  • Compatible clinical presentation
  • An MRI finding consistent with PML
  • No other alternative diagnosis.

With an ultrasensitive PCR technique, sensitivities should approach or may exceed 95%, but PCR sensitivity remains at 75% in some laboratories. Because the viral copy numbers in the CSF may be low, particularly in a patient treated with a monoclonal antibody such as natalizumab, the CSF PCR may be falsely negative.

If clinical suspicion of the disease remains high in the face of an initially negative CSF JCV, the CSF analysis should be repeated. CSF analysis for JCV is approximately 99% specific, but recent studies demonstrating low copy numbers of JCV in the CSF of patients with MS have raised concerns about potential pitfalls of this assay.20

PROGNOSIS

Until recently, PML was regarded as virtually universally fatal. The mean survival in the pre-AIDS era was approximately 6 months, and mortality was 80% within 9 months of disease onset. Rarely, patients had long survivals that ranged from 5 years to 19 years.

In the early years of the AIDS era, survival with PML did not appear to differ significantly from that observed in the pre-AIDS years. In the largest study of HIV-associated PML in the era prior to HAART, the median survival was 183 days.2 However, the majority of individuals were dead within 3 months of diagnosis. Only 8% to 10% of patients survived longer than 12 months, which has been regarded as “prolonged survival.” This long survival skewed the mean and median survival rates in this population.

Several factors have since been identified that correlate with prolonged survival in HIV-associated PML, including PML as the heralding manifestation of HIV, CD4 counts exceeding 300 cells per mm3, contrast enhancement of the lesions on radiographic imaging, low copy number or decreasing JCV titers in CSF,21–24 and the presence of JCV-specific cytotoxic T cells.25 A better prognosis has also been postulated for higher CSF levels of macrophage chemoattractant protein-126 and PML associated with JCV VP1 loop-specific polymorphisms.27

Prognosis of HIV-associated PML improves with immune system restoration

In the era of HAART, not only has the incidence of HIV-associated PML declined, but the prognosis of affected patients has improved as well. This development highlights the importance of restoration of the immune system in both disease prevention and survival. Some estimate that as many as 50% of HAART-treated patients with PML exhibit prolonged survival. In one study of 25 patients, the median survival was more than 46 weeks.28

Nonetheless, PML continues to have the worst prognosis of any AIDS-related cerebral disorder, with those having advanced immunosuppression being most susceptible to the disorder. For AIDS patients with PML, those who were HAART-naïve at the time of diagnosis appear to have better survival than treatment-experienced patients.29 Survival also correlates with reduced JCV load in the CSF30 and improved CD4 lymphocyte counts (CD4 counts > 100 cells/mm3).31

Prognosis of natalizumab-associated PML is different

The prognosis of natalizumab-associated PML differs from that of HIV-associated PML. In a series of 35 patients, 25 (71%) patients were alive on average 6 months after diagnosis.32 Prognosis was worse with a longer time to diagnosis and the presence of widespread disease.

Most deaths in patients taking natalizumab who developed PML have occurred during IRIS. Steroid treatment of IRIS appears to improve prognosis,8 but no scientifically rigorous study has been undertaken to demonstrate this recommendation.7 Among the survivors, neurologic deficit was mild in one-third, moderate in one-third, and severe in one-third of patients.

CONCLUSION: DISPELLING SOME MYTHS

Several assumptions about PML are not necessarily true. For example, although PML implies the presence of multifocal lesions as a characteristic of the disease, the lesions may be monofocal, especially with natalizumab-associated PML. The lesions of PML may show early gadolinium enhancement on neuroimaging. Although lesions typically are seen in subcortical white matter, cortical involvement also may be observed. Cerebellar granular cell degeneration may occur in association with PML or in isolation. Disease progression and death are not inevitable, even in the absence of treatment. The most important determinant for survival is restoration of the immune system.

DISCUSSION

Dr. Calabrese: Why are sensory deficits so common?

Dr. Berger: We don’t know. Because we see involvement in the parietal lobe, we would anticipate observing sensory deficits. I think that a lot of sensation occurs deep in the thalamic area, which is not often involved in PML. Also, we often don’t test for some of the deficits that may occur.

Dr. Rudick: Do you know of any cases of natalizumab-associated PML detected as an incidental finding on MRI, making a case for screening MRI in patients without clinical symptoms?

Dr. Berger: There have been a handful of cases, including one of the seminal cases of natalizumab-associated PML, in which MRI abnormalities were observed in advance of clinically recognized symptomatology.

Dr. Calabrese: The correlate question is, if a patient with a risk factor—be it HIV or treatment with a biologic agent—has a common neurocognitive sign or perhaps some subtle motor findings, does a normal MRI have 100% negative predictive value?

Dr. Berger: I have yet to see somebody with PML who has a normal MRI.

Dr. Simpson: What you may see are lesions that are not typical MRI lesions of white matter hypointensity. In some cases, as Dr. Berger mentioned in his summary, we’ll see cerebellar degeneration—atrophy—but not necessarily white matter lesions.

Progressive multifocal leukoencephalopathy (PML) was a rare disease until the era of human immunodeficiency virus (HIV) infection, when the number of cases of PML markedly increased. We are now entering a new era in which PML is being observed in patients treated with biologic agents for diseases not associated with development of PML.

This article reviews the epidemiology and symptoms that characterize PML, the identification of lesions on radiographic imaging that support the diagnosis, the value of laboratory studies and immunocytochemistry in the diagnosis, and clinical outcomes.

CHANGING EPIDEMIOLOGY OF PML

Figure. Although certain findings are considered classic in progressive multifocal leukoencephalopathy (PML) patients, the percentage of patients affected has shifted as the disorder has evolved from the pre–acquired immunodeficiency syndrome (AIDS) era to PML associated with biologic therapies such as natalizumab. Prior to the AIDS era, nearly one-half of patients with PML were affected by visual deficits. Among patients with AIDS-related PML, motor weakness is a feature for more than one-half, while cognitive impairment is the most common clinical presentation of natalizumab-related PML, affecting nearly one-half of patients.1,2,6,9
The presentation and epidemiology of PML have evolved over the last several decades. Prior to the acquired immunodeficiency syndrome (AIDS) era, nearly one-half of patients with PML were affected by visual deficits. Among patients with AIDS-related PML, motor weakness is a feature for more than one-half, while cognitive impairment is the most common clinical presentation in natalizumab-related PML, affecting nearly one-half of patients (Figure).

The pre-AIDS era

Lesions of subcortical white matter characterize PML and the patient’s clinical manifestations reflect their location. Brooks and Walker1 reviewed 69 pathologically confirmed and 40 virologically and pathologically confirmed cases of PML in the era before AIDS, and categorized the neurologic signs and symptoms at onset and during disease progression; the clinical picture had three significant findings:

  • Impaired vision: Defective vision, most commonly homonymous hemianopsia, was the most frequent presenting sign, present in 35% to 45% of cases. At the time of diagnosis, 6% to 8% of the patients were cortically blind because of bioccipital pathology.
  • Motor weakness: Motor weakness was the initial sign in 25% to 33% of patients. At the time of diagnosis, hemiparesis or hemiplegia was present in nearly all patients.
  • Changes in mentation: A change in mentation, including personality change, difficulty with memory, emotional lability, and frank dementia, was the presenting sign in approximately one-third of cases and eventually involved most patients.

AIDS-related PML

The epidemiology of PML changed with the AIDS pandemic. From 1958 to 1984, Brooks and Walker1 identified 230 cases of PML; in the period from 1981 to 1994, Berger and colleagues2 described 154 cases of AIDS-related PML that had been identified by the University of Miami Medical Center and the Broward County medical examiner’s office. The frequency of PML from 1991 through 1994 was 12-fold greater than the frequency 10 years earlier, from 1981 through 1984. Among the patients with AIDS-related PML, the most common initial symptoms were weakness (42%), speech abnormalities (40%), cognitive abnormalities (36%), gait abnormalities (29%), sensory loss (19%), and visual impairment (19%), followed by seizures, diplopia, and limb incoordination. The most common findings at the time of initial physical examination were weakness (54%), followed by gait abnormalities (20%), cognitive abnormalities (20%), dysarthria (24%), aphasia (19%), sensory loss (19%), visual impairment (17%), and oculomotor palsy (6%). For about 5% of patients with PML, it is the heralding manifestation of AIDS.

Although clinical features consistent with cerebral hemisphere lesions are most common, brainstem and cerebellar findings are also observed. Among these are ataxia, dysmetria, dysarthria, and oculomotor nerve palsies.2–4 Other signs and symptoms associated with PML include headache, vertigo, seizures, sensory deficits, parkinsonism, 5 aphasia, and neglect syndromes.1–4 In some cases, the coexistence of encephalitis with HIV infection could have accounted for some of the symptoms.

PML associated with monoclonal antibody therapy

Natalizumab is an alpha-4-beta-1 integrin inhibitor approved for the treatment of relapsing-remitting multiple sclerosis (MS); patients taking natalizumab represent the second largest group with PML (the largest group is patients with AIDS). Natalizumab-associated PML has some noteworthy features. The most common clinical presentations are cognitive disorders (48%), motor abnormalities (37%), language disturbances (31%), and visual defects (26%). Lesions are often monofocal rather than multifocal and the most common site of involvement is the frontal lobe.6 Among MS patients with natalizumab-associated PML, 30% to 40% have gadolinium-enhancing lesions on magnetic resonance imaging (MRI) at the time of diagnosis.

IMMUNE RECONSTITUTION INFLAMMATORY SYNDROME

The immune reconstitution inflammatory syndrome (IRIS) is a paradoxical clinical deterioration that typically occurs in the face of immunologic recovery (Table).7,8 Although not unique to PML or opportunistic infections, it is frequently observed in the setting of HIV infection and natalizumab-associated PML, and it may be concurrent with the diagnosis of PML.6,9

Among patients with HIV, predictors for the development of IRIS include antiretroviral naiveté, profoundly low CD4 lymphocyte counts (< 50 cells/mm3), a rapid decrease in HIV load, and the presence of active or subclinical opportunistic infections at the time of initiation of combined antiretroviral therapy. Tan and colleagues8 have reported the largest series to date. Of the 54 patients in their series, 36 developed PML and IRIS simultaneously, and 18 had worsening of preexisting PML. Although some investigators have recommended corticosteroid therapy for PML-IRIS, no controlled trials have been conducted and caution has been advised, particularly in patients without contrast enhancement on MRI or mass effect.

DIAGNOSTIC TESTING: NEUROIMAGING, CEREBROSPINAL FLUID ANALYSIS

Neuroimaging, including computed tomography (CT) and MRI, is a useful diagnostic tool for investigating a patient with PML. Cerebrospinal fluid (CSF) analysis for the presence of JC virus (JCV) may play a significant role, but it primarily serves to rule out other illnesses.

Computed tomography: lesion size may not reflect clinical status

On CT, demyelinating lesions appear as subcortical hypodensities, often with a propensity for parietooccipital areas that are confined to the white matter at the junction interface of the gray-white junction of the cortex.9–11 Lesions may be seen in the corpus callosum, thalamus, and basal ganglia,9 but changes in the size of lesions observed on CT do not necessarily reflect clinical progression.12 Prior to the availability of highly active antiretroviral therapy (HAART) for the treatment of AIDS, mass effect was exceptionally rare. However, the development of IRIS with PML, typically in AIDS patients following the use of HAART, may be associated with edema.13 Single-dose intravenous contrast and delayed, double-dose contrast CT scanning enhancement is observed in a minority of patients, typically fewer than 10%.8 This enhancement is generally faint and peripherally located.

Magnetic resonance imaging may show lesions before clinical disease

MRI is vastly more sensitive than CT in detecting the demyelinating lesions of PML.9,14 On rare occasions, MRI will clearly demonstrate pathology when CT is normal. In fact, MRI may show lesions in advance of clinically apparent disease.15 The characteristics of these lesions are hyperintensity on T2-weighted imaging, fluid-attenuated inversion recovery sequences, and hypointensity on T1-weighted image. Apparent diffusion coefficients (ADC) on MRI are typically normal to low in new lesions and at the advancing edge of lesions; the ADC was typically higher in the center of lesions.16

As observed on CT, approximately 10% of patients exhibit a faint rim of gadolinium enhancement.2,9 Enhancement is more common with PML-IRIS, and the distribution of lesions parallels what is seen pathologically. Enhancement PML lesions have altered signal characteristics compared with the surrounding white matter.9,17–19 In contrast, 15% of HIV-associated PML showed gadolinium enhancement on MRI at the time of diagnosis.6,9

Cerebrospinal fluid analysis

With the exception of polymerase chain reaction (PCR) for JCV, the primary utility of lumbar puncture in the setting of possible PML is to exclude the presence of other illnesses, including treatable infections.

CSF findings in patients with PML are nonspecific, with most patients demon strating a normal profile. A mild lymphocytic pleocytosis, which is rarely (if ever) more than 25 leukocytes/mL, occurs in 15% of patients. Total protein level is mildly elevated in approximately 20% to 30% of patients.

The CSF examination in HIV-infected patients with PML may reflect changes associated with HIV: low-grade lymphocytic pleocytosis (< 20 cells/mm3), mildly elevated protein (< 65 mg/dL), and elevated immunoglobulin G and oligoclonal bands. These abnormalities should not be attributed to PML.

 

 

DIAGNOSIS

The most reliable and accurate method for the diagnosis of PML remains brain biopsy that demonstrates the characteristic triad of histopathologic findings (demyelination, bizarre astrocytes, and enlarged oligodendrocyte nuclei) coupled with evidence of JCV infection. With respect to the latter, in situ hybridization or immunocytochemistry can be employed. In situ DNADNA hybridization is a method of annealing JCV DNA to complementary strands either in paraffin-embedded tissue or in frozen sections from biopsy samples.

In immunocytochemistry, antibodies to both T antigen and the common polyomavirus capsid antigen are used to detect cells undergoing productive viral infection. Cells that are positive by in situ hybridization are in a stage of active viral replication. Cells positive by immunocytochemistry that are expressing viral capsid antigens are in a stage of viral transcription and translation (ie, undergoing productive infection). In addition to their utility in confirming a diagnosis of PML, these techniques have demonstrated the presence of JCV in perivascular locations and at sites distant from foci of demyelination. Alternatively, PCR may be used to demonstrate JCV in brain tissue.

In the absence of biopsy, which few deem necessary today, a widely employed approach to diagnosis requires the demonstration of:

  • JCV in the CSF by PCR
  • Compatible clinical presentation
  • An MRI finding consistent with PML
  • No other alternative diagnosis.

With an ultrasensitive PCR technique, sensitivities should approach or may exceed 95%, but PCR sensitivity remains at 75% in some laboratories. Because the viral copy numbers in the CSF may be low, particularly in a patient treated with a monoclonal antibody such as natalizumab, the CSF PCR may be falsely negative.

If clinical suspicion of the disease remains high in the face of an initially negative CSF JCV, the CSF analysis should be repeated. CSF analysis for JCV is approximately 99% specific, but recent studies demonstrating low copy numbers of JCV in the CSF of patients with MS have raised concerns about potential pitfalls of this assay.20

PROGNOSIS

Until recently, PML was regarded as virtually universally fatal. The mean survival in the pre-AIDS era was approximately 6 months, and mortality was 80% within 9 months of disease onset. Rarely, patients had long survivals that ranged from 5 years to 19 years.

In the early years of the AIDS era, survival with PML did not appear to differ significantly from that observed in the pre-AIDS years. In the largest study of HIV-associated PML in the era prior to HAART, the median survival was 183 days.2 However, the majority of individuals were dead within 3 months of diagnosis. Only 8% to 10% of patients survived longer than 12 months, which has been regarded as “prolonged survival.” This long survival skewed the mean and median survival rates in this population.

Several factors have since been identified that correlate with prolonged survival in HIV-associated PML, including PML as the heralding manifestation of HIV, CD4 counts exceeding 300 cells per mm3, contrast enhancement of the lesions on radiographic imaging, low copy number or decreasing JCV titers in CSF,21–24 and the presence of JCV-specific cytotoxic T cells.25 A better prognosis has also been postulated for higher CSF levels of macrophage chemoattractant protein-126 and PML associated with JCV VP1 loop-specific polymorphisms.27

Prognosis of HIV-associated PML improves with immune system restoration

In the era of HAART, not only has the incidence of HIV-associated PML declined, but the prognosis of affected patients has improved as well. This development highlights the importance of restoration of the immune system in both disease prevention and survival. Some estimate that as many as 50% of HAART-treated patients with PML exhibit prolonged survival. In one study of 25 patients, the median survival was more than 46 weeks.28

Nonetheless, PML continues to have the worst prognosis of any AIDS-related cerebral disorder, with those having advanced immunosuppression being most susceptible to the disorder. For AIDS patients with PML, those who were HAART-naïve at the time of diagnosis appear to have better survival than treatment-experienced patients.29 Survival also correlates with reduced JCV load in the CSF30 and improved CD4 lymphocyte counts (CD4 counts > 100 cells/mm3).31

Prognosis of natalizumab-associated PML is different

The prognosis of natalizumab-associated PML differs from that of HIV-associated PML. In a series of 35 patients, 25 (71%) patients were alive on average 6 months after diagnosis.32 Prognosis was worse with a longer time to diagnosis and the presence of widespread disease.

Most deaths in patients taking natalizumab who developed PML have occurred during IRIS. Steroid treatment of IRIS appears to improve prognosis,8 but no scientifically rigorous study has been undertaken to demonstrate this recommendation.7 Among the survivors, neurologic deficit was mild in one-third, moderate in one-third, and severe in one-third of patients.

CONCLUSION: DISPELLING SOME MYTHS

Several assumptions about PML are not necessarily true. For example, although PML implies the presence of multifocal lesions as a characteristic of the disease, the lesions may be monofocal, especially with natalizumab-associated PML. The lesions of PML may show early gadolinium enhancement on neuroimaging. Although lesions typically are seen in subcortical white matter, cortical involvement also may be observed. Cerebellar granular cell degeneration may occur in association with PML or in isolation. Disease progression and death are not inevitable, even in the absence of treatment. The most important determinant for survival is restoration of the immune system.

DISCUSSION

Dr. Calabrese: Why are sensory deficits so common?

Dr. Berger: We don’t know. Because we see involvement in the parietal lobe, we would anticipate observing sensory deficits. I think that a lot of sensation occurs deep in the thalamic area, which is not often involved in PML. Also, we often don’t test for some of the deficits that may occur.

Dr. Rudick: Do you know of any cases of natalizumab-associated PML detected as an incidental finding on MRI, making a case for screening MRI in patients without clinical symptoms?

Dr. Berger: There have been a handful of cases, including one of the seminal cases of natalizumab-associated PML, in which MRI abnormalities were observed in advance of clinically recognized symptomatology.

Dr. Calabrese: The correlate question is, if a patient with a risk factor—be it HIV or treatment with a biologic agent—has a common neurocognitive sign or perhaps some subtle motor findings, does a normal MRI have 100% negative predictive value?

Dr. Berger: I have yet to see somebody with PML who has a normal MRI.

Dr. Simpson: What you may see are lesions that are not typical MRI lesions of white matter hypointensity. In some cases, as Dr. Berger mentioned in his summary, we’ll see cerebellar degeneration—atrophy—but not necessarily white matter lesions.

References
  1. Brooks BR, Walker DL. Progressive multifocal leukoencephalopathy. Neurol Clin 1984; 2:299313.
  2. Berger JR, Pall L, Lanska D, Whiteman M. Progressive multifocal leukoencephalopathy in patients with HIV infection. J Neurovirol 1998; 4:5968.
  3. Parr J, Horoupian DS, Winkelman AC. Cerebellar form of progressive multifocal leukoencephalopathy (PML). Can J Neurol Sci 1979; 6:123128.
  4. Jones HR, Hedley-Whyte ET, Freidberg SR, Kelleher JE, Krolikowski J. Primary cerebellopontine progressive multifocal leukoencephalopathy diagnosed premortem by cerebellar biopsy. Ann Neurol 1982; 11:199202.
  5. O’Riordan S, McGuigan C, Farrell M, Hutchinson M. Progressive multifocal leucoencephalopathy presenting with parkinsonism. J Neurol 2003; 250:13791381.
  6. Clifford DB, De Luca A, Simpson DM, Arendt G, Giovannoni G, Nath A. Natalizumab-associated progressive multifocal leukoencephalopathy in patients with multiple sclerosis: lessons from 28 cases. Lancet Neurol 2010; 9:438446.
  7. Berger JR. Steroids for PML-IRIS. A double-edged sword? Neurology 2009; 72:14541455.
  8. Tan K, Roda R, Ostrow L, McArthur J, Nath A. PML-IRIS in patients with HIV infection: clinical manifestations and treatment with steroids. Neurology 2009; 72:14581464.
  9. Whiteman ML, Post MJ, Berger JR, Tate LG, Bell MD, Limonte LP. Progressive multifocal leukoencephalopathy in 47 HIV-seropositive patients: neuroimaging with clinical and pathologic correlation. Radiology 1993; 187:233240.
  10. Conomy JP, Weinstein MA, Agamanolis D, Holt WS. Computed tomography in progressive multifocal leukoencephalopathy. AJR Am J Roentgenol 1976; 127:663665.
  11. Huckman MS. Computed tomography in the diagnosis of degenerative brain disease. Radiol Clin North Am 1982; 20:169183.
  12. Krupp LB, Lipton RB, Swerdlow ML, Leeds NE, Llena J. Progressive multifocal leukoencephalopathy: clinical and radiographic features. Ann Neurol 1985; 17:344349.
  13. Rushing EJ, Liappis A, Smirniotopoulos JD, et al. Immune reconstitution inflammatory syndrome of the brain: case illustrations of a challenging entity. J Neuropathol Exp Neurol 2008; 67:819827.
  14. Post MJ, Sheldon JJ, Hensley GT, et al. Central nervous system disease in acquired immunodeficiency syndrome: prospective correlation using CT, MR imaging, and pathologic studies. Radiology 1986; 158:141148.
  15. Langer-Gould A, Atlas SW, Green AJ, Bollen AW, Pelletier D. Progressive multifocal leukoencephalopathy in a patient treated with natalizumab. N Engl J Med 2005; 353:375381.
  16. Bergui M, Bradac GB, Oguz KK, et al. Progressive multifocal leukoencephalopathy: diffusion-weighted imaging and pathological correlations. Neuroradiology 2004; 46:2225.
  17. Guilleux MH, Steiner RE, Young IR. MR imaging in progressive multifocal leukoencephalopathy. AJNR Am J Neuroradiol 1986; 7:10331035.
  18. Levy JD, Cottingham KL, Campbell RJ, et al. Progressive multifocal leukoencephalopathy and magnetic resonance imaging. Ann Neurol 1986; 19:399401.
  19. Mark AS, Atlas SW. Progressive multifocal leukoencephalopathy in patients with AIDS: appearance on MR images. Radiology 1989; 173:517520.
  20. Iacobaeus E, Ryschkewitsch C, Gravell M, et al. Analysis of cerebrospinal fluid and cerebrospinal fluid cells from patients with multiple sclerosis for detection of JC virus DNA. Mult Scler 2009; 15:2835.
  21. Berger JR, Levy RM, Flomenhoft D, Dobbs M. Predictive factors for prolonged survival in acquired immunodeficiency syndrome-associated progressive multifocal leukoencephalopathy. Ann Neurol 1998; 44:341349.
  22. Taoufik Y, Gasnault J, Karaterki A, et al. Prognostic value of JC virus load in cerebrospinal fluid of patients with progressive multifocal leukoencephalopathy. J Infect Dis 1998; 178:18161820.
  23. Yiannoutsos CT, Major EO, Curfman B, et al. Relation of JC virus DNA in the cerebrospinal fluid to survival in acquired immunodeficiency syndrome patients with biopsy-proven progressive multifocal leukoencephalopathy. Ann Neurol 1999; 45:816821.
  24. Bossolasco S, Calori G, Moretti F, et al. Prognostic significance of JC virus DNA levels in cerebrospinal fluid of patients with HIV-associated progressive multifocal leukoencephalopathy. Clin Infect Dis 2005; 40:738744.
  25. Du Pasquier RA, Clark KW, Smith PS, et al. JCV-specific cellular immune response correlates with a favorable clinical outcome in HIV-infected individuals with progressive multifocal leukoencephalopathy. J Neurovirol 2001; 7:318322.
  26. Marzocchetti A, Cingolani A, Giambenedetto SD, et al. Macrophage chemoattractant protein-1 levels in cerebrospinal fluid correlate with containment of JC virus and prognosis of acquired immunodeficiency syndrome-associated progressive multifocal leukoencephalopathy. J Neurovirol 2005; 11:219224.
  27. Delbue S, Branchetti E, Bertolacci S, et al. JC virus VP1 loop-specific polymorphisms are associated with favorable prognosis for progressive multifocal leukoencephalopathy. J Neurovirol 2009; 15:5156.
  28. Clifford DB, Yiannoutsos C, Glicksman M, et al. HAART improves prognosis in HIV-associated progressive multifocal leukoencephalopathy. Neurology 1999; 52:623625.
  29. Wyen C, Hoffmann C, Schmeisser N, et al. Progressive multi focal leukoencephalopathy in patients on highly active antiretroviral therapy: survival and risk factors of death. J Acquir Immune Defic Syndr 2004; 37:12631268.
  30. De Luca A, Giancola ML, Ammassari A, et al. The effect of potent antiretroviral therapy and JC virus load in cerebrospinal fluid on clinical outcome of patients with AIDS-associated progressive multifocal leukoencephalopathy. J Infect Dis 2000; 182:10771083.
  31. Berenguer J, Miralles P, Arrizabalaga J, et al. Clinical course and prognostic factors of progressive multifocal leukoencephalopathy in patients treated with highly active antiretroviral therapy. Clin Infect Dis 2003; 36:10471052.
  32. Vermersch P, Kappos L, Gold R, et al. Clinical outcomes of natalizumab associated progressive multifocal leukoencephalopathy. Neurology 2011; 76:16971704.
References
  1. Brooks BR, Walker DL. Progressive multifocal leukoencephalopathy. Neurol Clin 1984; 2:299313.
  2. Berger JR, Pall L, Lanska D, Whiteman M. Progressive multifocal leukoencephalopathy in patients with HIV infection. J Neurovirol 1998; 4:5968.
  3. Parr J, Horoupian DS, Winkelman AC. Cerebellar form of progressive multifocal leukoencephalopathy (PML). Can J Neurol Sci 1979; 6:123128.
  4. Jones HR, Hedley-Whyte ET, Freidberg SR, Kelleher JE, Krolikowski J. Primary cerebellopontine progressive multifocal leukoencephalopathy diagnosed premortem by cerebellar biopsy. Ann Neurol 1982; 11:199202.
  5. O’Riordan S, McGuigan C, Farrell M, Hutchinson M. Progressive multifocal leucoencephalopathy presenting with parkinsonism. J Neurol 2003; 250:13791381.
  6. Clifford DB, De Luca A, Simpson DM, Arendt G, Giovannoni G, Nath A. Natalizumab-associated progressive multifocal leukoencephalopathy in patients with multiple sclerosis: lessons from 28 cases. Lancet Neurol 2010; 9:438446.
  7. Berger JR. Steroids for PML-IRIS. A double-edged sword? Neurology 2009; 72:14541455.
  8. Tan K, Roda R, Ostrow L, McArthur J, Nath A. PML-IRIS in patients with HIV infection: clinical manifestations and treatment with steroids. Neurology 2009; 72:14581464.
  9. Whiteman ML, Post MJ, Berger JR, Tate LG, Bell MD, Limonte LP. Progressive multifocal leukoencephalopathy in 47 HIV-seropositive patients: neuroimaging with clinical and pathologic correlation. Radiology 1993; 187:233240.
  10. Conomy JP, Weinstein MA, Agamanolis D, Holt WS. Computed tomography in progressive multifocal leukoencephalopathy. AJR Am J Roentgenol 1976; 127:663665.
  11. Huckman MS. Computed tomography in the diagnosis of degenerative brain disease. Radiol Clin North Am 1982; 20:169183.
  12. Krupp LB, Lipton RB, Swerdlow ML, Leeds NE, Llena J. Progressive multifocal leukoencephalopathy: clinical and radiographic features. Ann Neurol 1985; 17:344349.
  13. Rushing EJ, Liappis A, Smirniotopoulos JD, et al. Immune reconstitution inflammatory syndrome of the brain: case illustrations of a challenging entity. J Neuropathol Exp Neurol 2008; 67:819827.
  14. Post MJ, Sheldon JJ, Hensley GT, et al. Central nervous system disease in acquired immunodeficiency syndrome: prospective correlation using CT, MR imaging, and pathologic studies. Radiology 1986; 158:141148.
  15. Langer-Gould A, Atlas SW, Green AJ, Bollen AW, Pelletier D. Progressive multifocal leukoencephalopathy in a patient treated with natalizumab. N Engl J Med 2005; 353:375381.
  16. Bergui M, Bradac GB, Oguz KK, et al. Progressive multifocal leukoencephalopathy: diffusion-weighted imaging and pathological correlations. Neuroradiology 2004; 46:2225.
  17. Guilleux MH, Steiner RE, Young IR. MR imaging in progressive multifocal leukoencephalopathy. AJNR Am J Neuroradiol 1986; 7:10331035.
  18. Levy JD, Cottingham KL, Campbell RJ, et al. Progressive multifocal leukoencephalopathy and magnetic resonance imaging. Ann Neurol 1986; 19:399401.
  19. Mark AS, Atlas SW. Progressive multifocal leukoencephalopathy in patients with AIDS: appearance on MR images. Radiology 1989; 173:517520.
  20. Iacobaeus E, Ryschkewitsch C, Gravell M, et al. Analysis of cerebrospinal fluid and cerebrospinal fluid cells from patients with multiple sclerosis for detection of JC virus DNA. Mult Scler 2009; 15:2835.
  21. Berger JR, Levy RM, Flomenhoft D, Dobbs M. Predictive factors for prolonged survival in acquired immunodeficiency syndrome-associated progressive multifocal leukoencephalopathy. Ann Neurol 1998; 44:341349.
  22. Taoufik Y, Gasnault J, Karaterki A, et al. Prognostic value of JC virus load in cerebrospinal fluid of patients with progressive multifocal leukoencephalopathy. J Infect Dis 1998; 178:18161820.
  23. Yiannoutsos CT, Major EO, Curfman B, et al. Relation of JC virus DNA in the cerebrospinal fluid to survival in acquired immunodeficiency syndrome patients with biopsy-proven progressive multifocal leukoencephalopathy. Ann Neurol 1999; 45:816821.
  24. Bossolasco S, Calori G, Moretti F, et al. Prognostic significance of JC virus DNA levels in cerebrospinal fluid of patients with HIV-associated progressive multifocal leukoencephalopathy. Clin Infect Dis 2005; 40:738744.
  25. Du Pasquier RA, Clark KW, Smith PS, et al. JCV-specific cellular immune response correlates with a favorable clinical outcome in HIV-infected individuals with progressive multifocal leukoencephalopathy. J Neurovirol 2001; 7:318322.
  26. Marzocchetti A, Cingolani A, Giambenedetto SD, et al. Macrophage chemoattractant protein-1 levels in cerebrospinal fluid correlate with containment of JC virus and prognosis of acquired immunodeficiency syndrome-associated progressive multifocal leukoencephalopathy. J Neurovirol 2005; 11:219224.
  27. Delbue S, Branchetti E, Bertolacci S, et al. JC virus VP1 loop-specific polymorphisms are associated with favorable prognosis for progressive multifocal leukoencephalopathy. J Neurovirol 2009; 15:5156.
  28. Clifford DB, Yiannoutsos C, Glicksman M, et al. HAART improves prognosis in HIV-associated progressive multifocal leukoencephalopathy. Neurology 1999; 52:623625.
  29. Wyen C, Hoffmann C, Schmeisser N, et al. Progressive multi focal leukoencephalopathy in patients on highly active antiretroviral therapy: survival and risk factors of death. J Acquir Immune Defic Syndr 2004; 37:12631268.
  30. De Luca A, Giancola ML, Ammassari A, et al. The effect of potent antiretroviral therapy and JC virus load in cerebrospinal fluid on clinical outcome of patients with AIDS-associated progressive multifocal leukoencephalopathy. J Infect Dis 2000; 182:10771083.
  31. Berenguer J, Miralles P, Arrizabalaga J, et al. Clinical course and prognostic factors of progressive multifocal leukoencephalopathy in patients treated with highly active antiretroviral therapy. Clin Infect Dis 2003; 36:10471052.
  32. Vermersch P, Kappos L, Gold R, et al. Clinical outcomes of natalizumab associated progressive multifocal leukoencephalopathy. Neurology 2011; 76:16971704.
Page Number
S8-S12
Page Number
S8-S12
Publications
Publications
Article Type
Display Headline
The clinical features of PML
Display Headline
The clinical features of PML
Citation Override
Cleveland Clinic Journal of Medicine 2011 November;78(suppl 2):S8-S12
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media

Pharmacovigilance and PML in the oncology setting

Article Type
Changed
Tue, 10/02/2018 - 09:23
Display Headline
Pharmacovigilance and PML in the oncology setting

Rare and serious drug-related events are often not detected until after clinical trials have been completed and a drug becomes widely used. Methods traditionally used by pharmaceutical companies and the US Food and Drug Administration (FDA) are not the most effective ways to promptly identify a treatment-related adverse event and quickly notify the medical community. In 1998, an academically based surveillance group was created to identify and disseminate information on unrecognized adverse drug reactions.1 In 2010, with funding from the state of South Carolina, this program became the Southern Network on Adverse Reactions (SONAR), the only state-funded pharmacovigilance initiative in the nation. SONAR and its earlier incarnation have identified potentially fatal and previously unreported side effects associated with 43 drugs—with the majority of these drugs involving the hematology and oncology discisplines.

While the earlier incarnation of drug safety monitoring relied on data mining, or detecting specific signals from large amounts of data, investigations of possible adverse drug event occurrences have a much broader scope. SONAR, an enhanced surveillance program, was created to address this issue. Based jointly at the South Carolina College of Pharmacy and a National Cancer Institute–designated cancer center, the Hollings Cancer Center at the Medical University of South Carolina, SONAR more accurately reflects the nature of our adverse effects investigations: identification of small numbers of important cases from a variety of unique data sources, including case reports, the medical literature, FDA MedWatch, and pharmaceutical manufacturers.

This article reviews methods that underlie the successful investigations of the SONAR initiative, and it examines our SONAR investigation of the association between the immune modulatory monoclonal antibody rituximab and progressive multifocal leukoencephalopathy (PML).

DETECTING, INVESTIGATING, AND DISSEMINATING FINDINGS

Surveillance programs are needed because important rare side effects are seldom discovered in a clinical trial. The Safety and Efficacy of Natalizumab in Combination with Interferon Beta-1a in Patients with Relapsing Remitting Multiple Sclerosis (SENTINEL) trial was unusual in that it detected two cases of PML associated with the use of natalizumab.2 Most rare side effects are undetected at the time of FDA approval, and usually many years elapse from the time a potential problem is detected until it is identified as a rare side effect of the drug. The average time for a “black box” warning to appear on a package insert following FDA approval is 7 to 10 years.3

Timely and thorough data collection

Reprinted, with permission, from The Journal of the American Medical Association (Bennett CL, et al. JAMA 2005; 29:211–2140), Copyright © 2005 American Medical Association. All rights reserved.
Figure. Algorithm of Research on Adverse Drug Events and Reports (RADAR) protocol for investigation of adverse events and dissemination of results.3 ADR = adverse drug and device reaction; FDA = US Food and Drug Administration; IRB = institutional review board
Academic pharmacovigilance organizations such as SONAR operate differently from the FDA and pharmaceutical manufacturers in their search for adverse drug events (Figure).4 SONAR collects reports from investigators, clinicians, attorneys, patients, and family members on suspected treatment-related adverse events and investigates these reports carefully. Direct calls to hospitals and large centers can be useful in searching for cases, using information obtained from Internal Review Boards and medical records.

SONAR investigators perform extensive literature reviews, may request more data from authors, and request and review additional FDA case reports. Unfortunately, obtaining data from the FDA can be difficult and time-consuming. Data can be requested through the Freedom of Information Act (FOIA), but receiving it may take more than a year, and the information in the public record may be redacted. SONAR obtains laboratory tests and imaging records and works with scientists to better understand the pathophysiology of potential treatment-related rare adverse events, investigate epidemiologic estimates of the side effect rate, and evaluate risk factors for development of toxicity.

Adverse events are usually identified by SONAR within 2 years post–drug approval—a 5-year improvement over the FDA on this important metric. Once an adverse event is positively identified, the information is disseminated throughout the worldwide medical community via journal articles and presentations at medical conferences. Funding is grant-based from sources such as the National Institutes of Health (NIH), the state of South Carolina, and the University of South Carolina.

FDA, manufacturer reports may be incomplete and delayed

In contrast with SONAR, the FDA relies heavily on MedWatch to detect cases of adverse events. The safety record compiled by MedWatch is often incomplete because the program relies on voluntary submissions of adverse events; further, the inordinate amount of followup required of physicians discourages many from participating. The time to identify an adverse event can be several years, and the FDA disseminates adverse event reports via package inserts. The network that evaluates the safety information and identifies initial safety signals is mainly internal to FDA employees, as is the funding.

Pharmaceutical manufacturers frequently compile data from their own proprietary databases. Although they attempt to follow up on reports of rare adverse events, it is often difficult or impossible for the company to obtain followup information from busy clinicians. Identification of an adverse event typically takes 7 to 12 years for most pharmaceutical manufacturers—reflecting the barriers experienced in obtaining detailed information from clinicians about potential new serious adverse drug reactions. Findings are frequently disseminated through “Dear Doctor” letters. Manufacturers’ investigative networks, like those of the FDA, are largely internal and the amount of funding of they allocate to drug safety investigations is unknown.

RARE EVENTS MAY INVOLVE FEW CASES

Of our major publications,5–14 many findings are based on a small number of cases—for example, only 13 cases for clopidogrel-associated thrombotic thrombocytopenic purpura (TTP)13 and 9 for pure red cell aplasia caused by epoetin alfa.12 Important findings also come from meta-analyses,8,10 although this avenue in our pharmacovigilance approach is less typical.

The 2008 study6 on mortality and venous thromboembolism associated with erythropoiesis-stimulating agents highlights the importance of basic scientific investigation in identifying rare events. Administration of epoetin alfa to raise hemoglobin levels had been approved by the FDA in 1989 for use in patients undergoing dialysis and in 1993 for supportive use in patients with some types of cancers. We discovered that epoetin alfa promoted cancer growth based on analysis of published data and reports in conjunction with basic scientific studies of erythropoietin and erythropoietin receptors in solid cancers.

 

 

RITUXIMAB AND VIRAL REACTIVATION

In the case of viral reactivation associated with the use of rituximab, a warning about hepatitis B reactivation was added to the package insert in 2004.15 In 2006, a warning about other viral infections was added to the package insert.16 In late 2006, a letter was sent to health care professionals from the manufacturer and the FDA with the warning that PML had been observed in two patients with systemic lupus erythematosus (SLE) who were treated with rituximab (an off-label use), both of whom were negative for human immunodeficiency virus (HIV).16 A few months later, a black box warning to this effect was added to the package insert.16

After we identified PML as an adverse event from rituximab in HIV-negative patients,14 we obtained case reports from clinicians at 12 cancer centers or academic hospitals (22 cases). We also reviewed FDA reports (11 cases), the manufacturer’s database (30 cases), and publications (18 cases) using the search terms “leukoencephalopathy,” “rituximab,” “immunosuppressed,” “lymphoma,” and “leukemia.” The unique data sources included clinical observations, the medical literature, FDA MedWatch, and the manufacturer.17

Of rituximab-treated patients who developed PML, the mean age was 61 years (range, 30 to 89 years), 56% of patients were women, and the mean number of rituximab doses was six (range, 1 to 28). Six patients had undergone stem cell transplants (four autologous), and 26 were also taking a purine analogue.17

Among 57 patients, a median of 16 months elapsed between first taking rituximab to development of PML (range, 1.0 to 90.0 months), and 5.5 months from the last dose of rituximab to development of PML (range, 0.3 to 66.0 months). The median time from diagnosis of PML to death was only 2.0 months (range, 0.4 to 122 months). Reported survival rates for patients with rituximab-associated PML who did not undergo stem cell transplantation was less than 10%.17

The symptoms of PML are easily confused with those that might be expected in an older patient with lymphoma, making early detection especially difficult. More than one-half (54.4%) had confusion or disorientation, and many had focal motor weakness (33.3%), loss of coordination (24.6%), difficulty speaking (21.2%), and vision changes (17.5%).

Effects on T and B cells and role of JC virus

At the time of PML diagnosis, 90% of patients had either a severely low CD4+ count or a low CD4+:CD8+ ratio. Based on clinical trial data, cytotoxic chemotherapy and not rituximab appears responsible for the abnormal CD4+ count and the low CD4+:CD8+ ratio in rituximab-treated patients.

Little is known about how T cells function after rituximab administration. In idiopathic thrombocytopenic purpura, the response to B-cell depletion induced by rituximab is associated with significant changes in the T-cell compartment.18 In a study of patients with either SLE or Evans syndrome, rituximab therapy was found to modify T-cell phenotype and cytokine profiles.19 The rapid effect of rituximab in multiple sclerosis suggests that it targets a process thought to be T-cell mediated.20

Our early hypothesis was that rituximab contributes to viral reactivation and PML through inhibiting T-and B-lymphocyte interactions. We now believe that the bone marrow plays an important role, which may explain the process by which natalizumab can cause PML. Five of five bone marrow samples from patients with lymphoma and PML tested positive for JC virus (JCV) compared with only two of 86 bone marrow samples from patients without PML. The JCV is latent in CD34+ hematopoietic cells and probably in early B lymphocytes. Chemotherapy mobilizes the stem cells from bone marrow and causes quantitative T-cell depletion. Rituximab reduces the qualitative T-cell response, and B-cell depletion results in expansion of progenitor cells containing the latent JCV. The hypothesis is limited in that it is based on a retrospective case series and is not verified in a laboratory model.

Of the 57 cases of PML identified in 2009, two patients were given rituximab for hematologic disorders and had no chemotherapy other than steroids. These data suggest that rituximab confers risk on its own.17

Quantifying risk of developing PML from rituximab

Calculating the odds of developing PML from rituximab therapy is difficult. The background rate of PML is an important consideration. One population-based study estimated the incidence of PML in patients with hematologic malignancies at 0.07%. This estimate was based on three cases of PML observed in patients with hematologic malignancies over a period of 11 years in a single Canadian province.21 Another study found a higher incidence of 0.52% in patients with chronic lymphocytic leukemia, although all of these patients were also treated with fludarabine.22 Accurately calculating the risk of PML attributable to the underlying malignancy as opposed to immune suppression from treatment is complicated by the rarity of the disease. Fludarabine is the chemotherapeutic agent most closely associated with PML. However, its well known side effects of T-lymphocyte depletion and complicating opportunistic infections similar to those seen in acquired immunodeficiency syndrome (AIDS) make such an association intuitive.23

Kavenaugh and Matteson reported that about 8,000 SLE patients had received rituximab treatment and two of these patients had developed PML.24 PML has been reported previously among 30 SLE patients who had not received rituximab, suggesting that SLE is a predisposing disorder.25,26

In the setting of hematologic malignancy, rituximab-associated PML incidence estimates are complicated by a low basal risk of PML seen among persons with the disease state prompting rituximab therapy and an inability to determine risk attributable to rituximab. A recent study demonstrated an association between rituximab and PML in patients with non-Hodgkin lymphoma (NHL). The retrospective, monocentric cohort study assessed data from 976 NHL patients diagnosed in Italy from 1994 to 2008, including 517 patients who received at least one dose of rituximab. Inclusion of rituximab into standard chemotherapy regimens for NHL caused a significantly higher incidence of PML cases (rate difference, 2.2 every 1,000 patient-years; 95% confidence interval, 0.1–4.3).27 More such studies of viral reactivation syndromes are obviously needed.

Ideally, randomized clinical trials of the use of rituximab in patients with lymphoma would serve as guidance, but because the drug, as the standard of care for treatment of lymphoma, is so widely used, randomization would be impractical.

Future planned studies include a case-control study of T-cell markers after chemotherapy administration with or without exposure to rituximab, a case-control study of bone marrow specimens from disease-matched and treatment-matched controls, and a cohort study using a large electronic medical records database or a government database.

CONCLUSION

The methods developed in the SONAR project will permit exploration of important hypotheses regarding the detection and prevention of rare adverse events in oncology, forming a basis for subsequent investigations. Based on our recent findings, rituximab may be associated with multiple viral reactivation syndromes; screening and early detection can potentially be helpful in preventing these complications.

DISCUSSION

Dr. Calabrese: Your approach to identifying rare adverse events is novel and aggressive, but the seeming limitations in a disease such as lymphoma are (1) rituximab is now a standard of care so everybody with lymphoma gets it, and (2) going back to the earliest descriptions of PML, lymphoma has always been represented as a predisposing factor. Moving ahead, how then can you calculate an effect size for a drug like rituximab?

Dr. Bennett: There’s no way to do it; we’re sort of stuck. Of the 57 cases with PML that we reported in Blood,17 two patients received rituximab for hematologic disorders and received no chemotherapy besides steroids. In those two patients, we could not blame the development of PML on lymphoma. Those types of patients suggest that rituximab may be implicated, but examining this question with a case-control or even a cohort study is an expensive proposition.

Dr. Simpson: My experience in terms of collaborating with the FDA has been distinctly unrewarding. Some years ago I had been looking into an adverse effect related to the nucleoside analog reverse transcriptase inhibitor d4T, in which there was a rapidly progressive neuromuscular weakness syndrome that looked like Guillain-Barré and lactic acidosis. The FDA itself reported 12 cases at an international AIDS conference and did not have any answers. I was charged by the AIDS Clinical Trials Group and other branches at the NIH to try to figure it out. When I requested access to FDA data, I ran into an unbelievable bureaucratic morass. Ultimately, we had to go through the FOIA to get them to release anything.

Dr. Bennett: The FOIA is the only way to get anything from the FDA. It takes about a year and a half and much information is redacted.

Dr. Berger: As we roll out these newer compounds, we need a mechanism to look for both foreseen and unforeseen consequences, perhaps with close collaboration between pharmaceutical companies and governmental agencies.

Dr. Bennett: The Risk Evaluation and Mitigation Strategies program authorizes the FDA to require post-marketing surveillance of all adverse events from manufacturers. We published 11 cases of TTP in association with clopidogrel, obtained from surveillance of directors of plasmapheresis centers in the United States. Not one of them had been reported to the FDA directly. However, we had an article 6 weeks after clopidogrel received FDA approval. Now, 10 years later, there are about 120 clopidogrel-associated TTP cases in the FDA database. Its estimated incidence is still one in a million, although we hear about the side effect every night on TV during commercials for the drug on the evening news.

Dr. Major: The FDA is more open now than in the past to trying to get a handle on what’s going on with biologic therapies. We need to do a little more homework up front on biologic agents in order to anticipate some adverse events. For example, the migratory nature of CD34+ cells through the circulation following natalizumab therapy was not appreciated, even though data in the literature already supported this phenomenon when integrin receptors are blocked.

References
  1. Hampton T. Postmarket “pharmacovigilance” program on alert for adverse events from drugs. JAMA 2007; 298:851852.
  2. Rudick RA, Stuart WH, Calabresi PA, et al. Natalizumab plus interferon beta-1a for relapsing multiple sclerosis. N Engl J Med 2006; 354:911923.
  3. Lasser KE, Allen PD, Woolhandler SJ, Himmelstein Du, Wolfe SM, Bor DH. Timing of new black box warnings and withdrawals for prescription medications. JAMA 2002; 287:22152220.
  4. Bennett CL, Nebeker JR, Lyons EA, et al. The Research on Adverse Drug Events and Reports (RADAR) project. JAMA 2005; 293:21312140.
  5. Edwards BJ, Gounder M, McKoy JM, et al. Pharmacovigilance and reporting oversight in US FDA fast-track process: bisphosphonates and osteonecrosis of the jaw. Lancet Oncol 2008; 9:11661172.
  6. Bennett CL, Silver SM, Djulbegovic B, et al. Venous thromboembolism and mortality associated with recombinant erythropoietin and darbepoetin administration for the treatment of cancer-associated anemia. JAMA 2008; 299:914924.
  7. Beohar N, Davidson CJ, Kip KE, et al. Outcomes and complications associated with off-label and untested use of drug-eluting stents. JAMA 2007; 297:19922000.
  8. Hershman D, Neugut AI, Jacobson JS, et al. Acute myeloid leukemia or myelodysplastic syndrome following use of granulocyte colony-stimulating factors during breast cancer adjuvant chemotherapy. J Natl Cancer Inst 2007; 99:196205.
  9. Bohlius J, Wilson J, Seidenfeld J, et al. Recombinant human erythropoietins and cancer patients: updated meta-analysis of 57 studies including 9353 patients. J Natl Cancer Inst 2006; 98:708714.
  10. Nebeker JR, Virmani R, Bennett CL, et al. Hypersensitivity cases associated with drug-eluting coronary stents: a review of available cases from the Research on Adverse Drug Events and Reports (RADAR) project. J Am Coll Cardiol 2006; 47:175181.
  11. Bennett CL, Angelotta C, Yarnold PR, et al. Thalidomide- and lenalidomide-associated thromboembolism among patients with cancer. JAMA 2006; 296:25582560.
  12. Bennett CL, Luminari S, Nissenson AR, et al. Pure red-cell aplasia and epoetin therapy. N Engl J Med 2004; 351:14031408.
  13. Bennett CL, Connors JM, Carwile JM, et al. Thrombotic thrombocytopenic purpura associated with clopidogrel. N Engl J Med 2000; 342:17731777.
  14. Bennett CL, Kiss JE, Weinberg PD, et al. Thrombotic thrombocytopenic purpura after stenting and ticlopidine. Lancet 1998; 352:10361037.
  15. Rituxan (rituximab) Oct 2004. U.S. Food and Drug Administration Web site. http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsforHumanMedicalProducts/ucm166521.htm. Updated June 19, 2009. Accessed September 13, 2011.
  16. Rituxan (rituximab). U.S. Food and Drug Administration Web site. http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsforHumanMedicalProducts/ucm150747.htm. Updated June 19, 2009. Accessed September 13, 2011.
  17. Carson KR, Evens AM, Richey EA, et al. Progressive multifocal leukoencephalopathy after rituximab therapy in HIV-negative patients: a report of 57 cases from the Research on Adverse Drug Events and Reports project. Blood 2009; 113:48344840.
  18. Stasi R, Del Poeta G, Stipa E, et al. Response to B-cell depleting therapy with rituximab reverts the abnormalities of T-cell subsets in patients with idiopathic thrombocytopenic purpura. Blood 2007; 110:29242930.
  19. Tamimoto Y, Horiuchi T, Tsukamoto H, et al. A dose-escalation study of rituximab for treatment of systemic lupus erythematosus and Evans’ syndrome: immunological analysis of B cells, T cells and cytokines. Rheumatology (Oxford) 2008; 47:821827.
  20. McFarland HF. The B cell—old player, new position on the team. N Engl J Med 2008; 358:664665.
  21. Power C, Gladden JG, Halliday W, et al. AIDS- and non-AIDS-related PML association with distinct p53 polymorphism. Neurology 2000; 54:743746.
  22. Gonzalez H, Bolgert F, Camporo P, Leblond V. Progressive multi focal leukoencephalitis (PML) in three patients treated with standard-dose fludarabine (FAMP). Hematol Cell Ther 1999; 41:183186.
  23. Garcia-Suárez J, de Miguel D, Krsnik I, Bañas H, Arribas I, Burgaleta C. Changes in the natural history of progressive multifocal leukoencephalopathy in HIV-negative lymphoproliferative disorders: impact of novel therapies. Am J Hematol 2005; 80:271281.
  24. Kavanaugh A, Matteson E. Hotline: rituximab and progressive multifocal leukoencephalopathy. American College of Rheumatology Web site. http://www.rheumatology.org/publications/hotline/0107leuko.asp. Updated January 2007. Accessed September 13, 2011.
  25. Molloy ES, Calabrese LH. Progressive multifocal leukoencephalopathy in patients with rheumatic diseases: are patients with systemic lupus erythematosus at particular risk? Autoimmun Rev 2008; 8:144146.
  26. Calabrese LH, Molloy ES. Progressive multifocal leucoencephalopathy in the rheumatic diseases: assessing the risks of biological immunosuppressive therapies. Ann Rheum Dis 2008; 67 (suppl 3):iii64iii65.
  27. Tuccori M, Focosi D, Blandizzi C, et al. Inclusion of rituximab in treatment protocols for non-Hodgkin’s lymphomas and risk for progressive multifocal leukoencephalopathy. Oncologist 2010; 15:12141219. Epub 2010 Nov 1.
Article PDF
Author and Disclosure Information

Charles L. Bennett, MD, PhD, MPP
Endowed Chair, Center of Economic Excellence for Medication Safety and Efficacy; Josie M. Fletcher Professor of Pharmacy; South Carolina College of Pharmacy, Hollings Cancer Center, and Arnold School of Public Health; University of South Carolina and Medical University of South Carolina, Columbia and Charleston, SC

Correspondence: Charles L. Bennett, MD, PhD, MPP, CoEE Endowed Chair in Medication Safety and Efficacy, South Carolina College of Pharmacy/USC Campus, 715 Sumter Street, Columbia, SC 29208; charlesleebennett@gmail.com

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

This article was developed from an audio transcript of Dr. Bennett’s presentation at a roundtable convened at Cleveland Clinic on January 31, 2011. The transcript was edited by the Cleveland Clinic Journal of Medicine staff for clarity and conciseness, and was then reviewed, revised, and approved by Dr. Bennett.

Publications
Page Number
S13-S17
Author and Disclosure Information

Charles L. Bennett, MD, PhD, MPP
Endowed Chair, Center of Economic Excellence for Medication Safety and Efficacy; Josie M. Fletcher Professor of Pharmacy; South Carolina College of Pharmacy, Hollings Cancer Center, and Arnold School of Public Health; University of South Carolina and Medical University of South Carolina, Columbia and Charleston, SC

Correspondence: Charles L. Bennett, MD, PhD, MPP, CoEE Endowed Chair in Medication Safety and Efficacy, South Carolina College of Pharmacy/USC Campus, 715 Sumter Street, Columbia, SC 29208; charlesleebennett@gmail.com

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

This article was developed from an audio transcript of Dr. Bennett’s presentation at a roundtable convened at Cleveland Clinic on January 31, 2011. The transcript was edited by the Cleveland Clinic Journal of Medicine staff for clarity and conciseness, and was then reviewed, revised, and approved by Dr. Bennett.

Author and Disclosure Information

Charles L. Bennett, MD, PhD, MPP
Endowed Chair, Center of Economic Excellence for Medication Safety and Efficacy; Josie M. Fletcher Professor of Pharmacy; South Carolina College of Pharmacy, Hollings Cancer Center, and Arnold School of Public Health; University of South Carolina and Medical University of South Carolina, Columbia and Charleston, SC

Correspondence: Charles L. Bennett, MD, PhD, MPP, CoEE Endowed Chair in Medication Safety and Efficacy, South Carolina College of Pharmacy/USC Campus, 715 Sumter Street, Columbia, SC 29208; charlesleebennett@gmail.com

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

This article was developed from an audio transcript of Dr. Bennett’s presentation at a roundtable convened at Cleveland Clinic on January 31, 2011. The transcript was edited by the Cleveland Clinic Journal of Medicine staff for clarity and conciseness, and was then reviewed, revised, and approved by Dr. Bennett.

Article PDF
Article PDF

Rare and serious drug-related events are often not detected until after clinical trials have been completed and a drug becomes widely used. Methods traditionally used by pharmaceutical companies and the US Food and Drug Administration (FDA) are not the most effective ways to promptly identify a treatment-related adverse event and quickly notify the medical community. In 1998, an academically based surveillance group was created to identify and disseminate information on unrecognized adverse drug reactions.1 In 2010, with funding from the state of South Carolina, this program became the Southern Network on Adverse Reactions (SONAR), the only state-funded pharmacovigilance initiative in the nation. SONAR and its earlier incarnation have identified potentially fatal and previously unreported side effects associated with 43 drugs—with the majority of these drugs involving the hematology and oncology discisplines.

While the earlier incarnation of drug safety monitoring relied on data mining, or detecting specific signals from large amounts of data, investigations of possible adverse drug event occurrences have a much broader scope. SONAR, an enhanced surveillance program, was created to address this issue. Based jointly at the South Carolina College of Pharmacy and a National Cancer Institute–designated cancer center, the Hollings Cancer Center at the Medical University of South Carolina, SONAR more accurately reflects the nature of our adverse effects investigations: identification of small numbers of important cases from a variety of unique data sources, including case reports, the medical literature, FDA MedWatch, and pharmaceutical manufacturers.

This article reviews methods that underlie the successful investigations of the SONAR initiative, and it examines our SONAR investigation of the association between the immune modulatory monoclonal antibody rituximab and progressive multifocal leukoencephalopathy (PML).

DETECTING, INVESTIGATING, AND DISSEMINATING FINDINGS

Surveillance programs are needed because important rare side effects are seldom discovered in a clinical trial. The Safety and Efficacy of Natalizumab in Combination with Interferon Beta-1a in Patients with Relapsing Remitting Multiple Sclerosis (SENTINEL) trial was unusual in that it detected two cases of PML associated with the use of natalizumab.2 Most rare side effects are undetected at the time of FDA approval, and usually many years elapse from the time a potential problem is detected until it is identified as a rare side effect of the drug. The average time for a “black box” warning to appear on a package insert following FDA approval is 7 to 10 years.3

Timely and thorough data collection

Reprinted, with permission, from The Journal of the American Medical Association (Bennett CL, et al. JAMA 2005; 29:211–2140), Copyright © 2005 American Medical Association. All rights reserved.
Figure. Algorithm of Research on Adverse Drug Events and Reports (RADAR) protocol for investigation of adverse events and dissemination of results.3 ADR = adverse drug and device reaction; FDA = US Food and Drug Administration; IRB = institutional review board
Academic pharmacovigilance organizations such as SONAR operate differently from the FDA and pharmaceutical manufacturers in their search for adverse drug events (Figure).4 SONAR collects reports from investigators, clinicians, attorneys, patients, and family members on suspected treatment-related adverse events and investigates these reports carefully. Direct calls to hospitals and large centers can be useful in searching for cases, using information obtained from Internal Review Boards and medical records.

SONAR investigators perform extensive literature reviews, may request more data from authors, and request and review additional FDA case reports. Unfortunately, obtaining data from the FDA can be difficult and time-consuming. Data can be requested through the Freedom of Information Act (FOIA), but receiving it may take more than a year, and the information in the public record may be redacted. SONAR obtains laboratory tests and imaging records and works with scientists to better understand the pathophysiology of potential treatment-related rare adverse events, investigate epidemiologic estimates of the side effect rate, and evaluate risk factors for development of toxicity.

Adverse events are usually identified by SONAR within 2 years post–drug approval—a 5-year improvement over the FDA on this important metric. Once an adverse event is positively identified, the information is disseminated throughout the worldwide medical community via journal articles and presentations at medical conferences. Funding is grant-based from sources such as the National Institutes of Health (NIH), the state of South Carolina, and the University of South Carolina.

FDA, manufacturer reports may be incomplete and delayed

In contrast with SONAR, the FDA relies heavily on MedWatch to detect cases of adverse events. The safety record compiled by MedWatch is often incomplete because the program relies on voluntary submissions of adverse events; further, the inordinate amount of followup required of physicians discourages many from participating. The time to identify an adverse event can be several years, and the FDA disseminates adverse event reports via package inserts. The network that evaluates the safety information and identifies initial safety signals is mainly internal to FDA employees, as is the funding.

Pharmaceutical manufacturers frequently compile data from their own proprietary databases. Although they attempt to follow up on reports of rare adverse events, it is often difficult or impossible for the company to obtain followup information from busy clinicians. Identification of an adverse event typically takes 7 to 12 years for most pharmaceutical manufacturers—reflecting the barriers experienced in obtaining detailed information from clinicians about potential new serious adverse drug reactions. Findings are frequently disseminated through “Dear Doctor” letters. Manufacturers’ investigative networks, like those of the FDA, are largely internal and the amount of funding of they allocate to drug safety investigations is unknown.

RARE EVENTS MAY INVOLVE FEW CASES

Of our major publications,5–14 many findings are based on a small number of cases—for example, only 13 cases for clopidogrel-associated thrombotic thrombocytopenic purpura (TTP)13 and 9 for pure red cell aplasia caused by epoetin alfa.12 Important findings also come from meta-analyses,8,10 although this avenue in our pharmacovigilance approach is less typical.

The 2008 study6 on mortality and venous thromboembolism associated with erythropoiesis-stimulating agents highlights the importance of basic scientific investigation in identifying rare events. Administration of epoetin alfa to raise hemoglobin levels had been approved by the FDA in 1989 for use in patients undergoing dialysis and in 1993 for supportive use in patients with some types of cancers. We discovered that epoetin alfa promoted cancer growth based on analysis of published data and reports in conjunction with basic scientific studies of erythropoietin and erythropoietin receptors in solid cancers.

 

 

RITUXIMAB AND VIRAL REACTIVATION

In the case of viral reactivation associated with the use of rituximab, a warning about hepatitis B reactivation was added to the package insert in 2004.15 In 2006, a warning about other viral infections was added to the package insert.16 In late 2006, a letter was sent to health care professionals from the manufacturer and the FDA with the warning that PML had been observed in two patients with systemic lupus erythematosus (SLE) who were treated with rituximab (an off-label use), both of whom were negative for human immunodeficiency virus (HIV).16 A few months later, a black box warning to this effect was added to the package insert.16

After we identified PML as an adverse event from rituximab in HIV-negative patients,14 we obtained case reports from clinicians at 12 cancer centers or academic hospitals (22 cases). We also reviewed FDA reports (11 cases), the manufacturer’s database (30 cases), and publications (18 cases) using the search terms “leukoencephalopathy,” “rituximab,” “immunosuppressed,” “lymphoma,” and “leukemia.” The unique data sources included clinical observations, the medical literature, FDA MedWatch, and the manufacturer.17

Of rituximab-treated patients who developed PML, the mean age was 61 years (range, 30 to 89 years), 56% of patients were women, and the mean number of rituximab doses was six (range, 1 to 28). Six patients had undergone stem cell transplants (four autologous), and 26 were also taking a purine analogue.17

Among 57 patients, a median of 16 months elapsed between first taking rituximab to development of PML (range, 1.0 to 90.0 months), and 5.5 months from the last dose of rituximab to development of PML (range, 0.3 to 66.0 months). The median time from diagnosis of PML to death was only 2.0 months (range, 0.4 to 122 months). Reported survival rates for patients with rituximab-associated PML who did not undergo stem cell transplantation was less than 10%.17

The symptoms of PML are easily confused with those that might be expected in an older patient with lymphoma, making early detection especially difficult. More than one-half (54.4%) had confusion or disorientation, and many had focal motor weakness (33.3%), loss of coordination (24.6%), difficulty speaking (21.2%), and vision changes (17.5%).

Effects on T and B cells and role of JC virus

At the time of PML diagnosis, 90% of patients had either a severely low CD4+ count or a low CD4+:CD8+ ratio. Based on clinical trial data, cytotoxic chemotherapy and not rituximab appears responsible for the abnormal CD4+ count and the low CD4+:CD8+ ratio in rituximab-treated patients.

Little is known about how T cells function after rituximab administration. In idiopathic thrombocytopenic purpura, the response to B-cell depletion induced by rituximab is associated with significant changes in the T-cell compartment.18 In a study of patients with either SLE or Evans syndrome, rituximab therapy was found to modify T-cell phenotype and cytokine profiles.19 The rapid effect of rituximab in multiple sclerosis suggests that it targets a process thought to be T-cell mediated.20

Our early hypothesis was that rituximab contributes to viral reactivation and PML through inhibiting T-and B-lymphocyte interactions. We now believe that the bone marrow plays an important role, which may explain the process by which natalizumab can cause PML. Five of five bone marrow samples from patients with lymphoma and PML tested positive for JC virus (JCV) compared with only two of 86 bone marrow samples from patients without PML. The JCV is latent in CD34+ hematopoietic cells and probably in early B lymphocytes. Chemotherapy mobilizes the stem cells from bone marrow and causes quantitative T-cell depletion. Rituximab reduces the qualitative T-cell response, and B-cell depletion results in expansion of progenitor cells containing the latent JCV. The hypothesis is limited in that it is based on a retrospective case series and is not verified in a laboratory model.

Of the 57 cases of PML identified in 2009, two patients were given rituximab for hematologic disorders and had no chemotherapy other than steroids. These data suggest that rituximab confers risk on its own.17

Quantifying risk of developing PML from rituximab

Calculating the odds of developing PML from rituximab therapy is difficult. The background rate of PML is an important consideration. One population-based study estimated the incidence of PML in patients with hematologic malignancies at 0.07%. This estimate was based on three cases of PML observed in patients with hematologic malignancies over a period of 11 years in a single Canadian province.21 Another study found a higher incidence of 0.52% in patients with chronic lymphocytic leukemia, although all of these patients were also treated with fludarabine.22 Accurately calculating the risk of PML attributable to the underlying malignancy as opposed to immune suppression from treatment is complicated by the rarity of the disease. Fludarabine is the chemotherapeutic agent most closely associated with PML. However, its well known side effects of T-lymphocyte depletion and complicating opportunistic infections similar to those seen in acquired immunodeficiency syndrome (AIDS) make such an association intuitive.23

Kavenaugh and Matteson reported that about 8,000 SLE patients had received rituximab treatment and two of these patients had developed PML.24 PML has been reported previously among 30 SLE patients who had not received rituximab, suggesting that SLE is a predisposing disorder.25,26

In the setting of hematologic malignancy, rituximab-associated PML incidence estimates are complicated by a low basal risk of PML seen among persons with the disease state prompting rituximab therapy and an inability to determine risk attributable to rituximab. A recent study demonstrated an association between rituximab and PML in patients with non-Hodgkin lymphoma (NHL). The retrospective, monocentric cohort study assessed data from 976 NHL patients diagnosed in Italy from 1994 to 2008, including 517 patients who received at least one dose of rituximab. Inclusion of rituximab into standard chemotherapy regimens for NHL caused a significantly higher incidence of PML cases (rate difference, 2.2 every 1,000 patient-years; 95% confidence interval, 0.1–4.3).27 More such studies of viral reactivation syndromes are obviously needed.

Ideally, randomized clinical trials of the use of rituximab in patients with lymphoma would serve as guidance, but because the drug, as the standard of care for treatment of lymphoma, is so widely used, randomization would be impractical.

Future planned studies include a case-control study of T-cell markers after chemotherapy administration with or without exposure to rituximab, a case-control study of bone marrow specimens from disease-matched and treatment-matched controls, and a cohort study using a large electronic medical records database or a government database.

CONCLUSION

The methods developed in the SONAR project will permit exploration of important hypotheses regarding the detection and prevention of rare adverse events in oncology, forming a basis for subsequent investigations. Based on our recent findings, rituximab may be associated with multiple viral reactivation syndromes; screening and early detection can potentially be helpful in preventing these complications.

DISCUSSION

Dr. Calabrese: Your approach to identifying rare adverse events is novel and aggressive, but the seeming limitations in a disease such as lymphoma are (1) rituximab is now a standard of care so everybody with lymphoma gets it, and (2) going back to the earliest descriptions of PML, lymphoma has always been represented as a predisposing factor. Moving ahead, how then can you calculate an effect size for a drug like rituximab?

Dr. Bennett: There’s no way to do it; we’re sort of stuck. Of the 57 cases with PML that we reported in Blood,17 two patients received rituximab for hematologic disorders and received no chemotherapy besides steroids. In those two patients, we could not blame the development of PML on lymphoma. Those types of patients suggest that rituximab may be implicated, but examining this question with a case-control or even a cohort study is an expensive proposition.

Dr. Simpson: My experience in terms of collaborating with the FDA has been distinctly unrewarding. Some years ago I had been looking into an adverse effect related to the nucleoside analog reverse transcriptase inhibitor d4T, in which there was a rapidly progressive neuromuscular weakness syndrome that looked like Guillain-Barré and lactic acidosis. The FDA itself reported 12 cases at an international AIDS conference and did not have any answers. I was charged by the AIDS Clinical Trials Group and other branches at the NIH to try to figure it out. When I requested access to FDA data, I ran into an unbelievable bureaucratic morass. Ultimately, we had to go through the FOIA to get them to release anything.

Dr. Bennett: The FOIA is the only way to get anything from the FDA. It takes about a year and a half and much information is redacted.

Dr. Berger: As we roll out these newer compounds, we need a mechanism to look for both foreseen and unforeseen consequences, perhaps with close collaboration between pharmaceutical companies and governmental agencies.

Dr. Bennett: The Risk Evaluation and Mitigation Strategies program authorizes the FDA to require post-marketing surveillance of all adverse events from manufacturers. We published 11 cases of TTP in association with clopidogrel, obtained from surveillance of directors of plasmapheresis centers in the United States. Not one of them had been reported to the FDA directly. However, we had an article 6 weeks after clopidogrel received FDA approval. Now, 10 years later, there are about 120 clopidogrel-associated TTP cases in the FDA database. Its estimated incidence is still one in a million, although we hear about the side effect every night on TV during commercials for the drug on the evening news.

Dr. Major: The FDA is more open now than in the past to trying to get a handle on what’s going on with biologic therapies. We need to do a little more homework up front on biologic agents in order to anticipate some adverse events. For example, the migratory nature of CD34+ cells through the circulation following natalizumab therapy was not appreciated, even though data in the literature already supported this phenomenon when integrin receptors are blocked.

Rare and serious drug-related events are often not detected until after clinical trials have been completed and a drug becomes widely used. Methods traditionally used by pharmaceutical companies and the US Food and Drug Administration (FDA) are not the most effective ways to promptly identify a treatment-related adverse event and quickly notify the medical community. In 1998, an academically based surveillance group was created to identify and disseminate information on unrecognized adverse drug reactions.1 In 2010, with funding from the state of South Carolina, this program became the Southern Network on Adverse Reactions (SONAR), the only state-funded pharmacovigilance initiative in the nation. SONAR and its earlier incarnation have identified potentially fatal and previously unreported side effects associated with 43 drugs—with the majority of these drugs involving the hematology and oncology discisplines.

While the earlier incarnation of drug safety monitoring relied on data mining, or detecting specific signals from large amounts of data, investigations of possible adverse drug event occurrences have a much broader scope. SONAR, an enhanced surveillance program, was created to address this issue. Based jointly at the South Carolina College of Pharmacy and a National Cancer Institute–designated cancer center, the Hollings Cancer Center at the Medical University of South Carolina, SONAR more accurately reflects the nature of our adverse effects investigations: identification of small numbers of important cases from a variety of unique data sources, including case reports, the medical literature, FDA MedWatch, and pharmaceutical manufacturers.

This article reviews methods that underlie the successful investigations of the SONAR initiative, and it examines our SONAR investigation of the association between the immune modulatory monoclonal antibody rituximab and progressive multifocal leukoencephalopathy (PML).

DETECTING, INVESTIGATING, AND DISSEMINATING FINDINGS

Surveillance programs are needed because important rare side effects are seldom discovered in a clinical trial. The Safety and Efficacy of Natalizumab in Combination with Interferon Beta-1a in Patients with Relapsing Remitting Multiple Sclerosis (SENTINEL) trial was unusual in that it detected two cases of PML associated with the use of natalizumab.2 Most rare side effects are undetected at the time of FDA approval, and usually many years elapse from the time a potential problem is detected until it is identified as a rare side effect of the drug. The average time for a “black box” warning to appear on a package insert following FDA approval is 7 to 10 years.3

Timely and thorough data collection

Reprinted, with permission, from The Journal of the American Medical Association (Bennett CL, et al. JAMA 2005; 29:211–2140), Copyright © 2005 American Medical Association. All rights reserved.
Figure. Algorithm of Research on Adverse Drug Events and Reports (RADAR) protocol for investigation of adverse events and dissemination of results.3 ADR = adverse drug and device reaction; FDA = US Food and Drug Administration; IRB = institutional review board
Academic pharmacovigilance organizations such as SONAR operate differently from the FDA and pharmaceutical manufacturers in their search for adverse drug events (Figure).4 SONAR collects reports from investigators, clinicians, attorneys, patients, and family members on suspected treatment-related adverse events and investigates these reports carefully. Direct calls to hospitals and large centers can be useful in searching for cases, using information obtained from Internal Review Boards and medical records.

SONAR investigators perform extensive literature reviews, may request more data from authors, and request and review additional FDA case reports. Unfortunately, obtaining data from the FDA can be difficult and time-consuming. Data can be requested through the Freedom of Information Act (FOIA), but receiving it may take more than a year, and the information in the public record may be redacted. SONAR obtains laboratory tests and imaging records and works with scientists to better understand the pathophysiology of potential treatment-related rare adverse events, investigate epidemiologic estimates of the side effect rate, and evaluate risk factors for development of toxicity.

Adverse events are usually identified by SONAR within 2 years post–drug approval—a 5-year improvement over the FDA on this important metric. Once an adverse event is positively identified, the information is disseminated throughout the worldwide medical community via journal articles and presentations at medical conferences. Funding is grant-based from sources such as the National Institutes of Health (NIH), the state of South Carolina, and the University of South Carolina.

FDA, manufacturer reports may be incomplete and delayed

In contrast with SONAR, the FDA relies heavily on MedWatch to detect cases of adverse events. The safety record compiled by MedWatch is often incomplete because the program relies on voluntary submissions of adverse events; further, the inordinate amount of followup required of physicians discourages many from participating. The time to identify an adverse event can be several years, and the FDA disseminates adverse event reports via package inserts. The network that evaluates the safety information and identifies initial safety signals is mainly internal to FDA employees, as is the funding.

Pharmaceutical manufacturers frequently compile data from their own proprietary databases. Although they attempt to follow up on reports of rare adverse events, it is often difficult or impossible for the company to obtain followup information from busy clinicians. Identification of an adverse event typically takes 7 to 12 years for most pharmaceutical manufacturers—reflecting the barriers experienced in obtaining detailed information from clinicians about potential new serious adverse drug reactions. Findings are frequently disseminated through “Dear Doctor” letters. Manufacturers’ investigative networks, like those of the FDA, are largely internal and the amount of funding of they allocate to drug safety investigations is unknown.

RARE EVENTS MAY INVOLVE FEW CASES

Of our major publications,5–14 many findings are based on a small number of cases—for example, only 13 cases for clopidogrel-associated thrombotic thrombocytopenic purpura (TTP)13 and 9 for pure red cell aplasia caused by epoetin alfa.12 Important findings also come from meta-analyses,8,10 although this avenue in our pharmacovigilance approach is less typical.

The 2008 study6 on mortality and venous thromboembolism associated with erythropoiesis-stimulating agents highlights the importance of basic scientific investigation in identifying rare events. Administration of epoetin alfa to raise hemoglobin levels had been approved by the FDA in 1989 for use in patients undergoing dialysis and in 1993 for supportive use in patients with some types of cancers. We discovered that epoetin alfa promoted cancer growth based on analysis of published data and reports in conjunction with basic scientific studies of erythropoietin and erythropoietin receptors in solid cancers.

 

 

RITUXIMAB AND VIRAL REACTIVATION

In the case of viral reactivation associated with the use of rituximab, a warning about hepatitis B reactivation was added to the package insert in 2004.15 In 2006, a warning about other viral infections was added to the package insert.16 In late 2006, a letter was sent to health care professionals from the manufacturer and the FDA with the warning that PML had been observed in two patients with systemic lupus erythematosus (SLE) who were treated with rituximab (an off-label use), both of whom were negative for human immunodeficiency virus (HIV).16 A few months later, a black box warning to this effect was added to the package insert.16

After we identified PML as an adverse event from rituximab in HIV-negative patients,14 we obtained case reports from clinicians at 12 cancer centers or academic hospitals (22 cases). We also reviewed FDA reports (11 cases), the manufacturer’s database (30 cases), and publications (18 cases) using the search terms “leukoencephalopathy,” “rituximab,” “immunosuppressed,” “lymphoma,” and “leukemia.” The unique data sources included clinical observations, the medical literature, FDA MedWatch, and the manufacturer.17

Of rituximab-treated patients who developed PML, the mean age was 61 years (range, 30 to 89 years), 56% of patients were women, and the mean number of rituximab doses was six (range, 1 to 28). Six patients had undergone stem cell transplants (four autologous), and 26 were also taking a purine analogue.17

Among 57 patients, a median of 16 months elapsed between first taking rituximab to development of PML (range, 1.0 to 90.0 months), and 5.5 months from the last dose of rituximab to development of PML (range, 0.3 to 66.0 months). The median time from diagnosis of PML to death was only 2.0 months (range, 0.4 to 122 months). Reported survival rates for patients with rituximab-associated PML who did not undergo stem cell transplantation was less than 10%.17

The symptoms of PML are easily confused with those that might be expected in an older patient with lymphoma, making early detection especially difficult. More than one-half (54.4%) had confusion or disorientation, and many had focal motor weakness (33.3%), loss of coordination (24.6%), difficulty speaking (21.2%), and vision changes (17.5%).

Effects on T and B cells and role of JC virus

At the time of PML diagnosis, 90% of patients had either a severely low CD4+ count or a low CD4+:CD8+ ratio. Based on clinical trial data, cytotoxic chemotherapy and not rituximab appears responsible for the abnormal CD4+ count and the low CD4+:CD8+ ratio in rituximab-treated patients.

Little is known about how T cells function after rituximab administration. In idiopathic thrombocytopenic purpura, the response to B-cell depletion induced by rituximab is associated with significant changes in the T-cell compartment.18 In a study of patients with either SLE or Evans syndrome, rituximab therapy was found to modify T-cell phenotype and cytokine profiles.19 The rapid effect of rituximab in multiple sclerosis suggests that it targets a process thought to be T-cell mediated.20

Our early hypothesis was that rituximab contributes to viral reactivation and PML through inhibiting T-and B-lymphocyte interactions. We now believe that the bone marrow plays an important role, which may explain the process by which natalizumab can cause PML. Five of five bone marrow samples from patients with lymphoma and PML tested positive for JC virus (JCV) compared with only two of 86 bone marrow samples from patients without PML. The JCV is latent in CD34+ hematopoietic cells and probably in early B lymphocytes. Chemotherapy mobilizes the stem cells from bone marrow and causes quantitative T-cell depletion. Rituximab reduces the qualitative T-cell response, and B-cell depletion results in expansion of progenitor cells containing the latent JCV. The hypothesis is limited in that it is based on a retrospective case series and is not verified in a laboratory model.

Of the 57 cases of PML identified in 2009, two patients were given rituximab for hematologic disorders and had no chemotherapy other than steroids. These data suggest that rituximab confers risk on its own.17

Quantifying risk of developing PML from rituximab

Calculating the odds of developing PML from rituximab therapy is difficult. The background rate of PML is an important consideration. One population-based study estimated the incidence of PML in patients with hematologic malignancies at 0.07%. This estimate was based on three cases of PML observed in patients with hematologic malignancies over a period of 11 years in a single Canadian province.21 Another study found a higher incidence of 0.52% in patients with chronic lymphocytic leukemia, although all of these patients were also treated with fludarabine.22 Accurately calculating the risk of PML attributable to the underlying malignancy as opposed to immune suppression from treatment is complicated by the rarity of the disease. Fludarabine is the chemotherapeutic agent most closely associated with PML. However, its well known side effects of T-lymphocyte depletion and complicating opportunistic infections similar to those seen in acquired immunodeficiency syndrome (AIDS) make such an association intuitive.23

Kavenaugh and Matteson reported that about 8,000 SLE patients had received rituximab treatment and two of these patients had developed PML.24 PML has been reported previously among 30 SLE patients who had not received rituximab, suggesting that SLE is a predisposing disorder.25,26

In the setting of hematologic malignancy, rituximab-associated PML incidence estimates are complicated by a low basal risk of PML seen among persons with the disease state prompting rituximab therapy and an inability to determine risk attributable to rituximab. A recent study demonstrated an association between rituximab and PML in patients with non-Hodgkin lymphoma (NHL). The retrospective, monocentric cohort study assessed data from 976 NHL patients diagnosed in Italy from 1994 to 2008, including 517 patients who received at least one dose of rituximab. Inclusion of rituximab into standard chemotherapy regimens for NHL caused a significantly higher incidence of PML cases (rate difference, 2.2 every 1,000 patient-years; 95% confidence interval, 0.1–4.3).27 More such studies of viral reactivation syndromes are obviously needed.

Ideally, randomized clinical trials of the use of rituximab in patients with lymphoma would serve as guidance, but because the drug, as the standard of care for treatment of lymphoma, is so widely used, randomization would be impractical.

Future planned studies include a case-control study of T-cell markers after chemotherapy administration with or without exposure to rituximab, a case-control study of bone marrow specimens from disease-matched and treatment-matched controls, and a cohort study using a large electronic medical records database or a government database.

CONCLUSION

The methods developed in the SONAR project will permit exploration of important hypotheses regarding the detection and prevention of rare adverse events in oncology, forming a basis for subsequent investigations. Based on our recent findings, rituximab may be associated with multiple viral reactivation syndromes; screening and early detection can potentially be helpful in preventing these complications.

DISCUSSION

Dr. Calabrese: Your approach to identifying rare adverse events is novel and aggressive, but the seeming limitations in a disease such as lymphoma are (1) rituximab is now a standard of care so everybody with lymphoma gets it, and (2) going back to the earliest descriptions of PML, lymphoma has always been represented as a predisposing factor. Moving ahead, how then can you calculate an effect size for a drug like rituximab?

Dr. Bennett: There’s no way to do it; we’re sort of stuck. Of the 57 cases with PML that we reported in Blood,17 two patients received rituximab for hematologic disorders and received no chemotherapy besides steroids. In those two patients, we could not blame the development of PML on lymphoma. Those types of patients suggest that rituximab may be implicated, but examining this question with a case-control or even a cohort study is an expensive proposition.

Dr. Simpson: My experience in terms of collaborating with the FDA has been distinctly unrewarding. Some years ago I had been looking into an adverse effect related to the nucleoside analog reverse transcriptase inhibitor d4T, in which there was a rapidly progressive neuromuscular weakness syndrome that looked like Guillain-Barré and lactic acidosis. The FDA itself reported 12 cases at an international AIDS conference and did not have any answers. I was charged by the AIDS Clinical Trials Group and other branches at the NIH to try to figure it out. When I requested access to FDA data, I ran into an unbelievable bureaucratic morass. Ultimately, we had to go through the FOIA to get them to release anything.

Dr. Bennett: The FOIA is the only way to get anything from the FDA. It takes about a year and a half and much information is redacted.

Dr. Berger: As we roll out these newer compounds, we need a mechanism to look for both foreseen and unforeseen consequences, perhaps with close collaboration between pharmaceutical companies and governmental agencies.

Dr. Bennett: The Risk Evaluation and Mitigation Strategies program authorizes the FDA to require post-marketing surveillance of all adverse events from manufacturers. We published 11 cases of TTP in association with clopidogrel, obtained from surveillance of directors of plasmapheresis centers in the United States. Not one of them had been reported to the FDA directly. However, we had an article 6 weeks after clopidogrel received FDA approval. Now, 10 years later, there are about 120 clopidogrel-associated TTP cases in the FDA database. Its estimated incidence is still one in a million, although we hear about the side effect every night on TV during commercials for the drug on the evening news.

Dr. Major: The FDA is more open now than in the past to trying to get a handle on what’s going on with biologic therapies. We need to do a little more homework up front on biologic agents in order to anticipate some adverse events. For example, the migratory nature of CD34+ cells through the circulation following natalizumab therapy was not appreciated, even though data in the literature already supported this phenomenon when integrin receptors are blocked.

References
  1. Hampton T. Postmarket “pharmacovigilance” program on alert for adverse events from drugs. JAMA 2007; 298:851852.
  2. Rudick RA, Stuart WH, Calabresi PA, et al. Natalizumab plus interferon beta-1a for relapsing multiple sclerosis. N Engl J Med 2006; 354:911923.
  3. Lasser KE, Allen PD, Woolhandler SJ, Himmelstein Du, Wolfe SM, Bor DH. Timing of new black box warnings and withdrawals for prescription medications. JAMA 2002; 287:22152220.
  4. Bennett CL, Nebeker JR, Lyons EA, et al. The Research on Adverse Drug Events and Reports (RADAR) project. JAMA 2005; 293:21312140.
  5. Edwards BJ, Gounder M, McKoy JM, et al. Pharmacovigilance and reporting oversight in US FDA fast-track process: bisphosphonates and osteonecrosis of the jaw. Lancet Oncol 2008; 9:11661172.
  6. Bennett CL, Silver SM, Djulbegovic B, et al. Venous thromboembolism and mortality associated with recombinant erythropoietin and darbepoetin administration for the treatment of cancer-associated anemia. JAMA 2008; 299:914924.
  7. Beohar N, Davidson CJ, Kip KE, et al. Outcomes and complications associated with off-label and untested use of drug-eluting stents. JAMA 2007; 297:19922000.
  8. Hershman D, Neugut AI, Jacobson JS, et al. Acute myeloid leukemia or myelodysplastic syndrome following use of granulocyte colony-stimulating factors during breast cancer adjuvant chemotherapy. J Natl Cancer Inst 2007; 99:196205.
  9. Bohlius J, Wilson J, Seidenfeld J, et al. Recombinant human erythropoietins and cancer patients: updated meta-analysis of 57 studies including 9353 patients. J Natl Cancer Inst 2006; 98:708714.
  10. Nebeker JR, Virmani R, Bennett CL, et al. Hypersensitivity cases associated with drug-eluting coronary stents: a review of available cases from the Research on Adverse Drug Events and Reports (RADAR) project. J Am Coll Cardiol 2006; 47:175181.
  11. Bennett CL, Angelotta C, Yarnold PR, et al. Thalidomide- and lenalidomide-associated thromboembolism among patients with cancer. JAMA 2006; 296:25582560.
  12. Bennett CL, Luminari S, Nissenson AR, et al. Pure red-cell aplasia and epoetin therapy. N Engl J Med 2004; 351:14031408.
  13. Bennett CL, Connors JM, Carwile JM, et al. Thrombotic thrombocytopenic purpura associated with clopidogrel. N Engl J Med 2000; 342:17731777.
  14. Bennett CL, Kiss JE, Weinberg PD, et al. Thrombotic thrombocytopenic purpura after stenting and ticlopidine. Lancet 1998; 352:10361037.
  15. Rituxan (rituximab) Oct 2004. U.S. Food and Drug Administration Web site. http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsforHumanMedicalProducts/ucm166521.htm. Updated June 19, 2009. Accessed September 13, 2011.
  16. Rituxan (rituximab). U.S. Food and Drug Administration Web site. http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsforHumanMedicalProducts/ucm150747.htm. Updated June 19, 2009. Accessed September 13, 2011.
  17. Carson KR, Evens AM, Richey EA, et al. Progressive multifocal leukoencephalopathy after rituximab therapy in HIV-negative patients: a report of 57 cases from the Research on Adverse Drug Events and Reports project. Blood 2009; 113:48344840.
  18. Stasi R, Del Poeta G, Stipa E, et al. Response to B-cell depleting therapy with rituximab reverts the abnormalities of T-cell subsets in patients with idiopathic thrombocytopenic purpura. Blood 2007; 110:29242930.
  19. Tamimoto Y, Horiuchi T, Tsukamoto H, et al. A dose-escalation study of rituximab for treatment of systemic lupus erythematosus and Evans’ syndrome: immunological analysis of B cells, T cells and cytokines. Rheumatology (Oxford) 2008; 47:821827.
  20. McFarland HF. The B cell—old player, new position on the team. N Engl J Med 2008; 358:664665.
  21. Power C, Gladden JG, Halliday W, et al. AIDS- and non-AIDS-related PML association with distinct p53 polymorphism. Neurology 2000; 54:743746.
  22. Gonzalez H, Bolgert F, Camporo P, Leblond V. Progressive multi focal leukoencephalitis (PML) in three patients treated with standard-dose fludarabine (FAMP). Hematol Cell Ther 1999; 41:183186.
  23. Garcia-Suárez J, de Miguel D, Krsnik I, Bañas H, Arribas I, Burgaleta C. Changes in the natural history of progressive multifocal leukoencephalopathy in HIV-negative lymphoproliferative disorders: impact of novel therapies. Am J Hematol 2005; 80:271281.
  24. Kavanaugh A, Matteson E. Hotline: rituximab and progressive multifocal leukoencephalopathy. American College of Rheumatology Web site. http://www.rheumatology.org/publications/hotline/0107leuko.asp. Updated January 2007. Accessed September 13, 2011.
  25. Molloy ES, Calabrese LH. Progressive multifocal leukoencephalopathy in patients with rheumatic diseases: are patients with systemic lupus erythematosus at particular risk? Autoimmun Rev 2008; 8:144146.
  26. Calabrese LH, Molloy ES. Progressive multifocal leucoencephalopathy in the rheumatic diseases: assessing the risks of biological immunosuppressive therapies. Ann Rheum Dis 2008; 67 (suppl 3):iii64iii65.
  27. Tuccori M, Focosi D, Blandizzi C, et al. Inclusion of rituximab in treatment protocols for non-Hodgkin’s lymphomas and risk for progressive multifocal leukoencephalopathy. Oncologist 2010; 15:12141219. Epub 2010 Nov 1.
References
  1. Hampton T. Postmarket “pharmacovigilance” program on alert for adverse events from drugs. JAMA 2007; 298:851852.
  2. Rudick RA, Stuart WH, Calabresi PA, et al. Natalizumab plus interferon beta-1a for relapsing multiple sclerosis. N Engl J Med 2006; 354:911923.
  3. Lasser KE, Allen PD, Woolhandler SJ, Himmelstein Du, Wolfe SM, Bor DH. Timing of new black box warnings and withdrawals for prescription medications. JAMA 2002; 287:22152220.
  4. Bennett CL, Nebeker JR, Lyons EA, et al. The Research on Adverse Drug Events and Reports (RADAR) project. JAMA 2005; 293:21312140.
  5. Edwards BJ, Gounder M, McKoy JM, et al. Pharmacovigilance and reporting oversight in US FDA fast-track process: bisphosphonates and osteonecrosis of the jaw. Lancet Oncol 2008; 9:11661172.
  6. Bennett CL, Silver SM, Djulbegovic B, et al. Venous thromboembolism and mortality associated with recombinant erythropoietin and darbepoetin administration for the treatment of cancer-associated anemia. JAMA 2008; 299:914924.
  7. Beohar N, Davidson CJ, Kip KE, et al. Outcomes and complications associated with off-label and untested use of drug-eluting stents. JAMA 2007; 297:19922000.
  8. Hershman D, Neugut AI, Jacobson JS, et al. Acute myeloid leukemia or myelodysplastic syndrome following use of granulocyte colony-stimulating factors during breast cancer adjuvant chemotherapy. J Natl Cancer Inst 2007; 99:196205.
  9. Bohlius J, Wilson J, Seidenfeld J, et al. Recombinant human erythropoietins and cancer patients: updated meta-analysis of 57 studies including 9353 patients. J Natl Cancer Inst 2006; 98:708714.
  10. Nebeker JR, Virmani R, Bennett CL, et al. Hypersensitivity cases associated with drug-eluting coronary stents: a review of available cases from the Research on Adverse Drug Events and Reports (RADAR) project. J Am Coll Cardiol 2006; 47:175181.
  11. Bennett CL, Angelotta C, Yarnold PR, et al. Thalidomide- and lenalidomide-associated thromboembolism among patients with cancer. JAMA 2006; 296:25582560.
  12. Bennett CL, Luminari S, Nissenson AR, et al. Pure red-cell aplasia and epoetin therapy. N Engl J Med 2004; 351:14031408.
  13. Bennett CL, Connors JM, Carwile JM, et al. Thrombotic thrombocytopenic purpura associated with clopidogrel. N Engl J Med 2000; 342:17731777.
  14. Bennett CL, Kiss JE, Weinberg PD, et al. Thrombotic thrombocytopenic purpura after stenting and ticlopidine. Lancet 1998; 352:10361037.
  15. Rituxan (rituximab) Oct 2004. U.S. Food and Drug Administration Web site. http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsforHumanMedicalProducts/ucm166521.htm. Updated June 19, 2009. Accessed September 13, 2011.
  16. Rituxan (rituximab). U.S. Food and Drug Administration Web site. http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsforHumanMedicalProducts/ucm150747.htm. Updated June 19, 2009. Accessed September 13, 2011.
  17. Carson KR, Evens AM, Richey EA, et al. Progressive multifocal leukoencephalopathy after rituximab therapy in HIV-negative patients: a report of 57 cases from the Research on Adverse Drug Events and Reports project. Blood 2009; 113:48344840.
  18. Stasi R, Del Poeta G, Stipa E, et al. Response to B-cell depleting therapy with rituximab reverts the abnormalities of T-cell subsets in patients with idiopathic thrombocytopenic purpura. Blood 2007; 110:29242930.
  19. Tamimoto Y, Horiuchi T, Tsukamoto H, et al. A dose-escalation study of rituximab for treatment of systemic lupus erythematosus and Evans’ syndrome: immunological analysis of B cells, T cells and cytokines. Rheumatology (Oxford) 2008; 47:821827.
  20. McFarland HF. The B cell—old player, new position on the team. N Engl J Med 2008; 358:664665.
  21. Power C, Gladden JG, Halliday W, et al. AIDS- and non-AIDS-related PML association with distinct p53 polymorphism. Neurology 2000; 54:743746.
  22. Gonzalez H, Bolgert F, Camporo P, Leblond V. Progressive multi focal leukoencephalitis (PML) in three patients treated with standard-dose fludarabine (FAMP). Hematol Cell Ther 1999; 41:183186.
  23. Garcia-Suárez J, de Miguel D, Krsnik I, Bañas H, Arribas I, Burgaleta C. Changes in the natural history of progressive multifocal leukoencephalopathy in HIV-negative lymphoproliferative disorders: impact of novel therapies. Am J Hematol 2005; 80:271281.
  24. Kavanaugh A, Matteson E. Hotline: rituximab and progressive multifocal leukoencephalopathy. American College of Rheumatology Web site. http://www.rheumatology.org/publications/hotline/0107leuko.asp. Updated January 2007. Accessed September 13, 2011.
  25. Molloy ES, Calabrese LH. Progressive multifocal leukoencephalopathy in patients with rheumatic diseases: are patients with systemic lupus erythematosus at particular risk? Autoimmun Rev 2008; 8:144146.
  26. Calabrese LH, Molloy ES. Progressive multifocal leucoencephalopathy in the rheumatic diseases: assessing the risks of biological immunosuppressive therapies. Ann Rheum Dis 2008; 67 (suppl 3):iii64iii65.
  27. Tuccori M, Focosi D, Blandizzi C, et al. Inclusion of rituximab in treatment protocols for non-Hodgkin’s lymphomas and risk for progressive multifocal leukoencephalopathy. Oncologist 2010; 15:12141219. Epub 2010 Nov 1.
Page Number
S13-S17
Page Number
S13-S17
Publications
Publications
Article Type
Display Headline
Pharmacovigilance and PML in the oncology setting
Display Headline
Pharmacovigilance and PML in the oncology setting
Citation Override
Cleveland Clinic Journal of Medicine 2011 November;78(suppl 2):S13-S17
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media

Multiple sclerosis, natalizumab, and PML: Helping patients decide

Article Type
Changed
Tue, 10/02/2018 - 09:24
Display Headline
Multiple sclerosis, natalizumab, and PML: Helping patients decide

Multiple sclerosis (MS) is an autoimmune disease whose inflammatory process causes demyelination, axonal loss, and neurodegeneration, all of which can lead to progressive neurologic disability. Without treatment, the risk of progressive disability 15 to 20 years after the onset of MS has been estimated to be as high as 50%.1

Seven drugs have been approved by the US Food and Drug Administration (FDA) for the treatment of MS. Of these, interferon beta drugs and glatiramer acetate are generally considered as first-line agents based on extensive experience and relative safety. Indeed, reports with followup approaching 20 years have not identified significant safety concerns. However, these agents have only modest efficacy, reducing by approximately 30% the frequency of relapse in patients with relapsing-remitting MS.2–6

Natalizumab, and more recently, fingolimod, are generally used as second-line agents. Fingolimod, the first oral agent to receive FDA approval for the treatment of relapsing-remitting MS, is a functional antagonist of sphingosine-1-phosphate receptors. The reductions in annualized relapse rates in two phase 3 controlled trials of fingolimod were approximately 55% compared with placebo7 or intramuscular interferon beta-1a.8 Because of its more convenient oral route of administration and its documented efficacy, widespread use of fingolimod is anticipated. However, adverse reactions affecting more than 10% of patients include headache, influenza, diarrhea, back pain, liver transaminase elevations, and cough.9 Because sphingosine-1-phosphate receptors are widespread in many body tissues, off-target effects of fingolimod may be problematic and long-term toxicity is unknown.

In addition to natalizumab and fingolimod, which are currently available for use as second-line agents, several other MS therapies are showing promise. Oral cladribine, teriflunomide, and laquinimod have reported positive phase 3 results in publication or at national meetings, and several other drugs are in late stages of development (alemtuzumab, BG-12, ocrelizumab) based on encouraging phase 2 results.10–12 Thus, the options for MS patients are expanding, but drugs with higher efficacy also may pose greater risk.

NATALIZUMAB: ROBUST BENEFITS BUT ASSOCIATED RISK

Natalizumab is a humanized monoclonal antibody that binds to alpha-4 integrin on leukocytes. By inhibiting alpha-4 integrin, natalizumab, the first of a new class of selective adhesion-molecule inhibitors, impedes migration of activated mononuclear leukocytes into the brain and gut.13

Significant efficacy

Two phase 3 studies demonstrated more robust efficacy of natalizumab in patients with relapsing-remitting MS than had been observed in prior studies with other agents.14,15 In the Natalizumab Safety and Efficacy in Relapsing-Remitting Multiple Sclerosis (AFFIRM) study, which followed patients over 2 years of treatment, natalizumab was associated with a 68% reduction in the annualized relapse rate14; a 92% reduction in gadolinium-enhanced lesions on magnetic resonance imaging (MRI), which indicate new, active lesions16; and an 83% reduction in the mean number of new or enlarging T2 lesions16 compared with placebo. The likelihood of confirmed worsening on the Kurtzke Expanded Disability Status Score, which is the standard measure for MS-related disability, was also 42% lower in patients assigned to natalizumab compared with placebo.14

Other reported benefits from natalizumab therapy include a significantly increased probability of maintaining disease-free status17 and clinically significant improvements on patient-reported quality-of-life measures.18 Although there have been no head-to-head studies of natalizumab with interferon beta, glatiramer acetate, or fingolimod, there is a widespread view that treatment benefits of natalizumab exceed those of other disease-modifying drugs. In clinical practice, patients with MS who experience breakthrough disease on standard disease-modifying drugs are routinely observed to achieve disease control after switching to natalizumab. Thus, based purely on efficacy, patient-reported outcomes, and the convenience of once-monthly intravenous infusion, natalizumab represents an extremely attractive treatment option for patients with relapsing-remitting MS.

Use discontinued in 2005

Natalizumab was approved for treatment of relapsing-remitting MS in November 2004, using the FDA accelerated review pathway. The approval was based on the first-year results of the AFFIRM14 and the Natalizumab plus Interferon Beta-1a for Relapsing Remitting Multiple Sclerosis (SENTINEL)19 studies, both of which were completed in February 2005. In the 3 to 4 months between the drug’s approval and completion of the AFFIRM and SENTINEL studies, approximately 7,000 patients with relapsing-remitting MS received treatment with natalizumab. In February 2005, shortly after the release of the 2-year data, three cases of progressive multifocal leukoencephalopathy (PML) were identified in natalizumab-treated patients (one with Crohn disease, the other two with MS). Clinical and research use of natalizumab was abruptly suspended that month, pending a comprehensive safety review.

A safety study evaluated 3,116 patients who had received natalizumab over a mean exposure of 17.9 monthly doses.20 The study failed to identify any additional cases of PML and concluded that the risk of PML was approximately 1 in 1,000 patients. Abrupt discontinuation of natalizumab also allowed systematic assessment of disease behavior following treatment interruption. In 1,866 patients who had received natalizumab during clinical trials but who discontinued natalizumab after PML was recognized, MS relapses and gadolinium-enhancing lesions returned approximately to baseline levels within 4 to 7 months of natalizumab suspension. Reactivation of MS disease activity was observed even in patients who instituted one of the first-line disease-modifying drugs as substitute therapy.21

Based on the strong efficacy data and the extensive safety review, an FDA advisory committee recommended reintroduction, and natalizumab was returned to the market in June 2006. Natalizumab may be administered only in accredited infusion centers that agree to a monthly reporting regimen designed to identify all cases of PML. In the United States, natalizumab is available to patients only through the Tysabri Outreach Unified Commitment to Health (TOUCH) Prescribing Program, a restricted distribution program. Aggressive monitoring and reporting is also required in other regions of the world, so that ascertainment of PML associated with natalizumab is thought to be relatively complete.

 

 

Risk related to duration of therapy

Source: Data on file. Biogen Idec; 2011.
Figure 1. Progressive multifocal leukoencephalopathy (PML) incidence estimates by treatment duration (A) and treatment epoch (B), calculated based on natalizumab exposure through January 31, 2011, and 95 confirmed cases as of February 2, 2011. (A) The incidence for each time period is calculated as the number of PML cases divided by the number of patients exposed to natalizumab (eg, for ≥ 24 infusions, the number of PML cases diagnosed with exposure of 24 infusions or more is divided by the total number of patients exposed to at least 24 infusions). (B) The incidence for each epoch is calculated as the number of PML cases divided by the number of patients exposed to natalizumab (eg, for 25 to 36 infusions, the number of PML cases diagnosed during this period is divided by the total number of patients ever exposed to at least 25 infusions and therefore having risk of developing PML during this time).
As of June 30, 2011, some 88,100 patients had been dosed with natalizumab in the postmarketing setting.22 The overall risk of PML in these patients was originally estimated to be 1 in 1,000 individuals taking the drug; the risk now is estimated to be 1.66 in 1,000 patients. Postmarketing surveillance has revealed differences in risk when duration of therapy is considered (Figure 1).22 The most recent data indicate that, for those receiving between 1 and 24 infusions, the risk of PML is exceedingly low: 0.3 per 1,000 patients. For patients receiving between 25 and 36 infusions, the PML risk increases to approximately 1.5 per 1,000 patients. Although the number of cases receiving natalizumab for more than 36 infusions is more limited, the risk does not appear to increase further—the most recent estimated risk of PML for patients receiving 37 to 48 infusions is 0.9 per 1,000 patients.22 The use of immunosuppressive drugs, including antimetabolites (eg, azathioprine), alkylating agents (eg, cyclophosphamide), or anthracenediones (eg, mitoxantrone) prior to natalizumab exposure appears to be an independent risk factor for natalizumab-associated PML, raising risk by approximately two- to fourfold.

Of the first 35 cases of natalizumab-associated PML, 10 cases (29%) were fatal. Among surviving patients, the level of disability was found to be severe in 48%; moderate in 36%; and mild in 16%.23 Improved survival was associated with younger age, less MS-related disability prior to PML, more localized disease on brain MRI at diagnosis, and shorter time from symptom onset to PML diagnosis.23 As of August 4, 2011, there were 150 confirmed cases of natalizumab-associated PML (58 in the United States, 85 in Europe, and 7 from the rest of the world); of these, 29 (19%) have died.22

VARIATION IN PATIENT RISK TOLERANCE

The postmarketing surveillance of natalizumab clearly demonstrates that risk is associated with administration of the drug, but risk tolerance varies considerably among individuals with MS. Some patients elect to use natalizumab despite the risk of PML, even when they have relatively mild MS. Other patients decline use of natalizumab even when their MS is severe and has responded poorly to other disease-modifying drugs.

In most cases, based on my experience, patients accept the risk of natalizumab-associated PML if MS disease is their primary consideration. Another major factor is the patient’s prior experiences with disease-modifying drugs; patients who have experienced breakthrough disease activity despite treatment with first-line drugs commonly opt for natalizumab regardless of the risk of PML.

Interestingly, the treating neurologist’s perception of MS severity and risk of PML may differ from the patient’s perception. In a study of 69 natalizumab-treated MS patients and 66 neurologists, Heesen et al found that patients had a significantly worse perception of their disease and were more willing to assume treatment risks and continue natalizumab therapy than their neurologists were.24 About one-half of the neurologists said that they would discontinue natalizumab at a risk level of 1 in 5,000 or lower, whereas only 17% of the patients would stop at this risk level. This finding has significant implications for clinical practice and implies that the neurologist should discuss concerns about MS and risk of treatment with the patient in order to tailor the decision to the patient’s concerns.

Interest in identifying biomarkers to aid in quantifying risk is ongoing. Chen et al found that subclinical reactivation of the JC virus (JCV) occurred frequently in 19 natalizumab-treated MS patients.25 Another study of 24 natalizumab-treated MS patients found no JCV DNA in the blood, although JCV DNA was found in the urine in 25% of patients.26 A large survey of blood and urine from natalizumab-treated MS patients found low sensitivity and specificity for JCV DNA as a predictor for subsequent PML.27 In this study of more than 1,000 natalizumab-treated patients, JCV DNA was detected in 0.3% of patients’ plasma and in 26% of patients’ urine, but PML did not develop in any patient who was JCV-positive. Among five natalizumab-treated patients who developed PML, JCV DNA was detected in none before the advent of symptoms. The presence of JCV DNA in bodily fluids is important for the diagnosis of PML, but it currently holds no predictive clinical value. At present, measuring JCV DNA in blood, cells, or urine as a predictive biomarker for natalizumab-associated PML does not appear to be clinically useful.

Stratifying risk by measuring JCV serology, however, does appear to be a useful strategy. Investigators using a two-step assay to detect and quantify JCV antibodies found 53.6% of MS patients to be seropositive, with a false-negative rate of 2.5%. Of most interest, all 17 natalizumab-associated PML patients who had available blood samples taken an average of 2 years before onset of PML tested positive for JCV antibodies.28 Although studies are ongoing, classification according to JCV-antibody status may be helpful in advising patients. Patients who are JCV-antibody seronegative (about one-half of patients) appear to be at extraordinarily low risk for PML. In these patients, use of natalizumab could be liberalized and continued as long as the JCV-antibody status remains negative. In patients who are seropositive for JCV antibodies, caution is recommended, particularly for patients who had prior immunosuppressive drug therapy and for patients who have received treatment for more than 24 months. Even in JCV-antibody–seropositive patients, use of natalizumab may be advisable depending on disease severity, available options, and the patient’s risk tolerance. JCV-antibody testing is a rare example of a clinically useful biomarker that can guide specific treatment decisions in the field of neurology.

CURRENT PRACTICE: A PERSONAL MANAGEMENT ALGORITHM

Based on current evidence, the following opinion on the use of natalizumab for the treatment of MS is offered as a supplement to approved prescribing information. The neurologist must individualize the treatment decision for each patient and recognize that these general comments represent a personal opinion. Several factors affect decisions about the use of disease-modifying drugs in MS, and specifically use of natalizumab: How severe is the disease, and what is the prognosis for future disease progression from the neurologist’s perspective? How concerned is the patient about current or future MS symptoms and disability? What is the patient’s tolerance for medication side effects? For risk taking? Has there been prior immunosuppressive therapy? What is the JCV antibody status? What other options are available for disease management?

These issues require discussion among the neurologist, the patient, and the patient’s family. The neurologist should provide input on disease status, an opinion about prognosis, and a description of appropriate options for disease management. Many patients also want a global recommendation (ie, “Tell me what you think I should do”). The neurologist must tailor that global recommendation to the patient’s perceptions of his or her MS, its treatment, and preferences regarding treatment options.

Figure 2. (A) Decision-making algorithm for patients with active multiple sclerosis (MS) who are treatment-naïve, (B) for patients with MS who are already taking a first-line drug, and (C) for patients who have been receiving natalizumab for more than 2 years. JCV = JC virus
My current, evolving view of the management of relapsing-remitting MS is outlined in Figure 2. For treatment-naïve patients with active MS who are seronegative for JCV antibodies, I recommend that natalizumab be considered, with JCV serology repeated yearly (Figure 2A). For patients who are JCV-antibody–seronegative and have breakthrough disease, I also recommend natalizumab with JCV serology repeated yearly. If the patient is treatment-naïve and seropositive, I recommend a first-line drug or fingolimod. If the seropositive patient develops breakthrough disease, is risk tolerant, and has not been treated with prior immunosuppressive drugs, I advise switching to natalizumab for 1 to 2 years to determine response and then reassess.

For patients who are already receiving treatment with a first-line drug and whose disease is well controlled, I make no changes in treatment until a breakthrough occurs, at which time I recommend switching to natalizumab with JCV antibody status assessed yearly (Figure 2B). If a patient has been taking natalizumab for more than 2 years and is seronegative, I advise continuing natalizumab (Figure 2C). If seropositive after 2 or more years of natalizumab therapy, I recommend switching to fingolimod and monitoring for disease reactivation.

 

 

DISCUSSION

Dr. Calabrese: Have you perceived growing concern over PML among the MS patient population over the past 2 to 3 years?

Dr. Rudick: I would say that it’s pretty stable. Patients who are risk intolerant select out of natalizumab. Some patients would just as soon take their chances with MS rather than deal with additional risk. Since we talk about the risk of PML with patients prior to treatment, the patients who choose natalizumab are able to deal with the risk. The difficult cases are those patients who will be severely disabled before long but choose not to go on natalizumab because they’re very risk averse. It gets even more complicated when the closest family member (parents or spouse) want their relative to use natalizumab but the patient is risk averse. This can become quite complicated—for example, I’ve seen situations where the patient is a minor, and one parent wants their child to use natalizumab but the other is risk-averse. Spouses often see risk differently, and this has led to interesting and difficult discussions. In the case of a child with MS, I listen to preferences from family members, but otherwise I empower the patient to drive the decision.

Dr. Major: Our data seem to suggest a higher percentage of individuals who are seropositive—about 56%. The issue, however, is a lack of a standard to define seropositive and seronegative. I suggested that the natalizumab manufacturer collect a bank of samples and allocate them to laboratories with no vested interest for polymerase chain reaction (PCR) assays, from which consensus definitions of seropositive and seronegative could be developed.

Dr. Calabrese: Why is there no confirmatory immunoassay for this virus? We don’t have false positives for hepatitis B or human immunodeficiency virus. We still seem to be relying on older technologies.

Dr. Major: To determine the level of antibody, an enzyme-linked immunosorbent assay is just fine.

Dr. Calabrese: That’s for sensitivity, but what about specificity?

Dr. Major: Specificity is quite good. Everybody now uses the same antigen, the polyoma capsid antigen, VP1, to detect productive viral infection, but there are no set standards for the cutpoints to classify as seropositive and seronegative. Certainly, there’s high sensitivity with PCR to detect JCV DNA in cerebrospinal fluid (CSF), because we’re able to detect very low copy levels of JCV DNA in the CSF.

Dr. Simpson: I would like to see a quantitative measure of risk versus benefit for all of the drugs used in MS, not just natalizumab. When you look at any clinical trial, you see a table of adverse events and you see efficacy measures, but you don’t see the two combined. This really is necessary to compare drug A with drug B. Instead, we end up making decisions based on risk tolerance and rather soft criteria. One could argue that we don’t want to be so algorithmic that we take the art out of medicine, but the criteria we use to make decisions are quite soft. I wonder whether you have any suggestions on a more quantitative approach.

Dr. Rudick: This is an important point, but a difficult problem. We have much more information about PML associated with natalizumab than we do about many serious adverse events. For example, there are rare adverse events with interferon beta—severe depression, liver injury, and so forth. But we don’t have precise quantitative data on most adverse drug effects, and in general adverse events are underreported in clinical practice.

The natalizumab-PML situation is somewhat unique. PML is a dramatic, often fatal, disease that is virtually never observed spontaneously in MS, and the strict reporting requirements have resulted in near-complete ascertainment and more precise risk estimates. This situation doesn’t apply to most adverse events associated with other therapies—even for some severe adverse events. But you are correct—focusing exclusively on the risk of PML seems somewhat simplistic because there are clear risks with other drugs, and these need to be factored into treatment decisions.

Dr. Molloy: Do you have good tools that predict how a patient with MS will do over time?

Dr. Fox: We have fair tools, not great tools.

Dr. Rudick: We’re diagnosing MS earlier, sometimes at the first symptom. We’re even beginning to recognize it in patients without symptoms who have MS observed as an incidental MRI finding. It is difficult at the earliest stage of MS to predict severity with any confidence. The best predictor we have is the severity of the disease by MRI criteria. This can provide a general guide to treatment decisions, but it is an imprecise predictor.

References
  1. Weinshenker BG, Bass B, Rice GPA, et al. The natural history of multiple sclerosis: a geographically based study. I. Clinical course and disability. Brain 1999; 112:133146.
  2. Jacobs LD, Cookfair DL, Rudick RA, et al. Intramuscular interferon beta-1a for disease progression in relapsing multiple sclerosis. Ann Neurol 1996; 39:285294.
  3. The IFNB Multiple Sclerosis Study Group. Interferon beta-1b is effective in relapsing-remitting multiple sclerosis I: clinical results of a multicenter, randomized, double-blind, placebo-controlled trial. Neurology 1993; 43:655661.
  4. IFNB Multiple Sclerosis Study Group, The University of British Columbia MS/MRI Analysis Group. Interferon beta-1b in the treatment of multiple sclerosis: final outcome of the randomized controlled trial. The IFNB Multiple Sclerosis Study Group and the University of British Columbia MS/MRI Analysis Group. Neurology 1995; 45:12771285.
  5. Johnson KP, Brooks BR, Cohen JA, et al. Copolymer 1 reduces relapse rate and improves disability in relapsing-remitting multiple sclerosis: results of a phase III multicenter, double-blind, placebo-controlled trial. Neurology 1995; 45:12681276.
  6. PRISMS (Prevention of Relapses and Disability by Interferon beta-1a Subcutaneously in Multiple Sclerosis) Study Group. Randomised double-blind placebo-controlled study of interferon beta-1a in relapsing/remitting multiple sclerosis. Lancet 1998; 352:14981504.
  7. Kappos L, Radue EW, O’Connor P, et al. A placebo-controlled trial of oral fingolimod in relapsing multiple sclerosis. N Engl J Med 2010; 362:387401.
  8. Cohen JA, Barkhof F, Comi G, et al. Oral fingolimod or intramuscular interferon for relapsing multiple sclerosis. N Engl J Med 2010; 362:402415.
  9. Gilenya [package insert]. East Hanover, NJ: Novartis Pharmaceuticals Corporation; 2011.
  10. Coles AJ, Fox E, Vladic A, et al. Alemtuzumab versus interferon beta-1a in early relapsing-remitting multiple sclerosis: post-hoc and subset analyses of clinical efficacy outcomes. Lancet Neurol 2011; 10:338348.
  11. Nicholas R, Giannetti P, Alsanousi A, Friede T, Muraro PA. Development of oral immunomodulatory agents in the management of multiple sclerosis. Drug Des Devel Ther 2011; 5:255274.
  12. Consortium of Multiple Sclerosis Centers (CMSC). Phase II study with ocrelizumab shows significant reduction in disease activity. CMSC Web site. http://mscare.org/cmsc/index.php?option=com_content&task=view&id=1081&Itemid=1465. Published October 15, 2010. Accessed August 26, 2011.
  13. Tysabri [package insert]. South San Francisco, CA: Elan Pharmaceuticals, Inc.; 2011.
  14. Polman CH, O’Connor PW, Havrdova E, et al. A randomized, placebo-controlled trial of natalizumab for relapsing multiple sclerosis. N Engl J Med 2006; 354:899910.
  15. Miller DH, Khan OA, Sheremata WA, et al. A controlled trial of natalizumab for relapsing multiple sclerosis. N Engl J Med 2003; 348:1523.
  16. Miller DH, Soon D, Fernando KT, et al. MRI outcomes in a placebo-controlled trial of natalizumab in relapsing MS. Neurology 2007; 68:13901401.
  17. Havrdova E, Galetta S, Hutchinson M, et al. Effect of natalizumab on clinical and radiological disease activity in multiple sclerosis: a retrospective analysis of the Natalizumab Safety and Efficacy in Relapsing-Remitting Multiple Sclerosis (AFFIRM) study. Lancet Neurol 2009; 8:254260.
  18. Rudick RA, Miller D, Hass S, et al. Health-related quality of life in multiple sclerosis: effects of natalizumab. Ann Neurol 2007; 62:335346.
  19. Rudick RA, Stuart WH, Calabresi PA, et al. Natalizumab plus interferon beta-1a for relapsing multipls sclerosis. N Engl J Med 2006; 354:911923.
  20. Yousry TA, Major EO, Ryschkewitsch C, et al. Evaluation of patients treated with natalizumab for progressive multifocal leukoencephalopathy. N Engl J Med 2006; 354:924933.
  21. O’Connor PW, Goodman A, Kappos L, et al. Disease activity return during natalizumab treatment interruption in patients with multiple sclerosis [published online ahead of print May 4, 2011]. Neurology 2011; 76:18581865.
  22. Update on Tysabri and PML. National Multiple Sclerosis Society Web site. http://www.nationalmssociety.org/news/news-detail/index.aspx?nid=2308. Published April 11, 2011. Updated May 23, 2011. Accessed June 22, 2011.
  23. Vermersch P, Kappos L, Gold R, et al. Clinical outcomes of natalizumab-associated progressive multifocal leukoencephalopathy. Neurology 2011; 76:16971704.
  24. Heesen C, Kleiter I, Nguyen F, et al. Risk perception in natalizumab-treated multiple sclerosis patients and their neurologists. Mult Scler 2010; 16:15071512.
  25. Chen Y, Bord E, Tompkins T, et al. Asymptomatic reactivation of JC virus in patients treated with natalizumab. N Engl J Med 2009; 361:10671074.
  26. Jilek S, Jaquiery E, Hirsch HH, et al. Immune responses to JC virus in patients with multiple sclerosis treated with natalizumab: a cross-sectional and longitudinal study. Lancet Neurol 2010; 9:264272.
  27. Rudick RA, O’Connor PW, Polman CH, et al. Assessment of JC virus DNA in blood and urine from natalizumab-treated patients. Ann Neurol 2010; 68:304310.
  28. Gorelik L, Lerner M, Bixler S, et al. Anti-JC virus antibodies: implications for PML risk stratification. Ann Neurol 2010; 68:295303.
Article PDF
Author and Disclosure Information

Richard A. Rudick, MD
Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University; Director, Mellen Center for Multiple Sclerosis Treatment and Research; Vice Chair, Research and Development in the Neurological Institute; Cleveland Clinic, Cleveland, OH

Correspondence: Richard A. Rudick, MD, Director, Mellen Center for Multiple Sclerosis, Cleveland Clinic, 9500 Euclid Ave., U10, Cleveland, OH 44195; rudickr@ccf.org

Dr. Rudick reported consulting relationships with Biogen Idec, Genzyme Corporation, and Pfizer Inc.

This article was developed from an audio transcript of Dr. Rudick’s presentation at a roundtable convened at Cleveland Clinic on January 31, 2011. The transcript was edited by the Cleveland Clinic Journal of Medicine staff for clarity and conciseness, and was then reviewed, revised, and approved by Dr. Rudick.

Publications
Page Number
S18-S23
Author and Disclosure Information

Richard A. Rudick, MD
Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University; Director, Mellen Center for Multiple Sclerosis Treatment and Research; Vice Chair, Research and Development in the Neurological Institute; Cleveland Clinic, Cleveland, OH

Correspondence: Richard A. Rudick, MD, Director, Mellen Center for Multiple Sclerosis, Cleveland Clinic, 9500 Euclid Ave., U10, Cleveland, OH 44195; rudickr@ccf.org

Dr. Rudick reported consulting relationships with Biogen Idec, Genzyme Corporation, and Pfizer Inc.

This article was developed from an audio transcript of Dr. Rudick’s presentation at a roundtable convened at Cleveland Clinic on January 31, 2011. The transcript was edited by the Cleveland Clinic Journal of Medicine staff for clarity and conciseness, and was then reviewed, revised, and approved by Dr. Rudick.

Author and Disclosure Information

Richard A. Rudick, MD
Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University; Director, Mellen Center for Multiple Sclerosis Treatment and Research; Vice Chair, Research and Development in the Neurological Institute; Cleveland Clinic, Cleveland, OH

Correspondence: Richard A. Rudick, MD, Director, Mellen Center for Multiple Sclerosis, Cleveland Clinic, 9500 Euclid Ave., U10, Cleveland, OH 44195; rudickr@ccf.org

Dr. Rudick reported consulting relationships with Biogen Idec, Genzyme Corporation, and Pfizer Inc.

This article was developed from an audio transcript of Dr. Rudick’s presentation at a roundtable convened at Cleveland Clinic on January 31, 2011. The transcript was edited by the Cleveland Clinic Journal of Medicine staff for clarity and conciseness, and was then reviewed, revised, and approved by Dr. Rudick.

Article PDF
Article PDF

Multiple sclerosis (MS) is an autoimmune disease whose inflammatory process causes demyelination, axonal loss, and neurodegeneration, all of which can lead to progressive neurologic disability. Without treatment, the risk of progressive disability 15 to 20 years after the onset of MS has been estimated to be as high as 50%.1

Seven drugs have been approved by the US Food and Drug Administration (FDA) for the treatment of MS. Of these, interferon beta drugs and glatiramer acetate are generally considered as first-line agents based on extensive experience and relative safety. Indeed, reports with followup approaching 20 years have not identified significant safety concerns. However, these agents have only modest efficacy, reducing by approximately 30% the frequency of relapse in patients with relapsing-remitting MS.2–6

Natalizumab, and more recently, fingolimod, are generally used as second-line agents. Fingolimod, the first oral agent to receive FDA approval for the treatment of relapsing-remitting MS, is a functional antagonist of sphingosine-1-phosphate receptors. The reductions in annualized relapse rates in two phase 3 controlled trials of fingolimod were approximately 55% compared with placebo7 or intramuscular interferon beta-1a.8 Because of its more convenient oral route of administration and its documented efficacy, widespread use of fingolimod is anticipated. However, adverse reactions affecting more than 10% of patients include headache, influenza, diarrhea, back pain, liver transaminase elevations, and cough.9 Because sphingosine-1-phosphate receptors are widespread in many body tissues, off-target effects of fingolimod may be problematic and long-term toxicity is unknown.

In addition to natalizumab and fingolimod, which are currently available for use as second-line agents, several other MS therapies are showing promise. Oral cladribine, teriflunomide, and laquinimod have reported positive phase 3 results in publication or at national meetings, and several other drugs are in late stages of development (alemtuzumab, BG-12, ocrelizumab) based on encouraging phase 2 results.10–12 Thus, the options for MS patients are expanding, but drugs with higher efficacy also may pose greater risk.

NATALIZUMAB: ROBUST BENEFITS BUT ASSOCIATED RISK

Natalizumab is a humanized monoclonal antibody that binds to alpha-4 integrin on leukocytes. By inhibiting alpha-4 integrin, natalizumab, the first of a new class of selective adhesion-molecule inhibitors, impedes migration of activated mononuclear leukocytes into the brain and gut.13

Significant efficacy

Two phase 3 studies demonstrated more robust efficacy of natalizumab in patients with relapsing-remitting MS than had been observed in prior studies with other agents.14,15 In the Natalizumab Safety and Efficacy in Relapsing-Remitting Multiple Sclerosis (AFFIRM) study, which followed patients over 2 years of treatment, natalizumab was associated with a 68% reduction in the annualized relapse rate14; a 92% reduction in gadolinium-enhanced lesions on magnetic resonance imaging (MRI), which indicate new, active lesions16; and an 83% reduction in the mean number of new or enlarging T2 lesions16 compared with placebo. The likelihood of confirmed worsening on the Kurtzke Expanded Disability Status Score, which is the standard measure for MS-related disability, was also 42% lower in patients assigned to natalizumab compared with placebo.14

Other reported benefits from natalizumab therapy include a significantly increased probability of maintaining disease-free status17 and clinically significant improvements on patient-reported quality-of-life measures.18 Although there have been no head-to-head studies of natalizumab with interferon beta, glatiramer acetate, or fingolimod, there is a widespread view that treatment benefits of natalizumab exceed those of other disease-modifying drugs. In clinical practice, patients with MS who experience breakthrough disease on standard disease-modifying drugs are routinely observed to achieve disease control after switching to natalizumab. Thus, based purely on efficacy, patient-reported outcomes, and the convenience of once-monthly intravenous infusion, natalizumab represents an extremely attractive treatment option for patients with relapsing-remitting MS.

Use discontinued in 2005

Natalizumab was approved for treatment of relapsing-remitting MS in November 2004, using the FDA accelerated review pathway. The approval was based on the first-year results of the AFFIRM14 and the Natalizumab plus Interferon Beta-1a for Relapsing Remitting Multiple Sclerosis (SENTINEL)19 studies, both of which were completed in February 2005. In the 3 to 4 months between the drug’s approval and completion of the AFFIRM and SENTINEL studies, approximately 7,000 patients with relapsing-remitting MS received treatment with natalizumab. In February 2005, shortly after the release of the 2-year data, three cases of progressive multifocal leukoencephalopathy (PML) were identified in natalizumab-treated patients (one with Crohn disease, the other two with MS). Clinical and research use of natalizumab was abruptly suspended that month, pending a comprehensive safety review.

A safety study evaluated 3,116 patients who had received natalizumab over a mean exposure of 17.9 monthly doses.20 The study failed to identify any additional cases of PML and concluded that the risk of PML was approximately 1 in 1,000 patients. Abrupt discontinuation of natalizumab also allowed systematic assessment of disease behavior following treatment interruption. In 1,866 patients who had received natalizumab during clinical trials but who discontinued natalizumab after PML was recognized, MS relapses and gadolinium-enhancing lesions returned approximately to baseline levels within 4 to 7 months of natalizumab suspension. Reactivation of MS disease activity was observed even in patients who instituted one of the first-line disease-modifying drugs as substitute therapy.21

Based on the strong efficacy data and the extensive safety review, an FDA advisory committee recommended reintroduction, and natalizumab was returned to the market in June 2006. Natalizumab may be administered only in accredited infusion centers that agree to a monthly reporting regimen designed to identify all cases of PML. In the United States, natalizumab is available to patients only through the Tysabri Outreach Unified Commitment to Health (TOUCH) Prescribing Program, a restricted distribution program. Aggressive monitoring and reporting is also required in other regions of the world, so that ascertainment of PML associated with natalizumab is thought to be relatively complete.

 

 

Risk related to duration of therapy

Source: Data on file. Biogen Idec; 2011.
Figure 1. Progressive multifocal leukoencephalopathy (PML) incidence estimates by treatment duration (A) and treatment epoch (B), calculated based on natalizumab exposure through January 31, 2011, and 95 confirmed cases as of February 2, 2011. (A) The incidence for each time period is calculated as the number of PML cases divided by the number of patients exposed to natalizumab (eg, for ≥ 24 infusions, the number of PML cases diagnosed with exposure of 24 infusions or more is divided by the total number of patients exposed to at least 24 infusions). (B) The incidence for each epoch is calculated as the number of PML cases divided by the number of patients exposed to natalizumab (eg, for 25 to 36 infusions, the number of PML cases diagnosed during this period is divided by the total number of patients ever exposed to at least 25 infusions and therefore having risk of developing PML during this time).
As of June 30, 2011, some 88,100 patients had been dosed with natalizumab in the postmarketing setting.22 The overall risk of PML in these patients was originally estimated to be 1 in 1,000 individuals taking the drug; the risk now is estimated to be 1.66 in 1,000 patients. Postmarketing surveillance has revealed differences in risk when duration of therapy is considered (Figure 1).22 The most recent data indicate that, for those receiving between 1 and 24 infusions, the risk of PML is exceedingly low: 0.3 per 1,000 patients. For patients receiving between 25 and 36 infusions, the PML risk increases to approximately 1.5 per 1,000 patients. Although the number of cases receiving natalizumab for more than 36 infusions is more limited, the risk does not appear to increase further—the most recent estimated risk of PML for patients receiving 37 to 48 infusions is 0.9 per 1,000 patients.22 The use of immunosuppressive drugs, including antimetabolites (eg, azathioprine), alkylating agents (eg, cyclophosphamide), or anthracenediones (eg, mitoxantrone) prior to natalizumab exposure appears to be an independent risk factor for natalizumab-associated PML, raising risk by approximately two- to fourfold.

Of the first 35 cases of natalizumab-associated PML, 10 cases (29%) were fatal. Among surviving patients, the level of disability was found to be severe in 48%; moderate in 36%; and mild in 16%.23 Improved survival was associated with younger age, less MS-related disability prior to PML, more localized disease on brain MRI at diagnosis, and shorter time from symptom onset to PML diagnosis.23 As of August 4, 2011, there were 150 confirmed cases of natalizumab-associated PML (58 in the United States, 85 in Europe, and 7 from the rest of the world); of these, 29 (19%) have died.22

VARIATION IN PATIENT RISK TOLERANCE

The postmarketing surveillance of natalizumab clearly demonstrates that risk is associated with administration of the drug, but risk tolerance varies considerably among individuals with MS. Some patients elect to use natalizumab despite the risk of PML, even when they have relatively mild MS. Other patients decline use of natalizumab even when their MS is severe and has responded poorly to other disease-modifying drugs.

In most cases, based on my experience, patients accept the risk of natalizumab-associated PML if MS disease is their primary consideration. Another major factor is the patient’s prior experiences with disease-modifying drugs; patients who have experienced breakthrough disease activity despite treatment with first-line drugs commonly opt for natalizumab regardless of the risk of PML.

Interestingly, the treating neurologist’s perception of MS severity and risk of PML may differ from the patient’s perception. In a study of 69 natalizumab-treated MS patients and 66 neurologists, Heesen et al found that patients had a significantly worse perception of their disease and were more willing to assume treatment risks and continue natalizumab therapy than their neurologists were.24 About one-half of the neurologists said that they would discontinue natalizumab at a risk level of 1 in 5,000 or lower, whereas only 17% of the patients would stop at this risk level. This finding has significant implications for clinical practice and implies that the neurologist should discuss concerns about MS and risk of treatment with the patient in order to tailor the decision to the patient’s concerns.

Interest in identifying biomarkers to aid in quantifying risk is ongoing. Chen et al found that subclinical reactivation of the JC virus (JCV) occurred frequently in 19 natalizumab-treated MS patients.25 Another study of 24 natalizumab-treated MS patients found no JCV DNA in the blood, although JCV DNA was found in the urine in 25% of patients.26 A large survey of blood and urine from natalizumab-treated MS patients found low sensitivity and specificity for JCV DNA as a predictor for subsequent PML.27 In this study of more than 1,000 natalizumab-treated patients, JCV DNA was detected in 0.3% of patients’ plasma and in 26% of patients’ urine, but PML did not develop in any patient who was JCV-positive. Among five natalizumab-treated patients who developed PML, JCV DNA was detected in none before the advent of symptoms. The presence of JCV DNA in bodily fluids is important for the diagnosis of PML, but it currently holds no predictive clinical value. At present, measuring JCV DNA in blood, cells, or urine as a predictive biomarker for natalizumab-associated PML does not appear to be clinically useful.

Stratifying risk by measuring JCV serology, however, does appear to be a useful strategy. Investigators using a two-step assay to detect and quantify JCV antibodies found 53.6% of MS patients to be seropositive, with a false-negative rate of 2.5%. Of most interest, all 17 natalizumab-associated PML patients who had available blood samples taken an average of 2 years before onset of PML tested positive for JCV antibodies.28 Although studies are ongoing, classification according to JCV-antibody status may be helpful in advising patients. Patients who are JCV-antibody seronegative (about one-half of patients) appear to be at extraordinarily low risk for PML. In these patients, use of natalizumab could be liberalized and continued as long as the JCV-antibody status remains negative. In patients who are seropositive for JCV antibodies, caution is recommended, particularly for patients who had prior immunosuppressive drug therapy and for patients who have received treatment for more than 24 months. Even in JCV-antibody–seropositive patients, use of natalizumab may be advisable depending on disease severity, available options, and the patient’s risk tolerance. JCV-antibody testing is a rare example of a clinically useful biomarker that can guide specific treatment decisions in the field of neurology.

CURRENT PRACTICE: A PERSONAL MANAGEMENT ALGORITHM

Based on current evidence, the following opinion on the use of natalizumab for the treatment of MS is offered as a supplement to approved prescribing information. The neurologist must individualize the treatment decision for each patient and recognize that these general comments represent a personal opinion. Several factors affect decisions about the use of disease-modifying drugs in MS, and specifically use of natalizumab: How severe is the disease, and what is the prognosis for future disease progression from the neurologist’s perspective? How concerned is the patient about current or future MS symptoms and disability? What is the patient’s tolerance for medication side effects? For risk taking? Has there been prior immunosuppressive therapy? What is the JCV antibody status? What other options are available for disease management?

These issues require discussion among the neurologist, the patient, and the patient’s family. The neurologist should provide input on disease status, an opinion about prognosis, and a description of appropriate options for disease management. Many patients also want a global recommendation (ie, “Tell me what you think I should do”). The neurologist must tailor that global recommendation to the patient’s perceptions of his or her MS, its treatment, and preferences regarding treatment options.

Figure 2. (A) Decision-making algorithm for patients with active multiple sclerosis (MS) who are treatment-naïve, (B) for patients with MS who are already taking a first-line drug, and (C) for patients who have been receiving natalizumab for more than 2 years. JCV = JC virus
My current, evolving view of the management of relapsing-remitting MS is outlined in Figure 2. For treatment-naïve patients with active MS who are seronegative for JCV antibodies, I recommend that natalizumab be considered, with JCV serology repeated yearly (Figure 2A). For patients who are JCV-antibody–seronegative and have breakthrough disease, I also recommend natalizumab with JCV serology repeated yearly. If the patient is treatment-naïve and seropositive, I recommend a first-line drug or fingolimod. If the seropositive patient develops breakthrough disease, is risk tolerant, and has not been treated with prior immunosuppressive drugs, I advise switching to natalizumab for 1 to 2 years to determine response and then reassess.

For patients who are already receiving treatment with a first-line drug and whose disease is well controlled, I make no changes in treatment until a breakthrough occurs, at which time I recommend switching to natalizumab with JCV antibody status assessed yearly (Figure 2B). If a patient has been taking natalizumab for more than 2 years and is seronegative, I advise continuing natalizumab (Figure 2C). If seropositive after 2 or more years of natalizumab therapy, I recommend switching to fingolimod and monitoring for disease reactivation.

 

 

DISCUSSION

Dr. Calabrese: Have you perceived growing concern over PML among the MS patient population over the past 2 to 3 years?

Dr. Rudick: I would say that it’s pretty stable. Patients who are risk intolerant select out of natalizumab. Some patients would just as soon take their chances with MS rather than deal with additional risk. Since we talk about the risk of PML with patients prior to treatment, the patients who choose natalizumab are able to deal with the risk. The difficult cases are those patients who will be severely disabled before long but choose not to go on natalizumab because they’re very risk averse. It gets even more complicated when the closest family member (parents or spouse) want their relative to use natalizumab but the patient is risk averse. This can become quite complicated—for example, I’ve seen situations where the patient is a minor, and one parent wants their child to use natalizumab but the other is risk-averse. Spouses often see risk differently, and this has led to interesting and difficult discussions. In the case of a child with MS, I listen to preferences from family members, but otherwise I empower the patient to drive the decision.

Dr. Major: Our data seem to suggest a higher percentage of individuals who are seropositive—about 56%. The issue, however, is a lack of a standard to define seropositive and seronegative. I suggested that the natalizumab manufacturer collect a bank of samples and allocate them to laboratories with no vested interest for polymerase chain reaction (PCR) assays, from which consensus definitions of seropositive and seronegative could be developed.

Dr. Calabrese: Why is there no confirmatory immunoassay for this virus? We don’t have false positives for hepatitis B or human immunodeficiency virus. We still seem to be relying on older technologies.

Dr. Major: To determine the level of antibody, an enzyme-linked immunosorbent assay is just fine.

Dr. Calabrese: That’s for sensitivity, but what about specificity?

Dr. Major: Specificity is quite good. Everybody now uses the same antigen, the polyoma capsid antigen, VP1, to detect productive viral infection, but there are no set standards for the cutpoints to classify as seropositive and seronegative. Certainly, there’s high sensitivity with PCR to detect JCV DNA in cerebrospinal fluid (CSF), because we’re able to detect very low copy levels of JCV DNA in the CSF.

Dr. Simpson: I would like to see a quantitative measure of risk versus benefit for all of the drugs used in MS, not just natalizumab. When you look at any clinical trial, you see a table of adverse events and you see efficacy measures, but you don’t see the two combined. This really is necessary to compare drug A with drug B. Instead, we end up making decisions based on risk tolerance and rather soft criteria. One could argue that we don’t want to be so algorithmic that we take the art out of medicine, but the criteria we use to make decisions are quite soft. I wonder whether you have any suggestions on a more quantitative approach.

Dr. Rudick: This is an important point, but a difficult problem. We have much more information about PML associated with natalizumab than we do about many serious adverse events. For example, there are rare adverse events with interferon beta—severe depression, liver injury, and so forth. But we don’t have precise quantitative data on most adverse drug effects, and in general adverse events are underreported in clinical practice.

The natalizumab-PML situation is somewhat unique. PML is a dramatic, often fatal, disease that is virtually never observed spontaneously in MS, and the strict reporting requirements have resulted in near-complete ascertainment and more precise risk estimates. This situation doesn’t apply to most adverse events associated with other therapies—even for some severe adverse events. But you are correct—focusing exclusively on the risk of PML seems somewhat simplistic because there are clear risks with other drugs, and these need to be factored into treatment decisions.

Dr. Molloy: Do you have good tools that predict how a patient with MS will do over time?

Dr. Fox: We have fair tools, not great tools.

Dr. Rudick: We’re diagnosing MS earlier, sometimes at the first symptom. We’re even beginning to recognize it in patients without symptoms who have MS observed as an incidental MRI finding. It is difficult at the earliest stage of MS to predict severity with any confidence. The best predictor we have is the severity of the disease by MRI criteria. This can provide a general guide to treatment decisions, but it is an imprecise predictor.

Multiple sclerosis (MS) is an autoimmune disease whose inflammatory process causes demyelination, axonal loss, and neurodegeneration, all of which can lead to progressive neurologic disability. Without treatment, the risk of progressive disability 15 to 20 years after the onset of MS has been estimated to be as high as 50%.1

Seven drugs have been approved by the US Food and Drug Administration (FDA) for the treatment of MS. Of these, interferon beta drugs and glatiramer acetate are generally considered as first-line agents based on extensive experience and relative safety. Indeed, reports with followup approaching 20 years have not identified significant safety concerns. However, these agents have only modest efficacy, reducing by approximately 30% the frequency of relapse in patients with relapsing-remitting MS.2–6

Natalizumab, and more recently, fingolimod, are generally used as second-line agents. Fingolimod, the first oral agent to receive FDA approval for the treatment of relapsing-remitting MS, is a functional antagonist of sphingosine-1-phosphate receptors. The reductions in annualized relapse rates in two phase 3 controlled trials of fingolimod were approximately 55% compared with placebo7 or intramuscular interferon beta-1a.8 Because of its more convenient oral route of administration and its documented efficacy, widespread use of fingolimod is anticipated. However, adverse reactions affecting more than 10% of patients include headache, influenza, diarrhea, back pain, liver transaminase elevations, and cough.9 Because sphingosine-1-phosphate receptors are widespread in many body tissues, off-target effects of fingolimod may be problematic and long-term toxicity is unknown.

In addition to natalizumab and fingolimod, which are currently available for use as second-line agents, several other MS therapies are showing promise. Oral cladribine, teriflunomide, and laquinimod have reported positive phase 3 results in publication or at national meetings, and several other drugs are in late stages of development (alemtuzumab, BG-12, ocrelizumab) based on encouraging phase 2 results.10–12 Thus, the options for MS patients are expanding, but drugs with higher efficacy also may pose greater risk.

NATALIZUMAB: ROBUST BENEFITS BUT ASSOCIATED RISK

Natalizumab is a humanized monoclonal antibody that binds to alpha-4 integrin on leukocytes. By inhibiting alpha-4 integrin, natalizumab, the first of a new class of selective adhesion-molecule inhibitors, impedes migration of activated mononuclear leukocytes into the brain and gut.13

Significant efficacy

Two phase 3 studies demonstrated more robust efficacy of natalizumab in patients with relapsing-remitting MS than had been observed in prior studies with other agents.14,15 In the Natalizumab Safety and Efficacy in Relapsing-Remitting Multiple Sclerosis (AFFIRM) study, which followed patients over 2 years of treatment, natalizumab was associated with a 68% reduction in the annualized relapse rate14; a 92% reduction in gadolinium-enhanced lesions on magnetic resonance imaging (MRI), which indicate new, active lesions16; and an 83% reduction in the mean number of new or enlarging T2 lesions16 compared with placebo. The likelihood of confirmed worsening on the Kurtzke Expanded Disability Status Score, which is the standard measure for MS-related disability, was also 42% lower in patients assigned to natalizumab compared with placebo.14

Other reported benefits from natalizumab therapy include a significantly increased probability of maintaining disease-free status17 and clinically significant improvements on patient-reported quality-of-life measures.18 Although there have been no head-to-head studies of natalizumab with interferon beta, glatiramer acetate, or fingolimod, there is a widespread view that treatment benefits of natalizumab exceed those of other disease-modifying drugs. In clinical practice, patients with MS who experience breakthrough disease on standard disease-modifying drugs are routinely observed to achieve disease control after switching to natalizumab. Thus, based purely on efficacy, patient-reported outcomes, and the convenience of once-monthly intravenous infusion, natalizumab represents an extremely attractive treatment option for patients with relapsing-remitting MS.

Use discontinued in 2005

Natalizumab was approved for treatment of relapsing-remitting MS in November 2004, using the FDA accelerated review pathway. The approval was based on the first-year results of the AFFIRM14 and the Natalizumab plus Interferon Beta-1a for Relapsing Remitting Multiple Sclerosis (SENTINEL)19 studies, both of which were completed in February 2005. In the 3 to 4 months between the drug’s approval and completion of the AFFIRM and SENTINEL studies, approximately 7,000 patients with relapsing-remitting MS received treatment with natalizumab. In February 2005, shortly after the release of the 2-year data, three cases of progressive multifocal leukoencephalopathy (PML) were identified in natalizumab-treated patients (one with Crohn disease, the other two with MS). Clinical and research use of natalizumab was abruptly suspended that month, pending a comprehensive safety review.

A safety study evaluated 3,116 patients who had received natalizumab over a mean exposure of 17.9 monthly doses.20 The study failed to identify any additional cases of PML and concluded that the risk of PML was approximately 1 in 1,000 patients. Abrupt discontinuation of natalizumab also allowed systematic assessment of disease behavior following treatment interruption. In 1,866 patients who had received natalizumab during clinical trials but who discontinued natalizumab after PML was recognized, MS relapses and gadolinium-enhancing lesions returned approximately to baseline levels within 4 to 7 months of natalizumab suspension. Reactivation of MS disease activity was observed even in patients who instituted one of the first-line disease-modifying drugs as substitute therapy.21

Based on the strong efficacy data and the extensive safety review, an FDA advisory committee recommended reintroduction, and natalizumab was returned to the market in June 2006. Natalizumab may be administered only in accredited infusion centers that agree to a monthly reporting regimen designed to identify all cases of PML. In the United States, natalizumab is available to patients only through the Tysabri Outreach Unified Commitment to Health (TOUCH) Prescribing Program, a restricted distribution program. Aggressive monitoring and reporting is also required in other regions of the world, so that ascertainment of PML associated with natalizumab is thought to be relatively complete.

 

 

Risk related to duration of therapy

Source: Data on file. Biogen Idec; 2011.
Figure 1. Progressive multifocal leukoencephalopathy (PML) incidence estimates by treatment duration (A) and treatment epoch (B), calculated based on natalizumab exposure through January 31, 2011, and 95 confirmed cases as of February 2, 2011. (A) The incidence for each time period is calculated as the number of PML cases divided by the number of patients exposed to natalizumab (eg, for ≥ 24 infusions, the number of PML cases diagnosed with exposure of 24 infusions or more is divided by the total number of patients exposed to at least 24 infusions). (B) The incidence for each epoch is calculated as the number of PML cases divided by the number of patients exposed to natalizumab (eg, for 25 to 36 infusions, the number of PML cases diagnosed during this period is divided by the total number of patients ever exposed to at least 25 infusions and therefore having risk of developing PML during this time).
As of June 30, 2011, some 88,100 patients had been dosed with natalizumab in the postmarketing setting.22 The overall risk of PML in these patients was originally estimated to be 1 in 1,000 individuals taking the drug; the risk now is estimated to be 1.66 in 1,000 patients. Postmarketing surveillance has revealed differences in risk when duration of therapy is considered (Figure 1).22 The most recent data indicate that, for those receiving between 1 and 24 infusions, the risk of PML is exceedingly low: 0.3 per 1,000 patients. For patients receiving between 25 and 36 infusions, the PML risk increases to approximately 1.5 per 1,000 patients. Although the number of cases receiving natalizumab for more than 36 infusions is more limited, the risk does not appear to increase further—the most recent estimated risk of PML for patients receiving 37 to 48 infusions is 0.9 per 1,000 patients.22 The use of immunosuppressive drugs, including antimetabolites (eg, azathioprine), alkylating agents (eg, cyclophosphamide), or anthracenediones (eg, mitoxantrone) prior to natalizumab exposure appears to be an independent risk factor for natalizumab-associated PML, raising risk by approximately two- to fourfold.

Of the first 35 cases of natalizumab-associated PML, 10 cases (29%) were fatal. Among surviving patients, the level of disability was found to be severe in 48%; moderate in 36%; and mild in 16%.23 Improved survival was associated with younger age, less MS-related disability prior to PML, more localized disease on brain MRI at diagnosis, and shorter time from symptom onset to PML diagnosis.23 As of August 4, 2011, there were 150 confirmed cases of natalizumab-associated PML (58 in the United States, 85 in Europe, and 7 from the rest of the world); of these, 29 (19%) have died.22

VARIATION IN PATIENT RISK TOLERANCE

The postmarketing surveillance of natalizumab clearly demonstrates that risk is associated with administration of the drug, but risk tolerance varies considerably among individuals with MS. Some patients elect to use natalizumab despite the risk of PML, even when they have relatively mild MS. Other patients decline use of natalizumab even when their MS is severe and has responded poorly to other disease-modifying drugs.

In most cases, based on my experience, patients accept the risk of natalizumab-associated PML if MS disease is their primary consideration. Another major factor is the patient’s prior experiences with disease-modifying drugs; patients who have experienced breakthrough disease activity despite treatment with first-line drugs commonly opt for natalizumab regardless of the risk of PML.

Interestingly, the treating neurologist’s perception of MS severity and risk of PML may differ from the patient’s perception. In a study of 69 natalizumab-treated MS patients and 66 neurologists, Heesen et al found that patients had a significantly worse perception of their disease and were more willing to assume treatment risks and continue natalizumab therapy than their neurologists were.24 About one-half of the neurologists said that they would discontinue natalizumab at a risk level of 1 in 5,000 or lower, whereas only 17% of the patients would stop at this risk level. This finding has significant implications for clinical practice and implies that the neurologist should discuss concerns about MS and risk of treatment with the patient in order to tailor the decision to the patient’s concerns.

Interest in identifying biomarkers to aid in quantifying risk is ongoing. Chen et al found that subclinical reactivation of the JC virus (JCV) occurred frequently in 19 natalizumab-treated MS patients.25 Another study of 24 natalizumab-treated MS patients found no JCV DNA in the blood, although JCV DNA was found in the urine in 25% of patients.26 A large survey of blood and urine from natalizumab-treated MS patients found low sensitivity and specificity for JCV DNA as a predictor for subsequent PML.27 In this study of more than 1,000 natalizumab-treated patients, JCV DNA was detected in 0.3% of patients’ plasma and in 26% of patients’ urine, but PML did not develop in any patient who was JCV-positive. Among five natalizumab-treated patients who developed PML, JCV DNA was detected in none before the advent of symptoms. The presence of JCV DNA in bodily fluids is important for the diagnosis of PML, but it currently holds no predictive clinical value. At present, measuring JCV DNA in blood, cells, or urine as a predictive biomarker for natalizumab-associated PML does not appear to be clinically useful.

Stratifying risk by measuring JCV serology, however, does appear to be a useful strategy. Investigators using a two-step assay to detect and quantify JCV antibodies found 53.6% of MS patients to be seropositive, with a false-negative rate of 2.5%. Of most interest, all 17 natalizumab-associated PML patients who had available blood samples taken an average of 2 years before onset of PML tested positive for JCV antibodies.28 Although studies are ongoing, classification according to JCV-antibody status may be helpful in advising patients. Patients who are JCV-antibody seronegative (about one-half of patients) appear to be at extraordinarily low risk for PML. In these patients, use of natalizumab could be liberalized and continued as long as the JCV-antibody status remains negative. In patients who are seropositive for JCV antibodies, caution is recommended, particularly for patients who had prior immunosuppressive drug therapy and for patients who have received treatment for more than 24 months. Even in JCV-antibody–seropositive patients, use of natalizumab may be advisable depending on disease severity, available options, and the patient’s risk tolerance. JCV-antibody testing is a rare example of a clinically useful biomarker that can guide specific treatment decisions in the field of neurology.

CURRENT PRACTICE: A PERSONAL MANAGEMENT ALGORITHM

Based on current evidence, the following opinion on the use of natalizumab for the treatment of MS is offered as a supplement to approved prescribing information. The neurologist must individualize the treatment decision for each patient and recognize that these general comments represent a personal opinion. Several factors affect decisions about the use of disease-modifying drugs in MS, and specifically use of natalizumab: How severe is the disease, and what is the prognosis for future disease progression from the neurologist’s perspective? How concerned is the patient about current or future MS symptoms and disability? What is the patient’s tolerance for medication side effects? For risk taking? Has there been prior immunosuppressive therapy? What is the JCV antibody status? What other options are available for disease management?

These issues require discussion among the neurologist, the patient, and the patient’s family. The neurologist should provide input on disease status, an opinion about prognosis, and a description of appropriate options for disease management. Many patients also want a global recommendation (ie, “Tell me what you think I should do”). The neurologist must tailor that global recommendation to the patient’s perceptions of his or her MS, its treatment, and preferences regarding treatment options.

Figure 2. (A) Decision-making algorithm for patients with active multiple sclerosis (MS) who are treatment-naïve, (B) for patients with MS who are already taking a first-line drug, and (C) for patients who have been receiving natalizumab for more than 2 years. JCV = JC virus
My current, evolving view of the management of relapsing-remitting MS is outlined in Figure 2. For treatment-naïve patients with active MS who are seronegative for JCV antibodies, I recommend that natalizumab be considered, with JCV serology repeated yearly (Figure 2A). For patients who are JCV-antibody–seronegative and have breakthrough disease, I also recommend natalizumab with JCV serology repeated yearly. If the patient is treatment-naïve and seropositive, I recommend a first-line drug or fingolimod. If the seropositive patient develops breakthrough disease, is risk tolerant, and has not been treated with prior immunosuppressive drugs, I advise switching to natalizumab for 1 to 2 years to determine response and then reassess.

For patients who are already receiving treatment with a first-line drug and whose disease is well controlled, I make no changes in treatment until a breakthrough occurs, at which time I recommend switching to natalizumab with JCV antibody status assessed yearly (Figure 2B). If a patient has been taking natalizumab for more than 2 years and is seronegative, I advise continuing natalizumab (Figure 2C). If seropositive after 2 or more years of natalizumab therapy, I recommend switching to fingolimod and monitoring for disease reactivation.

 

 

DISCUSSION

Dr. Calabrese: Have you perceived growing concern over PML among the MS patient population over the past 2 to 3 years?

Dr. Rudick: I would say that it’s pretty stable. Patients who are risk intolerant select out of natalizumab. Some patients would just as soon take their chances with MS rather than deal with additional risk. Since we talk about the risk of PML with patients prior to treatment, the patients who choose natalizumab are able to deal with the risk. The difficult cases are those patients who will be severely disabled before long but choose not to go on natalizumab because they’re very risk averse. It gets even more complicated when the closest family member (parents or spouse) want their relative to use natalizumab but the patient is risk averse. This can become quite complicated—for example, I’ve seen situations where the patient is a minor, and one parent wants their child to use natalizumab but the other is risk-averse. Spouses often see risk differently, and this has led to interesting and difficult discussions. In the case of a child with MS, I listen to preferences from family members, but otherwise I empower the patient to drive the decision.

Dr. Major: Our data seem to suggest a higher percentage of individuals who are seropositive—about 56%. The issue, however, is a lack of a standard to define seropositive and seronegative. I suggested that the natalizumab manufacturer collect a bank of samples and allocate them to laboratories with no vested interest for polymerase chain reaction (PCR) assays, from which consensus definitions of seropositive and seronegative could be developed.

Dr. Calabrese: Why is there no confirmatory immunoassay for this virus? We don’t have false positives for hepatitis B or human immunodeficiency virus. We still seem to be relying on older technologies.

Dr. Major: To determine the level of antibody, an enzyme-linked immunosorbent assay is just fine.

Dr. Calabrese: That’s for sensitivity, but what about specificity?

Dr. Major: Specificity is quite good. Everybody now uses the same antigen, the polyoma capsid antigen, VP1, to detect productive viral infection, but there are no set standards for the cutpoints to classify as seropositive and seronegative. Certainly, there’s high sensitivity with PCR to detect JCV DNA in cerebrospinal fluid (CSF), because we’re able to detect very low copy levels of JCV DNA in the CSF.

Dr. Simpson: I would like to see a quantitative measure of risk versus benefit for all of the drugs used in MS, not just natalizumab. When you look at any clinical trial, you see a table of adverse events and you see efficacy measures, but you don’t see the two combined. This really is necessary to compare drug A with drug B. Instead, we end up making decisions based on risk tolerance and rather soft criteria. One could argue that we don’t want to be so algorithmic that we take the art out of medicine, but the criteria we use to make decisions are quite soft. I wonder whether you have any suggestions on a more quantitative approach.

Dr. Rudick: This is an important point, but a difficult problem. We have much more information about PML associated with natalizumab than we do about many serious adverse events. For example, there are rare adverse events with interferon beta—severe depression, liver injury, and so forth. But we don’t have precise quantitative data on most adverse drug effects, and in general adverse events are underreported in clinical practice.

The natalizumab-PML situation is somewhat unique. PML is a dramatic, often fatal, disease that is virtually never observed spontaneously in MS, and the strict reporting requirements have resulted in near-complete ascertainment and more precise risk estimates. This situation doesn’t apply to most adverse events associated with other therapies—even for some severe adverse events. But you are correct—focusing exclusively on the risk of PML seems somewhat simplistic because there are clear risks with other drugs, and these need to be factored into treatment decisions.

Dr. Molloy: Do you have good tools that predict how a patient with MS will do over time?

Dr. Fox: We have fair tools, not great tools.

Dr. Rudick: We’re diagnosing MS earlier, sometimes at the first symptom. We’re even beginning to recognize it in patients without symptoms who have MS observed as an incidental MRI finding. It is difficult at the earliest stage of MS to predict severity with any confidence. The best predictor we have is the severity of the disease by MRI criteria. This can provide a general guide to treatment decisions, but it is an imprecise predictor.

References
  1. Weinshenker BG, Bass B, Rice GPA, et al. The natural history of multiple sclerosis: a geographically based study. I. Clinical course and disability. Brain 1999; 112:133146.
  2. Jacobs LD, Cookfair DL, Rudick RA, et al. Intramuscular interferon beta-1a for disease progression in relapsing multiple sclerosis. Ann Neurol 1996; 39:285294.
  3. The IFNB Multiple Sclerosis Study Group. Interferon beta-1b is effective in relapsing-remitting multiple sclerosis I: clinical results of a multicenter, randomized, double-blind, placebo-controlled trial. Neurology 1993; 43:655661.
  4. IFNB Multiple Sclerosis Study Group, The University of British Columbia MS/MRI Analysis Group. Interferon beta-1b in the treatment of multiple sclerosis: final outcome of the randomized controlled trial. The IFNB Multiple Sclerosis Study Group and the University of British Columbia MS/MRI Analysis Group. Neurology 1995; 45:12771285.
  5. Johnson KP, Brooks BR, Cohen JA, et al. Copolymer 1 reduces relapse rate and improves disability in relapsing-remitting multiple sclerosis: results of a phase III multicenter, double-blind, placebo-controlled trial. Neurology 1995; 45:12681276.
  6. PRISMS (Prevention of Relapses and Disability by Interferon beta-1a Subcutaneously in Multiple Sclerosis) Study Group. Randomised double-blind placebo-controlled study of interferon beta-1a in relapsing/remitting multiple sclerosis. Lancet 1998; 352:14981504.
  7. Kappos L, Radue EW, O’Connor P, et al. A placebo-controlled trial of oral fingolimod in relapsing multiple sclerosis. N Engl J Med 2010; 362:387401.
  8. Cohen JA, Barkhof F, Comi G, et al. Oral fingolimod or intramuscular interferon for relapsing multiple sclerosis. N Engl J Med 2010; 362:402415.
  9. Gilenya [package insert]. East Hanover, NJ: Novartis Pharmaceuticals Corporation; 2011.
  10. Coles AJ, Fox E, Vladic A, et al. Alemtuzumab versus interferon beta-1a in early relapsing-remitting multiple sclerosis: post-hoc and subset analyses of clinical efficacy outcomes. Lancet Neurol 2011; 10:338348.
  11. Nicholas R, Giannetti P, Alsanousi A, Friede T, Muraro PA. Development of oral immunomodulatory agents in the management of multiple sclerosis. Drug Des Devel Ther 2011; 5:255274.
  12. Consortium of Multiple Sclerosis Centers (CMSC). Phase II study with ocrelizumab shows significant reduction in disease activity. CMSC Web site. http://mscare.org/cmsc/index.php?option=com_content&task=view&id=1081&Itemid=1465. Published October 15, 2010. Accessed August 26, 2011.
  13. Tysabri [package insert]. South San Francisco, CA: Elan Pharmaceuticals, Inc.; 2011.
  14. Polman CH, O’Connor PW, Havrdova E, et al. A randomized, placebo-controlled trial of natalizumab for relapsing multiple sclerosis. N Engl J Med 2006; 354:899910.
  15. Miller DH, Khan OA, Sheremata WA, et al. A controlled trial of natalizumab for relapsing multiple sclerosis. N Engl J Med 2003; 348:1523.
  16. Miller DH, Soon D, Fernando KT, et al. MRI outcomes in a placebo-controlled trial of natalizumab in relapsing MS. Neurology 2007; 68:13901401.
  17. Havrdova E, Galetta S, Hutchinson M, et al. Effect of natalizumab on clinical and radiological disease activity in multiple sclerosis: a retrospective analysis of the Natalizumab Safety and Efficacy in Relapsing-Remitting Multiple Sclerosis (AFFIRM) study. Lancet Neurol 2009; 8:254260.
  18. Rudick RA, Miller D, Hass S, et al. Health-related quality of life in multiple sclerosis: effects of natalizumab. Ann Neurol 2007; 62:335346.
  19. Rudick RA, Stuart WH, Calabresi PA, et al. Natalizumab plus interferon beta-1a for relapsing multipls sclerosis. N Engl J Med 2006; 354:911923.
  20. Yousry TA, Major EO, Ryschkewitsch C, et al. Evaluation of patients treated with natalizumab for progressive multifocal leukoencephalopathy. N Engl J Med 2006; 354:924933.
  21. O’Connor PW, Goodman A, Kappos L, et al. Disease activity return during natalizumab treatment interruption in patients with multiple sclerosis [published online ahead of print May 4, 2011]. Neurology 2011; 76:18581865.
  22. Update on Tysabri and PML. National Multiple Sclerosis Society Web site. http://www.nationalmssociety.org/news/news-detail/index.aspx?nid=2308. Published April 11, 2011. Updated May 23, 2011. Accessed June 22, 2011.
  23. Vermersch P, Kappos L, Gold R, et al. Clinical outcomes of natalizumab-associated progressive multifocal leukoencephalopathy. Neurology 2011; 76:16971704.
  24. Heesen C, Kleiter I, Nguyen F, et al. Risk perception in natalizumab-treated multiple sclerosis patients and their neurologists. Mult Scler 2010; 16:15071512.
  25. Chen Y, Bord E, Tompkins T, et al. Asymptomatic reactivation of JC virus in patients treated with natalizumab. N Engl J Med 2009; 361:10671074.
  26. Jilek S, Jaquiery E, Hirsch HH, et al. Immune responses to JC virus in patients with multiple sclerosis treated with natalizumab: a cross-sectional and longitudinal study. Lancet Neurol 2010; 9:264272.
  27. Rudick RA, O’Connor PW, Polman CH, et al. Assessment of JC virus DNA in blood and urine from natalizumab-treated patients. Ann Neurol 2010; 68:304310.
  28. Gorelik L, Lerner M, Bixler S, et al. Anti-JC virus antibodies: implications for PML risk stratification. Ann Neurol 2010; 68:295303.
References
  1. Weinshenker BG, Bass B, Rice GPA, et al. The natural history of multiple sclerosis: a geographically based study. I. Clinical course and disability. Brain 1999; 112:133146.
  2. Jacobs LD, Cookfair DL, Rudick RA, et al. Intramuscular interferon beta-1a for disease progression in relapsing multiple sclerosis. Ann Neurol 1996; 39:285294.
  3. The IFNB Multiple Sclerosis Study Group. Interferon beta-1b is effective in relapsing-remitting multiple sclerosis I: clinical results of a multicenter, randomized, double-blind, placebo-controlled trial. Neurology 1993; 43:655661.
  4. IFNB Multiple Sclerosis Study Group, The University of British Columbia MS/MRI Analysis Group. Interferon beta-1b in the treatment of multiple sclerosis: final outcome of the randomized controlled trial. The IFNB Multiple Sclerosis Study Group and the University of British Columbia MS/MRI Analysis Group. Neurology 1995; 45:12771285.
  5. Johnson KP, Brooks BR, Cohen JA, et al. Copolymer 1 reduces relapse rate and improves disability in relapsing-remitting multiple sclerosis: results of a phase III multicenter, double-blind, placebo-controlled trial. Neurology 1995; 45:12681276.
  6. PRISMS (Prevention of Relapses and Disability by Interferon beta-1a Subcutaneously in Multiple Sclerosis) Study Group. Randomised double-blind placebo-controlled study of interferon beta-1a in relapsing/remitting multiple sclerosis. Lancet 1998; 352:14981504.
  7. Kappos L, Radue EW, O’Connor P, et al. A placebo-controlled trial of oral fingolimod in relapsing multiple sclerosis. N Engl J Med 2010; 362:387401.
  8. Cohen JA, Barkhof F, Comi G, et al. Oral fingolimod or intramuscular interferon for relapsing multiple sclerosis. N Engl J Med 2010; 362:402415.
  9. Gilenya [package insert]. East Hanover, NJ: Novartis Pharmaceuticals Corporation; 2011.
  10. Coles AJ, Fox E, Vladic A, et al. Alemtuzumab versus interferon beta-1a in early relapsing-remitting multiple sclerosis: post-hoc and subset analyses of clinical efficacy outcomes. Lancet Neurol 2011; 10:338348.
  11. Nicholas R, Giannetti P, Alsanousi A, Friede T, Muraro PA. Development of oral immunomodulatory agents in the management of multiple sclerosis. Drug Des Devel Ther 2011; 5:255274.
  12. Consortium of Multiple Sclerosis Centers (CMSC). Phase II study with ocrelizumab shows significant reduction in disease activity. CMSC Web site. http://mscare.org/cmsc/index.php?option=com_content&task=view&id=1081&Itemid=1465. Published October 15, 2010. Accessed August 26, 2011.
  13. Tysabri [package insert]. South San Francisco, CA: Elan Pharmaceuticals, Inc.; 2011.
  14. Polman CH, O’Connor PW, Havrdova E, et al. A randomized, placebo-controlled trial of natalizumab for relapsing multiple sclerosis. N Engl J Med 2006; 354:899910.
  15. Miller DH, Khan OA, Sheremata WA, et al. A controlled trial of natalizumab for relapsing multiple sclerosis. N Engl J Med 2003; 348:1523.
  16. Miller DH, Soon D, Fernando KT, et al. MRI outcomes in a placebo-controlled trial of natalizumab in relapsing MS. Neurology 2007; 68:13901401.
  17. Havrdova E, Galetta S, Hutchinson M, et al. Effect of natalizumab on clinical and radiological disease activity in multiple sclerosis: a retrospective analysis of the Natalizumab Safety and Efficacy in Relapsing-Remitting Multiple Sclerosis (AFFIRM) study. Lancet Neurol 2009; 8:254260.
  18. Rudick RA, Miller D, Hass S, et al. Health-related quality of life in multiple sclerosis: effects of natalizumab. Ann Neurol 2007; 62:335346.
  19. Rudick RA, Stuart WH, Calabresi PA, et al. Natalizumab plus interferon beta-1a for relapsing multipls sclerosis. N Engl J Med 2006; 354:911923.
  20. Yousry TA, Major EO, Ryschkewitsch C, et al. Evaluation of patients treated with natalizumab for progressive multifocal leukoencephalopathy. N Engl J Med 2006; 354:924933.
  21. O’Connor PW, Goodman A, Kappos L, et al. Disease activity return during natalizumab treatment interruption in patients with multiple sclerosis [published online ahead of print May 4, 2011]. Neurology 2011; 76:18581865.
  22. Update on Tysabri and PML. National Multiple Sclerosis Society Web site. http://www.nationalmssociety.org/news/news-detail/index.aspx?nid=2308. Published April 11, 2011. Updated May 23, 2011. Accessed June 22, 2011.
  23. Vermersch P, Kappos L, Gold R, et al. Clinical outcomes of natalizumab-associated progressive multifocal leukoencephalopathy. Neurology 2011; 76:16971704.
  24. Heesen C, Kleiter I, Nguyen F, et al. Risk perception in natalizumab-treated multiple sclerosis patients and their neurologists. Mult Scler 2010; 16:15071512.
  25. Chen Y, Bord E, Tompkins T, et al. Asymptomatic reactivation of JC virus in patients treated with natalizumab. N Engl J Med 2009; 361:10671074.
  26. Jilek S, Jaquiery E, Hirsch HH, et al. Immune responses to JC virus in patients with multiple sclerosis treated with natalizumab: a cross-sectional and longitudinal study. Lancet Neurol 2010; 9:264272.
  27. Rudick RA, O’Connor PW, Polman CH, et al. Assessment of JC virus DNA in blood and urine from natalizumab-treated patients. Ann Neurol 2010; 68:304310.
  28. Gorelik L, Lerner M, Bixler S, et al. Anti-JC virus antibodies: implications for PML risk stratification. Ann Neurol 2010; 68:295303.
Page Number
S18-S23
Page Number
S18-S23
Publications
Publications
Article Type
Display Headline
Multiple sclerosis, natalizumab, and PML: Helping patients decide
Display Headline
Multiple sclerosis, natalizumab, and PML: Helping patients decide
Citation Override
Cleveland Clinic Journal of Medicine 2011 November;78(suppl 2):S18-S23
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media

HIV-associated PML: Changing epidemiology and clinical approach

Article Type
Changed
Tue, 10/02/2018 - 09:25
Display Headline
HIV-associated PML: Changing epidemiology and clinical approach

The appearance of progressive multifocal leukoencephalopathy (PML) as a complication of human immunodeficiency virus (HIV) infection dates to shortly after the first description of acquired immunodeficiency syndrome (AIDS). The advent of highly active antiretroviral therapy (HAART) dramatically altered the nature of HIV infection, resulting in a substantial decline in mortality and, in essence, turning AIDS into a chronic disease. As patients lived longer with HIV infection, one consequence was an increased incidence of neurologic complications. By the early 1980s, AIDS was well recognized as an underlying disorder that predisposed to PML.

As many as 70% of HIV patients will eventually have involvement of either the peripheral or central nervous system (CNS). Most patients with HIV are managed by primary care clinicians, including those in the fields of family practice, internal medicine, or infectious disease, and the complexity of the neurologic disorders associated with HIV often results in either delayed diagnosis or misdiagnosis. For example, the evolution of HIV in the plasma, where most clinicians measure it, may differ from its evolution in the spinal fluid and brain. An emerging issue is that of hepatitis C coinfection, which may itself be associated with central and peripheral neurologic complications.

Treatment of HIV with antiretroviral agents has numerous neurologic implications. These include the potential ability of these agents to penetrate the blood-brain barrier, their efficacy in both treating and preventing cognitive impairment and other CNS disorders, and their toxic effects in the CNS and peripheral nervous system.

NEUROLOGIC COMPLICATIONS OF AIDS

Neurologic disease in AIDS patients can be classified in several ways. One of the most logical, particularly for primary care clinicians, is the separation of primary from secondary neurologic disorders:

  • Primary neurologic disorders are enigmatic and difficult to characterize; they include HIV-associated neurocognitive disorders in adults, encephalopathy in children, myelopathy or spinal cord disease, and peripheral neuropathy.
  • Secondary complications are related to progressive immunosuppression. These include opportunistic infections such as cytomegalovirus, toxoplasmosis, or cryptococcal meningitis; and neoplasms such as primary CNS lymphoma. Opportunistic infections and neoplasms have declined in incidence in the HAART era.

The prevalence of neuropathy as a neurologic complication of HIV/AIDS is as high as 57%, while PML affects 5% or fewer of HIV-infected patients (Table). PML in these patients is often associated with advanced disease, as reflected by low CD4+ cell counts and increased plasma HIV viral loads.

AT-RISK POOLS FOR PML

The AIDS epidemic significantly changed the epidemiology of PML, turning a formerly rare disease into a much more common one. In South Florida, the incidence of PML in patients with AIDS increased by 12 times from the 5-year period 1981 to 1984 compared with 1991 to 1994. Only two non-AIDS cases of PML were reported in South Florida during this 15-year period.1

At present, nonimmunosuppressed, healthy individuals account for fewer than 1% of all cases of PML. Non-HIV–related PML represents 10% to 20% of all PML cases. Cancer survivors and patients with rheumatoid arthritis who are treated with immunotherapy constitute the largest at-risk pools among this group. PML related to HIV represents 80% to 90% of PML cases, drawing from a pool of 1.2 million HIV-infected individuals in the United States.

UNIQUE PRESENTATION OF HIV-ASSOCIATED PML

The brain lesion in PML is classically a nonenhancing focal lesion, preferentially in white matter, but lesion characteristics often depart from this characteristic picture. For example, relatively faint contrast enhancement of lesions on magnetic resonance imaging has been observed, as well as involvement of white matter and gray matter. The distribution and character of brain lesions in PML may also differ from the classic picture. For example, the lesion may not be focal, particularly when PML is combined with the symmetric white matter abnormalities that are seen in HIV encephalopathy; this nonclassic presentation can cause difficulty in radiologic differentiation of PML and HIV encephalopathy.

Cerebellar degeneration

A unique presentation of PML is possible in HIV-infected patients. In 1998, Tagliati et al2 described a syndrome of degeneration of the cerebellum in 10 HIV-infected patients. One patient had JC virus (JCV) detected by polymerase chain reaction (PCR) in cerebellar biopsy tissue. The authors proposed the possibility of latent JCV infection of cerebellar granular cells in HIV-infected patients with cerebellar atrophy, lacking further evidence of other features of PML.

Figure. Magnetic resonance imaging demonstrates diffuse cerebellar atrophy (A, arrow) without areas of enhancement and white matter abnormalities (B, arrow) in a patient with JC virus–associated cerebellar degeneration.3
In 2005, Koralnik et al3 described a case of JCV granule cell neuronopathy in a patient with JCV-associated cerebellar degeneration. Clinical symptoms included gait and limb ataxia, dysmetria, dysarthria, and nystagmus. Magnetic resonance imaging demonstrated diffuse cerebellar atrophy and occasional subtle white matter abnormalities within the cerebellum (Figure). Immunohistochemistry showed preservation of Purkinje cells, and in situ PCR revealed selective depletion of cerebellar granule cells and JCV infection of granule cell neurons.

 

 

MANAGEMENT OF HIV-ASSOCIATED PML

Optimize HAART

A suppressed plasma HIV viral load is the strongest prognostic factor for an improved course in PML.4 In the pre-HAART era, the mean survival of HIV-associated PML was 3 to 6 months, with long-term survival estimated at 10%.5 The use of HAART has achieved a dramatic improvement in long-term survival, to upwards of 50%.6 Neurologic deficits are often irreversible even with HAART, but most HAART recipients show stability in neurologic status for years.

Other key characteristics associated with improved survival in HIV-associated PML appear to be younger age, PML as the heralding manifestation of AIDS, initiation of HAART upon diagnosis of PML, higher CD4 count, and absence of severe neurologic impairment.5–7

Investigational therapies

Specific antiviral drug regimens targeting JCV have been tested empirically in case studies and in clinical trials in patients with AIDS- and non–AIDS-related PML.

Cytosine arabinoside (Ara-C). Ara-C is a nucleoside analog used as an antineoplastic agent; it terminates chain elongation and inhibits DNA polymerase to confer antiviral activity. Ara-C decreased JCV replication in vitro.8 Based on anecdotal reports of efficacy in cancer-related cases of PML,9 Ara-C was tested in a multicenter trial of 57 patients with HIV and biopsy-confirmed PML.10 Neither intravenous nor intrathecal Ara-C combined with established antiviral therapy for AIDS improved the prognosis of these patients, and Ara-C has since been abandoned as a strategy to treat HIV-related PML.

Cidofovir. The noncyclic nucleoside phosphonate cidofovir garnered attention as a potential treatment for PML based on case reports of efficacy in HIV as well as non-HIV patients. Subsequently, a large multicenter study failed to detect any significant added benefit with cidofovir beyond that of HAART.11 Retrospective European studies confirmed the lack of clinical benefit with cidofovir.6,7,12

Interferon alfa. Case reports with interferon alfa-2a and -2b for the treatment of PML show conflicting results with respect to clinical response, symptomatic improvement, and survival, but toxicity has been substantial. In a series of 97 patients with AIDS-related PML, Geschwind et al determined that interferon alfa had no effect on survival beyond that of HAART.13

Mirtazapine. Serotonin receptor antagonists such as mirtazapine can block JCV entry into glial cells via serotonin 5-hydroxytryptamine receptors, providing a rationale for their use as a potential treatment for PML. Verma et al describe a case of clinical improvement (stable neurologic deficit) and PML lesion regression in a 63-year-old bedbound woman with polycythemia vera with biopsy-proven non–HIV-related PML that had progressed to quadriparesis.14

Mefloquine. The antimalarial drug mefloquine inhibits viral replication in cultured human glial cells and astrocytes, inhibits JC viral DNA replication, and showed efficacy against two JCV strains in cell culture.15 A randomized study to assess the effectiveness of mefloquine for treatment of PML has been completed and its results await publication.

SUMMARY

The incidence of PML has remained unchanged from the pre-HAART to the HAART era, but the prognosis is greatly improved. The clinical presentation of PML in AIDS patients may deviate from the classic triad of progressive, multifocal, white matter disease. It may be static and unifocal, and it may involve gray matter and neurons as well as white matter. The number of neurologic manifestations is vast and can include the cerebellar syndrome. Lumbar puncture with a PCR negative for JCV does not confirm the absence of PML.

The standard of care for HIV-associated PML is HAART, with the goal of achieving immunologic recovery and optimal virologic control. Whether therapeutic results obtained in patients with HIV-associated PML can be translated to the setting of non–HIV-associated PML is unclear.

DISCUSSION

Dr. Simpson: As a followup to the Ara-C trial that was published,10 PML confirmed by brain biopsy was one of the enrolling criteria, and the planned study population was 65 patients. Longitudinal examination of viral load in cerebrospinal fluid (CSF) was a part of the study, and we found that the lower the viral load, the better the prognosis. Fifty-two patients were enrolled before the trial was stopped because it was clear that Ara-C was not producing a benefit. The patients had multifocal disease but, because Ara-C does not effectively cross the blood-brain barrier, penetration in the brain was minimal even with the use of an intrathecal shunt in this study.

Dr. Major: Do you think viral load in CSF is a predictor of disease severity and outcome in PML?

Dr. Rudick: Generally speaking, that’s probably true. We have found, as have many of our colleagues who run a lot of CSF samples, that high viral loads are not a good thing.

Dr. Bennett: How is it that the incidence of PML has not changed from the pre-HAART to the post-HAART era? How do you account for this in terms of the change in patients’ T-cell function from pre- to post-HAART?

Dr. Simpson: I don’t know. Intuitively, why do patients treated with HAART, who are relatively immune reconstituted, develop PML? The problem is that not everyone is immune reconstituted. HAART fails in some patients. Further, PML remains a disease that is more common in late-stage HIV among patients with low CD4 counts and high viral loads, meaning that a large population of patients is available to develop this disease. With that said, it is perplexing that the incidence has not gone down more than it has.

Dr. Major: There’s a phenomenon called “unmasking PML with HAART,” in which individuals have no signs of PML upon initiation of HAART, but then very shortly after, PML is diagnosed.

Dr. Berger: You’re talking about PML immune reconstitution inflammatory syndrome (IRIS).

Dr. Major: IRIS can occur before PML, or PML and IRIS can be concurrent. In some patients, once the infection starts, it persists; this suggests that the virus is carried to the brain through the infected lymphocyte populations and may explain why the incidence of PML has not changed from the pre-HAART to the HAART era.

Dr. Calabrese: In patients with HIV who develop PML within the first 6 months of HAART, are we seeing the IRIS phenomenon or is it a presenting sign of advanced HIV?

Dr. Simpson: It’s well known that a number of opportunistic infections can develop in the setting of HAART. In fact, whether one should delay HAART when initiating therapy for opportunistic infections has been debated for just this reason. Most people presume IRIS to be a massive immunologic hit to all organ systems, as CD4 counts rise dramatically to produce hyperimmune-mediated phenomena such as Guillain-Barré syndrome. To what extent immunologic recovery is or is not linked to PML and why it happens are fascinating questions.

Dr. Berger: Opportunistic infections, PML among them, that occur following the initiation of HAART and recovery of the immune system are almost always an IRIS-mediated phenomenon in which the disease has been smoldering and then surfaces because of the release of an inflammatory response.

Dr. Calabrese: In patients with cerebellar degeneration, do you typically detect JCV in PCR in the spinal fluid?

Dr. Simpson: Not in the early stages, but in some patients with later-stage disease,3 the answer is yes. Certainly, PCR of CSF samples to look for JCV is the diagnostic test of choice. But in the early days, when we had no idea what caused this cerebellar syndrome, we were doing cerebellar biopsies.

References
  1. Berger JR, Pall L, Lanska D, Whiteman M. Progressive multifocal leukoencephalopathy in patients with HIV infection. J Neurovirol 1998; 4:5968.
  2. Tagliati M, Simpson D, Margello S, Clifford D, Schwartz RL, Berger JR. Cerebellar degeneration associated with human immunodeficiency virus infection. Neurology 1998; 50:244251.
  3. Koralnik I, Wüthrich C, Dang X, et al. JC virus granule cell neuronopathy: a novel clinical syndrome distinct from progressive multifocal leukoencephalopathy. Ann Neurol 2005; 57:576580.
  4. Clifford DB, Yiannoutsos C, Glicksman M, et al. HAART improves prognosis in HIV-associated progressive multifocal leukoencephalopathy. Neurology 1999; 52:623625.
  5. Berger JR, Levy RM, Flomenhoff D, Dobbs M. Predictive factors for prolonged survival in acquired immunodeficiency syndrome-associated progressive multifocal leukoencephalopathy. Ann Neurol 1998; 44:341349.
  6. Antinori A, Cingolani A, Lorenzini P, et al. Clinical epidemiology and survival of progressive multifocal leukoencephalopathy in the era of highly active antiretroviral therapy: data from the Italian Registry Investigative Neuro AIDS (IRINA). J Neurovirol 2003; 9( suppl 1):4753.
  7. Berenguer J, Miralles P, Arrizabalaga J, et al. Clinical course and prognostic factors of progressive multifocal leukoencephalopathy in patients treated with highly active antiretroviral therapy. Clin Infect Dis 2003; 36:10471052.
  8. Hou J, Major EO. The efficacy of nucleoside analogs against JC virus multiplication in a persistently infected human fetal brain cell line. J Neurovirol 1998; 4:451456.
  9. Aksamit A. Treatment of non-AIDS progressive multifocal leukoencephalopathy with cytosine arabinoside. J Neurovirol 2001; 7:386390.
  10. Hall C, Dafni U, Simpson D, et al. Failure of cytarabine in progressive multifocal leukoencephalopathy associated with human immunodeficiency virus infection. AIDS Clinical Trials Group 243 Team. N Engl J Med 1998; 338:13451351.
  11. Marra CM, Rajicic N, Barker DE, et al. A pilot study of cidofovir for progressive multifocal leukoencephalopathy in AIDS. AIDS 2002; 16:17911797.
  12. Gasnault J, Kousignian P, Kahraman M, et al. Cidofovir in AIDS-associated progressive multifocal leukoencephalopathy: a monocenter observational study with clinical and JC virus load monitoring. J Neurovirol 2001; 7:375381.
  13. Geschwind MD, Skolasky RI, Royal WS, McArthur JC. The relative contributions of HAART and alpha-interferon for therapy of progressive multifocal leukoencephalopathy in AIDS. J Neurovirol 2001; 7:353357.
  14. Verma S, Cikurel K, Koralnik IJ, et al. Mirtazapine in progressive multifocal leukoencephalopathy. J Infect Dis 2007; 196:709711.
  15. Brickelmaier M, Lugovskoy A, Kartikeyan R, et al. Identification and characterization of mefloquine efficacy against JC virus in vitro. Antimicrob Agents Chemother 2009; 53:18401849.
Article PDF
Author and Disclosure Information

David M. Simpson, MD, FRCP, FAAN
Professor of Neurology; Director, Neuro-AIDS Research Program; Director, Clinical Neurophysiology Laboratories; Mount Sinai Medical Center, New York, NY

Correspondence: David M. Simpson, MD, FRCP, FAAN, Annenberg Building, Floor 2, Box 1052, One Gustave Levy Place, New York, NY 10029; david.simpson@mssm.edu

Dr. Simpson reported consulting relationships with Biogen Idec and Pfizer Inc.

This article was developed from an audio transcript of Dr. Simpson’s presentation at a roundtable convened at Cleveland Clinic on January 31, 2011. The transcript was edited by the Cleveland Clinic Journal of Medicine staff for clarity and conciseness, and was then reviewed, revised, and approved by Dr. Simpson.

Publications
Page Number
S24-S27
Author and Disclosure Information

David M. Simpson, MD, FRCP, FAAN
Professor of Neurology; Director, Neuro-AIDS Research Program; Director, Clinical Neurophysiology Laboratories; Mount Sinai Medical Center, New York, NY

Correspondence: David M. Simpson, MD, FRCP, FAAN, Annenberg Building, Floor 2, Box 1052, One Gustave Levy Place, New York, NY 10029; david.simpson@mssm.edu

Dr. Simpson reported consulting relationships with Biogen Idec and Pfizer Inc.

This article was developed from an audio transcript of Dr. Simpson’s presentation at a roundtable convened at Cleveland Clinic on January 31, 2011. The transcript was edited by the Cleveland Clinic Journal of Medicine staff for clarity and conciseness, and was then reviewed, revised, and approved by Dr. Simpson.

Author and Disclosure Information

David M. Simpson, MD, FRCP, FAAN
Professor of Neurology; Director, Neuro-AIDS Research Program; Director, Clinical Neurophysiology Laboratories; Mount Sinai Medical Center, New York, NY

Correspondence: David M. Simpson, MD, FRCP, FAAN, Annenberg Building, Floor 2, Box 1052, One Gustave Levy Place, New York, NY 10029; david.simpson@mssm.edu

Dr. Simpson reported consulting relationships with Biogen Idec and Pfizer Inc.

This article was developed from an audio transcript of Dr. Simpson’s presentation at a roundtable convened at Cleveland Clinic on January 31, 2011. The transcript was edited by the Cleveland Clinic Journal of Medicine staff for clarity and conciseness, and was then reviewed, revised, and approved by Dr. Simpson.

Article PDF
Article PDF

The appearance of progressive multifocal leukoencephalopathy (PML) as a complication of human immunodeficiency virus (HIV) infection dates to shortly after the first description of acquired immunodeficiency syndrome (AIDS). The advent of highly active antiretroviral therapy (HAART) dramatically altered the nature of HIV infection, resulting in a substantial decline in mortality and, in essence, turning AIDS into a chronic disease. As patients lived longer with HIV infection, one consequence was an increased incidence of neurologic complications. By the early 1980s, AIDS was well recognized as an underlying disorder that predisposed to PML.

As many as 70% of HIV patients will eventually have involvement of either the peripheral or central nervous system (CNS). Most patients with HIV are managed by primary care clinicians, including those in the fields of family practice, internal medicine, or infectious disease, and the complexity of the neurologic disorders associated with HIV often results in either delayed diagnosis or misdiagnosis. For example, the evolution of HIV in the plasma, where most clinicians measure it, may differ from its evolution in the spinal fluid and brain. An emerging issue is that of hepatitis C coinfection, which may itself be associated with central and peripheral neurologic complications.

Treatment of HIV with antiretroviral agents has numerous neurologic implications. These include the potential ability of these agents to penetrate the blood-brain barrier, their efficacy in both treating and preventing cognitive impairment and other CNS disorders, and their toxic effects in the CNS and peripheral nervous system.

NEUROLOGIC COMPLICATIONS OF AIDS

Neurologic disease in AIDS patients can be classified in several ways. One of the most logical, particularly for primary care clinicians, is the separation of primary from secondary neurologic disorders:

  • Primary neurologic disorders are enigmatic and difficult to characterize; they include HIV-associated neurocognitive disorders in adults, encephalopathy in children, myelopathy or spinal cord disease, and peripheral neuropathy.
  • Secondary complications are related to progressive immunosuppression. These include opportunistic infections such as cytomegalovirus, toxoplasmosis, or cryptococcal meningitis; and neoplasms such as primary CNS lymphoma. Opportunistic infections and neoplasms have declined in incidence in the HAART era.

The prevalence of neuropathy as a neurologic complication of HIV/AIDS is as high as 57%, while PML affects 5% or fewer of HIV-infected patients (Table). PML in these patients is often associated with advanced disease, as reflected by low CD4+ cell counts and increased plasma HIV viral loads.

AT-RISK POOLS FOR PML

The AIDS epidemic significantly changed the epidemiology of PML, turning a formerly rare disease into a much more common one. In South Florida, the incidence of PML in patients with AIDS increased by 12 times from the 5-year period 1981 to 1984 compared with 1991 to 1994. Only two non-AIDS cases of PML were reported in South Florida during this 15-year period.1

At present, nonimmunosuppressed, healthy individuals account for fewer than 1% of all cases of PML. Non-HIV–related PML represents 10% to 20% of all PML cases. Cancer survivors and patients with rheumatoid arthritis who are treated with immunotherapy constitute the largest at-risk pools among this group. PML related to HIV represents 80% to 90% of PML cases, drawing from a pool of 1.2 million HIV-infected individuals in the United States.

UNIQUE PRESENTATION OF HIV-ASSOCIATED PML

The brain lesion in PML is classically a nonenhancing focal lesion, preferentially in white matter, but lesion characteristics often depart from this characteristic picture. For example, relatively faint contrast enhancement of lesions on magnetic resonance imaging has been observed, as well as involvement of white matter and gray matter. The distribution and character of brain lesions in PML may also differ from the classic picture. For example, the lesion may not be focal, particularly when PML is combined with the symmetric white matter abnormalities that are seen in HIV encephalopathy; this nonclassic presentation can cause difficulty in radiologic differentiation of PML and HIV encephalopathy.

Cerebellar degeneration

A unique presentation of PML is possible in HIV-infected patients. In 1998, Tagliati et al2 described a syndrome of degeneration of the cerebellum in 10 HIV-infected patients. One patient had JC virus (JCV) detected by polymerase chain reaction (PCR) in cerebellar biopsy tissue. The authors proposed the possibility of latent JCV infection of cerebellar granular cells in HIV-infected patients with cerebellar atrophy, lacking further evidence of other features of PML.

Figure. Magnetic resonance imaging demonstrates diffuse cerebellar atrophy (A, arrow) without areas of enhancement and white matter abnormalities (B, arrow) in a patient with JC virus–associated cerebellar degeneration.3
In 2005, Koralnik et al3 described a case of JCV granule cell neuronopathy in a patient with JCV-associated cerebellar degeneration. Clinical symptoms included gait and limb ataxia, dysmetria, dysarthria, and nystagmus. Magnetic resonance imaging demonstrated diffuse cerebellar atrophy and occasional subtle white matter abnormalities within the cerebellum (Figure). Immunohistochemistry showed preservation of Purkinje cells, and in situ PCR revealed selective depletion of cerebellar granule cells and JCV infection of granule cell neurons.

 

 

MANAGEMENT OF HIV-ASSOCIATED PML

Optimize HAART

A suppressed plasma HIV viral load is the strongest prognostic factor for an improved course in PML.4 In the pre-HAART era, the mean survival of HIV-associated PML was 3 to 6 months, with long-term survival estimated at 10%.5 The use of HAART has achieved a dramatic improvement in long-term survival, to upwards of 50%.6 Neurologic deficits are often irreversible even with HAART, but most HAART recipients show stability in neurologic status for years.

Other key characteristics associated with improved survival in HIV-associated PML appear to be younger age, PML as the heralding manifestation of AIDS, initiation of HAART upon diagnosis of PML, higher CD4 count, and absence of severe neurologic impairment.5–7

Investigational therapies

Specific antiviral drug regimens targeting JCV have been tested empirically in case studies and in clinical trials in patients with AIDS- and non–AIDS-related PML.

Cytosine arabinoside (Ara-C). Ara-C is a nucleoside analog used as an antineoplastic agent; it terminates chain elongation and inhibits DNA polymerase to confer antiviral activity. Ara-C decreased JCV replication in vitro.8 Based on anecdotal reports of efficacy in cancer-related cases of PML,9 Ara-C was tested in a multicenter trial of 57 patients with HIV and biopsy-confirmed PML.10 Neither intravenous nor intrathecal Ara-C combined with established antiviral therapy for AIDS improved the prognosis of these patients, and Ara-C has since been abandoned as a strategy to treat HIV-related PML.

Cidofovir. The noncyclic nucleoside phosphonate cidofovir garnered attention as a potential treatment for PML based on case reports of efficacy in HIV as well as non-HIV patients. Subsequently, a large multicenter study failed to detect any significant added benefit with cidofovir beyond that of HAART.11 Retrospective European studies confirmed the lack of clinical benefit with cidofovir.6,7,12

Interferon alfa. Case reports with interferon alfa-2a and -2b for the treatment of PML show conflicting results with respect to clinical response, symptomatic improvement, and survival, but toxicity has been substantial. In a series of 97 patients with AIDS-related PML, Geschwind et al determined that interferon alfa had no effect on survival beyond that of HAART.13

Mirtazapine. Serotonin receptor antagonists such as mirtazapine can block JCV entry into glial cells via serotonin 5-hydroxytryptamine receptors, providing a rationale for their use as a potential treatment for PML. Verma et al describe a case of clinical improvement (stable neurologic deficit) and PML lesion regression in a 63-year-old bedbound woman with polycythemia vera with biopsy-proven non–HIV-related PML that had progressed to quadriparesis.14

Mefloquine. The antimalarial drug mefloquine inhibits viral replication in cultured human glial cells and astrocytes, inhibits JC viral DNA replication, and showed efficacy against two JCV strains in cell culture.15 A randomized study to assess the effectiveness of mefloquine for treatment of PML has been completed and its results await publication.

SUMMARY

The incidence of PML has remained unchanged from the pre-HAART to the HAART era, but the prognosis is greatly improved. The clinical presentation of PML in AIDS patients may deviate from the classic triad of progressive, multifocal, white matter disease. It may be static and unifocal, and it may involve gray matter and neurons as well as white matter. The number of neurologic manifestations is vast and can include the cerebellar syndrome. Lumbar puncture with a PCR negative for JCV does not confirm the absence of PML.

The standard of care for HIV-associated PML is HAART, with the goal of achieving immunologic recovery and optimal virologic control. Whether therapeutic results obtained in patients with HIV-associated PML can be translated to the setting of non–HIV-associated PML is unclear.

DISCUSSION

Dr. Simpson: As a followup to the Ara-C trial that was published,10 PML confirmed by brain biopsy was one of the enrolling criteria, and the planned study population was 65 patients. Longitudinal examination of viral load in cerebrospinal fluid (CSF) was a part of the study, and we found that the lower the viral load, the better the prognosis. Fifty-two patients were enrolled before the trial was stopped because it was clear that Ara-C was not producing a benefit. The patients had multifocal disease but, because Ara-C does not effectively cross the blood-brain barrier, penetration in the brain was minimal even with the use of an intrathecal shunt in this study.

Dr. Major: Do you think viral load in CSF is a predictor of disease severity and outcome in PML?

Dr. Rudick: Generally speaking, that’s probably true. We have found, as have many of our colleagues who run a lot of CSF samples, that high viral loads are not a good thing.

Dr. Bennett: How is it that the incidence of PML has not changed from the pre-HAART to the post-HAART era? How do you account for this in terms of the change in patients’ T-cell function from pre- to post-HAART?

Dr. Simpson: I don’t know. Intuitively, why do patients treated with HAART, who are relatively immune reconstituted, develop PML? The problem is that not everyone is immune reconstituted. HAART fails in some patients. Further, PML remains a disease that is more common in late-stage HIV among patients with low CD4 counts and high viral loads, meaning that a large population of patients is available to develop this disease. With that said, it is perplexing that the incidence has not gone down more than it has.

Dr. Major: There’s a phenomenon called “unmasking PML with HAART,” in which individuals have no signs of PML upon initiation of HAART, but then very shortly after, PML is diagnosed.

Dr. Berger: You’re talking about PML immune reconstitution inflammatory syndrome (IRIS).

Dr. Major: IRIS can occur before PML, or PML and IRIS can be concurrent. In some patients, once the infection starts, it persists; this suggests that the virus is carried to the brain through the infected lymphocyte populations and may explain why the incidence of PML has not changed from the pre-HAART to the HAART era.

Dr. Calabrese: In patients with HIV who develop PML within the first 6 months of HAART, are we seeing the IRIS phenomenon or is it a presenting sign of advanced HIV?

Dr. Simpson: It’s well known that a number of opportunistic infections can develop in the setting of HAART. In fact, whether one should delay HAART when initiating therapy for opportunistic infections has been debated for just this reason. Most people presume IRIS to be a massive immunologic hit to all organ systems, as CD4 counts rise dramatically to produce hyperimmune-mediated phenomena such as Guillain-Barré syndrome. To what extent immunologic recovery is or is not linked to PML and why it happens are fascinating questions.

Dr. Berger: Opportunistic infections, PML among them, that occur following the initiation of HAART and recovery of the immune system are almost always an IRIS-mediated phenomenon in which the disease has been smoldering and then surfaces because of the release of an inflammatory response.

Dr. Calabrese: In patients with cerebellar degeneration, do you typically detect JCV in PCR in the spinal fluid?

Dr. Simpson: Not in the early stages, but in some patients with later-stage disease,3 the answer is yes. Certainly, PCR of CSF samples to look for JCV is the diagnostic test of choice. But in the early days, when we had no idea what caused this cerebellar syndrome, we were doing cerebellar biopsies.

The appearance of progressive multifocal leukoencephalopathy (PML) as a complication of human immunodeficiency virus (HIV) infection dates to shortly after the first description of acquired immunodeficiency syndrome (AIDS). The advent of highly active antiretroviral therapy (HAART) dramatically altered the nature of HIV infection, resulting in a substantial decline in mortality and, in essence, turning AIDS into a chronic disease. As patients lived longer with HIV infection, one consequence was an increased incidence of neurologic complications. By the early 1980s, AIDS was well recognized as an underlying disorder that predisposed to PML.

As many as 70% of HIV patients will eventually have involvement of either the peripheral or central nervous system (CNS). Most patients with HIV are managed by primary care clinicians, including those in the fields of family practice, internal medicine, or infectious disease, and the complexity of the neurologic disorders associated with HIV often results in either delayed diagnosis or misdiagnosis. For example, the evolution of HIV in the plasma, where most clinicians measure it, may differ from its evolution in the spinal fluid and brain. An emerging issue is that of hepatitis C coinfection, which may itself be associated with central and peripheral neurologic complications.

Treatment of HIV with antiretroviral agents has numerous neurologic implications. These include the potential ability of these agents to penetrate the blood-brain barrier, their efficacy in both treating and preventing cognitive impairment and other CNS disorders, and their toxic effects in the CNS and peripheral nervous system.

NEUROLOGIC COMPLICATIONS OF AIDS

Neurologic disease in AIDS patients can be classified in several ways. One of the most logical, particularly for primary care clinicians, is the separation of primary from secondary neurologic disorders:

  • Primary neurologic disorders are enigmatic and difficult to characterize; they include HIV-associated neurocognitive disorders in adults, encephalopathy in children, myelopathy or spinal cord disease, and peripheral neuropathy.
  • Secondary complications are related to progressive immunosuppression. These include opportunistic infections such as cytomegalovirus, toxoplasmosis, or cryptococcal meningitis; and neoplasms such as primary CNS lymphoma. Opportunistic infections and neoplasms have declined in incidence in the HAART era.

The prevalence of neuropathy as a neurologic complication of HIV/AIDS is as high as 57%, while PML affects 5% or fewer of HIV-infected patients (Table). PML in these patients is often associated with advanced disease, as reflected by low CD4+ cell counts and increased plasma HIV viral loads.

AT-RISK POOLS FOR PML

The AIDS epidemic significantly changed the epidemiology of PML, turning a formerly rare disease into a much more common one. In South Florida, the incidence of PML in patients with AIDS increased by 12 times from the 5-year period 1981 to 1984 compared with 1991 to 1994. Only two non-AIDS cases of PML were reported in South Florida during this 15-year period.1

At present, nonimmunosuppressed, healthy individuals account for fewer than 1% of all cases of PML. Non-HIV–related PML represents 10% to 20% of all PML cases. Cancer survivors and patients with rheumatoid arthritis who are treated with immunotherapy constitute the largest at-risk pools among this group. PML related to HIV represents 80% to 90% of PML cases, drawing from a pool of 1.2 million HIV-infected individuals in the United States.

UNIQUE PRESENTATION OF HIV-ASSOCIATED PML

The brain lesion in PML is classically a nonenhancing focal lesion, preferentially in white matter, but lesion characteristics often depart from this characteristic picture. For example, relatively faint contrast enhancement of lesions on magnetic resonance imaging has been observed, as well as involvement of white matter and gray matter. The distribution and character of brain lesions in PML may also differ from the classic picture. For example, the lesion may not be focal, particularly when PML is combined with the symmetric white matter abnormalities that are seen in HIV encephalopathy; this nonclassic presentation can cause difficulty in radiologic differentiation of PML and HIV encephalopathy.

Cerebellar degeneration

A unique presentation of PML is possible in HIV-infected patients. In 1998, Tagliati et al2 described a syndrome of degeneration of the cerebellum in 10 HIV-infected patients. One patient had JC virus (JCV) detected by polymerase chain reaction (PCR) in cerebellar biopsy tissue. The authors proposed the possibility of latent JCV infection of cerebellar granular cells in HIV-infected patients with cerebellar atrophy, lacking further evidence of other features of PML.

Figure. Magnetic resonance imaging demonstrates diffuse cerebellar atrophy (A, arrow) without areas of enhancement and white matter abnormalities (B, arrow) in a patient with JC virus–associated cerebellar degeneration.3
In 2005, Koralnik et al3 described a case of JCV granule cell neuronopathy in a patient with JCV-associated cerebellar degeneration. Clinical symptoms included gait and limb ataxia, dysmetria, dysarthria, and nystagmus. Magnetic resonance imaging demonstrated diffuse cerebellar atrophy and occasional subtle white matter abnormalities within the cerebellum (Figure). Immunohistochemistry showed preservation of Purkinje cells, and in situ PCR revealed selective depletion of cerebellar granule cells and JCV infection of granule cell neurons.

 

 

MANAGEMENT OF HIV-ASSOCIATED PML

Optimize HAART

A suppressed plasma HIV viral load is the strongest prognostic factor for an improved course in PML.4 In the pre-HAART era, the mean survival of HIV-associated PML was 3 to 6 months, with long-term survival estimated at 10%.5 The use of HAART has achieved a dramatic improvement in long-term survival, to upwards of 50%.6 Neurologic deficits are often irreversible even with HAART, but most HAART recipients show stability in neurologic status for years.

Other key characteristics associated with improved survival in HIV-associated PML appear to be younger age, PML as the heralding manifestation of AIDS, initiation of HAART upon diagnosis of PML, higher CD4 count, and absence of severe neurologic impairment.5–7

Investigational therapies

Specific antiviral drug regimens targeting JCV have been tested empirically in case studies and in clinical trials in patients with AIDS- and non–AIDS-related PML.

Cytosine arabinoside (Ara-C). Ara-C is a nucleoside analog used as an antineoplastic agent; it terminates chain elongation and inhibits DNA polymerase to confer antiviral activity. Ara-C decreased JCV replication in vitro.8 Based on anecdotal reports of efficacy in cancer-related cases of PML,9 Ara-C was tested in a multicenter trial of 57 patients with HIV and biopsy-confirmed PML.10 Neither intravenous nor intrathecal Ara-C combined with established antiviral therapy for AIDS improved the prognosis of these patients, and Ara-C has since been abandoned as a strategy to treat HIV-related PML.

Cidofovir. The noncyclic nucleoside phosphonate cidofovir garnered attention as a potential treatment for PML based on case reports of efficacy in HIV as well as non-HIV patients. Subsequently, a large multicenter study failed to detect any significant added benefit with cidofovir beyond that of HAART.11 Retrospective European studies confirmed the lack of clinical benefit with cidofovir.6,7,12

Interferon alfa. Case reports with interferon alfa-2a and -2b for the treatment of PML show conflicting results with respect to clinical response, symptomatic improvement, and survival, but toxicity has been substantial. In a series of 97 patients with AIDS-related PML, Geschwind et al determined that interferon alfa had no effect on survival beyond that of HAART.13

Mirtazapine. Serotonin receptor antagonists such as mirtazapine can block JCV entry into glial cells via serotonin 5-hydroxytryptamine receptors, providing a rationale for their use as a potential treatment for PML. Verma et al describe a case of clinical improvement (stable neurologic deficit) and PML lesion regression in a 63-year-old bedbound woman with polycythemia vera with biopsy-proven non–HIV-related PML that had progressed to quadriparesis.14

Mefloquine. The antimalarial drug mefloquine inhibits viral replication in cultured human glial cells and astrocytes, inhibits JC viral DNA replication, and showed efficacy against two JCV strains in cell culture.15 A randomized study to assess the effectiveness of mefloquine for treatment of PML has been completed and its results await publication.

SUMMARY

The incidence of PML has remained unchanged from the pre-HAART to the HAART era, but the prognosis is greatly improved. The clinical presentation of PML in AIDS patients may deviate from the classic triad of progressive, multifocal, white matter disease. It may be static and unifocal, and it may involve gray matter and neurons as well as white matter. The number of neurologic manifestations is vast and can include the cerebellar syndrome. Lumbar puncture with a PCR negative for JCV does not confirm the absence of PML.

The standard of care for HIV-associated PML is HAART, with the goal of achieving immunologic recovery and optimal virologic control. Whether therapeutic results obtained in patients with HIV-associated PML can be translated to the setting of non–HIV-associated PML is unclear.

DISCUSSION

Dr. Simpson: As a followup to the Ara-C trial that was published,10 PML confirmed by brain biopsy was one of the enrolling criteria, and the planned study population was 65 patients. Longitudinal examination of viral load in cerebrospinal fluid (CSF) was a part of the study, and we found that the lower the viral load, the better the prognosis. Fifty-two patients were enrolled before the trial was stopped because it was clear that Ara-C was not producing a benefit. The patients had multifocal disease but, because Ara-C does not effectively cross the blood-brain barrier, penetration in the brain was minimal even with the use of an intrathecal shunt in this study.

Dr. Major: Do you think viral load in CSF is a predictor of disease severity and outcome in PML?

Dr. Rudick: Generally speaking, that’s probably true. We have found, as have many of our colleagues who run a lot of CSF samples, that high viral loads are not a good thing.

Dr. Bennett: How is it that the incidence of PML has not changed from the pre-HAART to the post-HAART era? How do you account for this in terms of the change in patients’ T-cell function from pre- to post-HAART?

Dr. Simpson: I don’t know. Intuitively, why do patients treated with HAART, who are relatively immune reconstituted, develop PML? The problem is that not everyone is immune reconstituted. HAART fails in some patients. Further, PML remains a disease that is more common in late-stage HIV among patients with low CD4 counts and high viral loads, meaning that a large population of patients is available to develop this disease. With that said, it is perplexing that the incidence has not gone down more than it has.

Dr. Major: There’s a phenomenon called “unmasking PML with HAART,” in which individuals have no signs of PML upon initiation of HAART, but then very shortly after, PML is diagnosed.

Dr. Berger: You’re talking about PML immune reconstitution inflammatory syndrome (IRIS).

Dr. Major: IRIS can occur before PML, or PML and IRIS can be concurrent. In some patients, once the infection starts, it persists; this suggests that the virus is carried to the brain through the infected lymphocyte populations and may explain why the incidence of PML has not changed from the pre-HAART to the HAART era.

Dr. Calabrese: In patients with HIV who develop PML within the first 6 months of HAART, are we seeing the IRIS phenomenon or is it a presenting sign of advanced HIV?

Dr. Simpson: It’s well known that a number of opportunistic infections can develop in the setting of HAART. In fact, whether one should delay HAART when initiating therapy for opportunistic infections has been debated for just this reason. Most people presume IRIS to be a massive immunologic hit to all organ systems, as CD4 counts rise dramatically to produce hyperimmune-mediated phenomena such as Guillain-Barré syndrome. To what extent immunologic recovery is or is not linked to PML and why it happens are fascinating questions.

Dr. Berger: Opportunistic infections, PML among them, that occur following the initiation of HAART and recovery of the immune system are almost always an IRIS-mediated phenomenon in which the disease has been smoldering and then surfaces because of the release of an inflammatory response.

Dr. Calabrese: In patients with cerebellar degeneration, do you typically detect JCV in PCR in the spinal fluid?

Dr. Simpson: Not in the early stages, but in some patients with later-stage disease,3 the answer is yes. Certainly, PCR of CSF samples to look for JCV is the diagnostic test of choice. But in the early days, when we had no idea what caused this cerebellar syndrome, we were doing cerebellar biopsies.

References
  1. Berger JR, Pall L, Lanska D, Whiteman M. Progressive multifocal leukoencephalopathy in patients with HIV infection. J Neurovirol 1998; 4:5968.
  2. Tagliati M, Simpson D, Margello S, Clifford D, Schwartz RL, Berger JR. Cerebellar degeneration associated with human immunodeficiency virus infection. Neurology 1998; 50:244251.
  3. Koralnik I, Wüthrich C, Dang X, et al. JC virus granule cell neuronopathy: a novel clinical syndrome distinct from progressive multifocal leukoencephalopathy. Ann Neurol 2005; 57:576580.
  4. Clifford DB, Yiannoutsos C, Glicksman M, et al. HAART improves prognosis in HIV-associated progressive multifocal leukoencephalopathy. Neurology 1999; 52:623625.
  5. Berger JR, Levy RM, Flomenhoff D, Dobbs M. Predictive factors for prolonged survival in acquired immunodeficiency syndrome-associated progressive multifocal leukoencephalopathy. Ann Neurol 1998; 44:341349.
  6. Antinori A, Cingolani A, Lorenzini P, et al. Clinical epidemiology and survival of progressive multifocal leukoencephalopathy in the era of highly active antiretroviral therapy: data from the Italian Registry Investigative Neuro AIDS (IRINA). J Neurovirol 2003; 9( suppl 1):4753.
  7. Berenguer J, Miralles P, Arrizabalaga J, et al. Clinical course and prognostic factors of progressive multifocal leukoencephalopathy in patients treated with highly active antiretroviral therapy. Clin Infect Dis 2003; 36:10471052.
  8. Hou J, Major EO. The efficacy of nucleoside analogs against JC virus multiplication in a persistently infected human fetal brain cell line. J Neurovirol 1998; 4:451456.
  9. Aksamit A. Treatment of non-AIDS progressive multifocal leukoencephalopathy with cytosine arabinoside. J Neurovirol 2001; 7:386390.
  10. Hall C, Dafni U, Simpson D, et al. Failure of cytarabine in progressive multifocal leukoencephalopathy associated with human immunodeficiency virus infection. AIDS Clinical Trials Group 243 Team. N Engl J Med 1998; 338:13451351.
  11. Marra CM, Rajicic N, Barker DE, et al. A pilot study of cidofovir for progressive multifocal leukoencephalopathy in AIDS. AIDS 2002; 16:17911797.
  12. Gasnault J, Kousignian P, Kahraman M, et al. Cidofovir in AIDS-associated progressive multifocal leukoencephalopathy: a monocenter observational study with clinical and JC virus load monitoring. J Neurovirol 2001; 7:375381.
  13. Geschwind MD, Skolasky RI, Royal WS, McArthur JC. The relative contributions of HAART and alpha-interferon for therapy of progressive multifocal leukoencephalopathy in AIDS. J Neurovirol 2001; 7:353357.
  14. Verma S, Cikurel K, Koralnik IJ, et al. Mirtazapine in progressive multifocal leukoencephalopathy. J Infect Dis 2007; 196:709711.
  15. Brickelmaier M, Lugovskoy A, Kartikeyan R, et al. Identification and characterization of mefloquine efficacy against JC virus in vitro. Antimicrob Agents Chemother 2009; 53:18401849.
References
  1. Berger JR, Pall L, Lanska D, Whiteman M. Progressive multifocal leukoencephalopathy in patients with HIV infection. J Neurovirol 1998; 4:5968.
  2. Tagliati M, Simpson D, Margello S, Clifford D, Schwartz RL, Berger JR. Cerebellar degeneration associated with human immunodeficiency virus infection. Neurology 1998; 50:244251.
  3. Koralnik I, Wüthrich C, Dang X, et al. JC virus granule cell neuronopathy: a novel clinical syndrome distinct from progressive multifocal leukoencephalopathy. Ann Neurol 2005; 57:576580.
  4. Clifford DB, Yiannoutsos C, Glicksman M, et al. HAART improves prognosis in HIV-associated progressive multifocal leukoencephalopathy. Neurology 1999; 52:623625.
  5. Berger JR, Levy RM, Flomenhoff D, Dobbs M. Predictive factors for prolonged survival in acquired immunodeficiency syndrome-associated progressive multifocal leukoencephalopathy. Ann Neurol 1998; 44:341349.
  6. Antinori A, Cingolani A, Lorenzini P, et al. Clinical epidemiology and survival of progressive multifocal leukoencephalopathy in the era of highly active antiretroviral therapy: data from the Italian Registry Investigative Neuro AIDS (IRINA). J Neurovirol 2003; 9( suppl 1):4753.
  7. Berenguer J, Miralles P, Arrizabalaga J, et al. Clinical course and prognostic factors of progressive multifocal leukoencephalopathy in patients treated with highly active antiretroviral therapy. Clin Infect Dis 2003; 36:10471052.
  8. Hou J, Major EO. The efficacy of nucleoside analogs against JC virus multiplication in a persistently infected human fetal brain cell line. J Neurovirol 1998; 4:451456.
  9. Aksamit A. Treatment of non-AIDS progressive multifocal leukoencephalopathy with cytosine arabinoside. J Neurovirol 2001; 7:386390.
  10. Hall C, Dafni U, Simpson D, et al. Failure of cytarabine in progressive multifocal leukoencephalopathy associated with human immunodeficiency virus infection. AIDS Clinical Trials Group 243 Team. N Engl J Med 1998; 338:13451351.
  11. Marra CM, Rajicic N, Barker DE, et al. A pilot study of cidofovir for progressive multifocal leukoencephalopathy in AIDS. AIDS 2002; 16:17911797.
  12. Gasnault J, Kousignian P, Kahraman M, et al. Cidofovir in AIDS-associated progressive multifocal leukoencephalopathy: a monocenter observational study with clinical and JC virus load monitoring. J Neurovirol 2001; 7:375381.
  13. Geschwind MD, Skolasky RI, Royal WS, McArthur JC. The relative contributions of HAART and alpha-interferon for therapy of progressive multifocal leukoencephalopathy in AIDS. J Neurovirol 2001; 7:353357.
  14. Verma S, Cikurel K, Koralnik IJ, et al. Mirtazapine in progressive multifocal leukoencephalopathy. J Infect Dis 2007; 196:709711.
  15. Brickelmaier M, Lugovskoy A, Kartikeyan R, et al. Identification and characterization of mefloquine efficacy against JC virus in vitro. Antimicrob Agents Chemother 2009; 53:18401849.
Page Number
S24-S27
Page Number
S24-S27
Publications
Publications
Article Type
Display Headline
HIV-associated PML: Changing epidemiology and clinical approach
Display Headline
HIV-associated PML: Changing epidemiology and clinical approach
Citation Override
Cleveland Clinic Journal of Medicine 2011 November;78(suppl 2):S24-S27
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media

PML and rheumatology: The contribution of disease and drugs

Article Type
Changed
Tue, 10/02/2018 - 09:25
Display Headline
PML and rheumatology: The contribution of disease and drugs

Progressive multifocal leukoencephalopathy (PML) is a rare, typically fatal, opportunistic infection caused by the JC virus (JCV). Formerly an example of neurologic arcana, PML became an important clinical concern when it developed in patients with human immunodeficiency virus (HIV) infection. More recently, PML has attracted the attention of rheumatologists following reports of its being associated with the use of targeted therapies such as natalizumab and rituximab.1

A recent survey of rheumatologists’ knowledge of and attitudes towards PML revealed that concerns over PML affect decisions on the use of biologic agents. Further, rheumatologists have important real and perceived learning gaps regarding PML; for example, 41% of those surveyed could not identify the diagnostic test of choice for PML.2

PML IN RHEUMATIC DISEASES

The US Food and Drug Administration (FDA) issued an alert in December 2006 following documentation of two fatal cases of PML in patients with systemic lupus erythematosus (SLE), both of whom had been treated with rituximab.3 We subsequently performed a literature search to identify cases of PML associated with rheumatic diseases.1,4 Patients were included only if the information provided was sufficient to substantiate the diagnosis of PML and the rheumatic disease in question; patients were excluded if they had HIV or cancer or had undergone organ transplantation. The search revealed 50 patients with rheumatic diseases who had PML (Table 1); SLE was overrepresented (n = 32) despite a much lower population prevalence of SLE compared with rheumatoid arthritis.

Examination of the immunosuppressive therapies prescribed to these patients within 6 months of the onset of neurologic symptoms attributed to PML revealed that low-dose (≤ 15 mg/d) prednisone, with or without an antimalarial agent, was the only immunosuppressive therapy in 31% of patients with SLE and in 11% of patients with rheumatic diseases other than SLE. Three patients had no documented immunosuppressive therapy in the 6 months prior to the onset of PML. Two patients with SLE were prescribed rituximab; no cases were reported in association with biologic therapies other than rituximab.4

In order to circumvent reporting bias, a nationwide hospital discharge database representing nearly 300 million patient discharges was used to determine the relative frequency of PML in patients with rheumatic diseases.5 Because of the reliance on diagnostic coding, rheumatic diseases were likely underreported in this sample; information on therapies was unavailable. After excluding patients who had HIV or cancer or were organ transplant recipients, four cases of PML were identified per 100,000 SLE discharges. This rate was 10-fold higher than the rate associated with rheumatoid arthritis and 20-fold higher than that of the background population.

These data show that PML is a rare occurrence in patients with rheumatic diseases, and SLE appears to be associated with a predisposition to PML. This predisposition in patients with SLE does not appear to be proportional to the degree of iatrogenic immunosuppression, emphasizing the role of host factors.

DISEASE-MODIFYING DRUGS AND PML RISK

In addition to certain disease states, disease-modifying biologic drugs have recently been associated with rare instances of PML.

Rituximab

The first case of rituximab-associated PML in the setting of rheumatoid arthritis was recorded in September 2008.6 The patient had longstanding rheumatoid arthritis and Sjögren syndrome. She received four courses of rituximab and was diagnosed with PML 18 months after the last dose; she died 1 month later. Her therapy for rheumatoid arthritis included a tumor necrosis factor (TNF) antagonist prior to rituximab initiation and treatment with methotrexate and steroids before, during, and after rituximab therapy. Oropharyngeal cancer developed in this patient 9 months prior to the onset of PML and was treated with chemotherapy and radiation therapy.

Another case of PML in a patient with rheumatic disease who had been treated with rituximab was notable because it was the first in which the patient had not previously been treated with an anti-TNF agent.7

Ascertaining cause of PML in patients treated with rituximab is difficult because the potential pathogenic mechanism remains unknown. Humoral immunity is not protective against PML, leading to speculation that the loss of other B-cell functions, such as those of antigen-presenting cells or cytokine production, may lead to a defect in cell-mediated immunity. Another theory posits that reconstitution of naïve B cells with latent JCV infection following B-cell depletion from rituximab therapy may somehow facilitate the development of PML.

 

 

Efalizumab

Efalizumab is a monoclonal antibody that targets CD11a, the alpha subunit of lymphocyte function–associated antigen 1. Efalizumab blocks binding to intercellular adhesion molecule 1, and thereby blocks T-cell adhesion and migration. CD11a is also expressed on a variety of other leukocytes and lymphocytes such as B cells, monocytes, and natural killer cells.

Efalizumab was approved in 2003 by the FDA for the treatment of moderate to severe plaque psoriasis. It is estimated that 46,000 patients have been treated with efalizumab worldwide since its approval. In 2008, a black box warning was added to the efalizumab prescribing information following the occurrence of serious infections, including pulmonary tuberculosis, necrotizing fasciitis, and invasive fungal infections.8 Subsequently, four cases of PML, three of which were fatal, were reported in psoriasis patients treated with efalizumab. Of note, these were the first cases of PML reported in patients with psoriasis. Of more concern, the affected patients were among a group of approximately 1,100 patients who had been treated with efalizumab for more than 3 years. In February 2009, a public health advisory was issued by the FDA,9 and efalizumab was voluntarily withdrawn by its manufacturer 2 months later.

Belatacept

Belatacept is a recombinant soluble fusion protein of the extracellular domain of human cytotoxic T-lymphocyte antigen-4 with a fragment of a modified Fc domain of immunoglobulin G1. Recently approved by the FDA for prophylaxis of renal transplant rejection, it is a second-generation, higher-avidity version of abatacept. Abatacept is licensed for the treatment of rheumatoid arthritis and is under investigation for the treatment of vasculitis and SLE. Belatacept differs from abatacept by only two amino acids.

Two cases of PML have been reported in association with belatacept, one in a patient following renal transplantation and the other in a patient following liver transplantation. Both patients had been treated with other standard immunosuppressive therapies for prophylaxis of organ transplant rejection, including mycophenolate mofetil.

Mycophenolate mofetil

Mycophenolate mofetil is the prodrug of mycophenolic acid. Both have been the subjects of FDA alerts regarding PML, based on a January 2008 report of 10 definite and 7 possible cases of PML occurring with mycophenolate mofetil. The patients affected included four with SLE, none of whom underwent a renal transplant.10

In a retrospective cohort study of 32,757 renal transplant patients, Neff et al11 found 14 cases of PML per 100,000 person-years among patients treated with mycophenolate mofetil following kidney transplant compared with none in patients who did not receive mycophenolate mofetil. It is difficult to ascertain risk with mycophenolate mofetil because it is standard therapy among renal transplant patients, leaving few patients in these groups unexposed.

Given the FDA alert with respect to mycophenolate mofetil and PML,10 the frequent use of mycophenolate mofetil in the setting of SLE, and the concerns about possible predisposition to PML among patients with SLE, it will be important to clarify the level of risk in patients with SLE who are treated with mycophenolate mofetil.

AGGREGATE EXPERIENCE: REVIEW OF FEDERAL DATABASE

We examined the aggregate experience of PML in association with autoimmune disorders and biologic disease-modifying antirheumatic drug (DMARD) exposures reported in the FDA Adverse Event Reporting System (AERS) database.12 A total of 19 confirmed cases of PML in patients with rheumatic diseases were uncovered: 10 in patients with SLE, 5 in patients with rheumatoid arthritis, 3 in patients with vasculitis, and 1 in a patient with dermatomyositis. The patients with PML included six who received rituximab for the management of rheumatic diseases (Table 2). In all six patients, rituximab was the most recently prescribed DMARD. Four cases were identified in patients treated with anti-TNF therapy, but three of these had received anti-TNF therapy prior to rituximab, and the other was receiving concomitant cyclophosphamide for rheumatoid vasculitis.

Ten cases of PML were confirmed with cyclophosphamide treatment, and cyclophosphamide was the most recent DMARD prescribed in two of these cases. Five cases were confirmed with mycophenolate mofetil (in four of which it was the most recently prescribed DMARD) and six with azathioprine (in three of which it was the most recently prescribed DMARD).

Risk of PML with DMARD therapy

Rituximab. The confirmation of six cases of PML among rituximab-treated rheumatoid arthritis patients is a source of concern. Nevertheless, PML is a rare adverse event. It occurs in fewer than 1 in 10,000 rituximab-treated patients who have rheumatoid arthritis, among a total of approximately 130,000 such patients. A better understanding of the potential mechanism responsible for the increased risk of developing PML may help in risk prediction and to guide patient selection for this agent.

Anti-TNF therapy. A paucity of confirmed cases in patients treated with anti-TNF therapy argues against a significant risk of PML associated with this therapy, especially considering the estimated 2 to 3 million rheumatoid arthritis patients who are receiving treatment with anti-TNF agents. A note of caution is sounded by a recent case report of PML in a rheumatoid arthritis patient. The patient had been treated with infliximab, with the only background therapy being methotrexate.13 Ongoing vigilance is therefore necessary.

Mycophenolate mofetil. All five confirmed cases of PML in mycophenolate mofetil-treated patients had earlier received treatment with cyclophosphamide. These data indicate no clear signal of excess risk with mycophenolate mofetil above that seen with other nonbiologic immunosuppressive agents, such as cyclophosphamide or azathioprine.

CONCLUSION AND RECOMMENDATIONS

PML has been reported in association with a variety of disease states, although a predisposition in patients with SLE has become apparent. Synthetic and biologic immunosuppressive therapies have also been implicated, but PML may also occur in the setting of minimal iatrogenic immunosuppression.

Until greater clarity can be achieved, all patients with systemic rheumatic diseases should be considered at risk for PML, regardless of the nature or intensity of their immunosuppressive therapy. In this context, differentiating PML from neurologic syndromes related to the underlying rheumatic disease (eg, neuropsychiatric SLE, cerebral vasculitis) is critical, particularly given the markedly different approaches to management.

PML should be considered in patients with unexplained subacute progressive focal and diffuse neurologic deficits, especially if their clinical or radiologic status worsens in the face of increased intensity of immunosuppressive therapy. Spinal cord or optic nerve involvement argues against PML. A normal magnetic resonance image (MRI) has a high negative predictive value, and frank infarction is not a feature of PML. In classic PML, contrast enhancement is typically absent and routine cerebrospinal fluid (CSF) analysis is typically normal. However, contrast enhancement and edema on MRI, lymphocytic CSF pleocytosis, and elevated CSF protein may be seen in the more recently described “inflammatory PML,” in which case the distinction from cerebral vasculitis or neuropsychiatric SLE may be more difficult. Angiography appears normal in patients with PML.

The diagnostic test of choice is a polymerase chain reaction (PCR) assay for JCV in CSF. If the PCR is repeatedly negative, then a brain biopsy should be considered, especially in the setting of progressive neurologic decline in patients receiving immunosuppressive therapy.

 

 

DISCUSSION

Dr. Simpson: To what extent are these lesions in the brain being attributed to the underlying vasculitis, particularly in SLE, as opposed to pursuing a PML diagnosis, and how might this result in dramatic underreporting of the complication?

Dr. Molloy: We found that PML is almost certainly underdiagnosed, particularly in SLE patients. If a patient succumbs to assumed neuropsychiatric SLE, how often is an autopsy undertaken? One telling paper from Sweden documented four cases of PML in SLE patients.14 In one of these, the diagnosis was made retrospectively from autopsy tissue that had been banked 20 years previously. It undoubtedly is underdiagnosed.

Dr. Calabrese: Even in the most recent rituximab-associated cases of PML, several patients were empirically given additional immunosuppressive therapy because it was presumed that they had a comorbid neuropsychiatric rheumatic complication. The presence of neuropsychiatric complications ascribed to an autoinflammatory disease generally warrants escalation of immunosuppressive therapy. It has always been standard practice for us to rule out opportunistic infection, but JCV infection has not been on the radar screen until very recently.

Dr. Molloy: I’d like to emphasize that, in our literature review, 50% of the rheumatic disease patients diagnosed with PML had been treated with more intensive immunosuppressive therapy. It was only after they continued to deteriorate that JCV infection was suspected and PML ultimately diagnosed.

Dr. Berger: Is it fair to say that the incidence of PML in SLE is about 10 times that in rheumatoid arthritis?

Dr. Molloy: In the hospital discharge database, it was 10-fold higher in SLE than in rheumatoid arthritis, but we can’t draw a conclusion from the AERS database because we don’t have a denominator. The database consists of voluntary submission of cases.

Dr. Calabrese: The information that we can expect to glean from the database is profoundly limited, for all the reasons that you enumerated. Despite the flaws, we’re obligated to continuously examine it because sometimes a case or two may provide some special insight.

Dr. Simpson: As neurologists, we often lag behind rheumatologists in the use of new treatments, including intravenous immune globulin (IVIG) and now rituximab. Rituximab is becoming the go-to drug for a number of neurologic diseases. I’m using it quite a bit and have observed some dramatic responses in patients with chronic inflammatory demyelinating polyneuropathy, for example, in whom IVIG or plasmapheresis was failing. Anecdotally, some of the turnarounds in polymyositis and even myasthenia gravis are remarkable as well. I’m not sure to what extent neurologists—particularly peripheral neurologists—who use rituximab are recognizing PML.

Dr. Fox: The index of suspicion is probably vastly different among multiple sclerosis (MS) specialists and general neurologists. Neurologists who treat MS will be acutely aware of PML because of its association with natalizumab.

Dr. Berger: Yes, but you’re talking about possibly two orders of magnitude difference between natalizumab and rituximab. In fact, PML is rarely reported in the setting of neurologic disease. It’s mostly reported in the setting of rheumatologic disease.

Dr. Rudick: I don’t necessarily agree with you. Ascertaining the true incidence of PML with agents other than natalizumab is difficult. One is unlikely to miss a case of PML in an MS patient treated with natalizumab, but most cases stemming from the use of these other disease-modifying drugs are probably being missed.

Dr. Calabrese: I get two messages out of this body of work. Number one is that while PML is rare, it is seen across the spectrum of immunosuppressive agents, including biologic and nonbiologic drugs. Number two is that PML is seriously underreported and underrecognized, which is probably leading to suboptimal patient care. Rituximab was recently approved for treatment of Wegener granulomatosis, and this disease is heavily pretreated with cyclophosphamide. You would expect that PML is on the radar among clinicians caring for patients whose diseases warrant the use of increasingly complex, potent, and novel immunosuppressives.

Dr. Berger: There is one other biologic agent you left out—alemtuzumab. It wipes out all of the B cells and T cells; the B cells repopulate but the T cells remain suppressed for a long period. If ever there was a drug whose action mirrors what happens in HIV, alemtuzumab is that drug. Yet, PML is rarely seen with alemtuzumab. Alemtuzumab-associated PML has not been reported in the MS population, and it has only been seen in two transplantations that I’m aware of. I’m not saying that it doesn’t occur, but we’re not seeing it with the same frequency that one would predict given its profile.

References
  1. Calabrese LH, Molloy ES, Huang D, Ransohoff RM. Progressive multifocal leukoencephalopathy in rheumatic diseases: evolving clinical and pathologic patterns of disease. Arthritis Rheum 2007; 6:21162128.
  2. Calabrese LH, Molloy ES, Taege AJ. Rheumatologists’ knowledge, attitudes and concerns regarding progressive multifocal encephalopathy (PML) [abstract]. Arthritis Rheum 2009; 60 (suppl 10):130.
  3. Rituxan (rituximab). U.S. Food and Drug Administration Web site. http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsforHumanMedicalProducts/ucm150747.htm. Updated June 19, 2009. Accessed September 15, 2011.
  4. Molloy ES, Calabrese LH. Progressive multifocal leukoencephalopathy in patients with rheumatic diseases: are patients with systemic lupus erythematosus at particular risk? Autoimmun Rev 2008; 8:144146.
  5. Molloy ES, Calabrese LH. Progressive multifocal leukoencephalopathy: a national estimate of frequency in systemic lupus erythematosus and other rheumatic diseases. Arthritis Rheum 2009; 60:37613765.
  6. Fleischmann RM. Progressive multifocal leukoencephalopathy following rituximab treatment in a patient with rheumatoid arthritis. Arthritis Rheum 2009; 60:32253228.
  7. Rituxan (rituximab) – PML. U.S. Food and Drug Administration Web site. http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsforHumanMedicalProducts/ucm187791.htm. Updated October 23, 2009. Accessed September 15, 2011.
  8. Raptiva (efalizumab) October 2008. U.S. Food and Drug Administration Web site. http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsforHumanMedicalProducts/ucm092089.htm. Updated June 19, 2009. Accessed September 15, 2011.
  9. Raptiva (efalizumab) Feb 2009. U.S. Food and Drug Administration Web site. http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsforHumanMedicalProducts/ucm149675.htm. Updated June 16, 2009. Accessed September 15, 2011.
  10. Communication about an ongoing safety review of CellCept (mycophenolate mofetil) and Myfortic (mycophenolic acid). U.S. Food and Drug Administration Web site. http://www.fda.gov/Drugs/DrugSafety/PostmarketDrugSafetyInformationforPatientsandProviders/DrugSafetyInformationforHeathcareProfessionals/ucm072438.htm. Updated October 30, 2009. Accessed September 15, 2011.
  11. Neff RT, Hurst FP, Falta EM, et al. Progressive multifocal leukoencephalopathy and use of mycophenolate mofetil after kidney transplantation. Transplantation 2008; 86:14741478.
  12. Molloy ES, Calabrese LH. Progressive multifocal leukoencephalopathy and biologic therapy in rheumatic diseases: an analysis of the FDA AERS database [ACR abstract 700]. Arthritis Rheum 2010; 62 (suppl):S292.
  13. Kumar D, Bouldin TW, Berger RG. A case of progressive multifocal leukoencephalopathy in a patient treated with infliximab. Arthritis Rheum 2010; 62:31913195.
  14. Nived O, Bengtsson AA, Jonsen A, Sturfelt G. Progressive multifocal leukoencephalopathy—the importance of early diagnosis illustrated in four cases. Lupus 2008; 17:10361041.
Article PDF
Author and Disclosure Information

Eamonn S. Molloy, MD, MS, MRCPI
Consultant Rheumatologist, Department of Rheumatology, St. Vincent’s University Hospital, Dublin, Ireland

Correspondence: Eamonn Molloy, MD, MS, MRCPI, Consultant Rheumatologist, Department of Rheumatology, St. Vincent’s University Hospital, Elm Park, Dublin 4, Ireland; E.Molloy@st-vincents.ie

Dr. Molloy reported teaching and speaking relationships with Abbott Laboratories, Bristol-Myers Squibb, and Roche Pharmaceuticals; and membership on an advisory committee or review panel for GlaxoSmithKline.

This article was developed from an audio transcript of Dr. Molloy’s presentation at a roundtable convened at Cleveland Clinic on January 31, 2011. The transcript was edited by the Cleveland Clinic Journal of Medicine staff for clarity and conciseness, and was then reviewed, revised, and approved by Dr. Molloy.

Publications
Page Number
S28-S32
Author and Disclosure Information

Eamonn S. Molloy, MD, MS, MRCPI
Consultant Rheumatologist, Department of Rheumatology, St. Vincent’s University Hospital, Dublin, Ireland

Correspondence: Eamonn Molloy, MD, MS, MRCPI, Consultant Rheumatologist, Department of Rheumatology, St. Vincent’s University Hospital, Elm Park, Dublin 4, Ireland; E.Molloy@st-vincents.ie

Dr. Molloy reported teaching and speaking relationships with Abbott Laboratories, Bristol-Myers Squibb, and Roche Pharmaceuticals; and membership on an advisory committee or review panel for GlaxoSmithKline.

This article was developed from an audio transcript of Dr. Molloy’s presentation at a roundtable convened at Cleveland Clinic on January 31, 2011. The transcript was edited by the Cleveland Clinic Journal of Medicine staff for clarity and conciseness, and was then reviewed, revised, and approved by Dr. Molloy.

Author and Disclosure Information

Eamonn S. Molloy, MD, MS, MRCPI
Consultant Rheumatologist, Department of Rheumatology, St. Vincent’s University Hospital, Dublin, Ireland

Correspondence: Eamonn Molloy, MD, MS, MRCPI, Consultant Rheumatologist, Department of Rheumatology, St. Vincent’s University Hospital, Elm Park, Dublin 4, Ireland; E.Molloy@st-vincents.ie

Dr. Molloy reported teaching and speaking relationships with Abbott Laboratories, Bristol-Myers Squibb, and Roche Pharmaceuticals; and membership on an advisory committee or review panel for GlaxoSmithKline.

This article was developed from an audio transcript of Dr. Molloy’s presentation at a roundtable convened at Cleveland Clinic on January 31, 2011. The transcript was edited by the Cleveland Clinic Journal of Medicine staff for clarity and conciseness, and was then reviewed, revised, and approved by Dr. Molloy.

Article PDF
Article PDF

Progressive multifocal leukoencephalopathy (PML) is a rare, typically fatal, opportunistic infection caused by the JC virus (JCV). Formerly an example of neurologic arcana, PML became an important clinical concern when it developed in patients with human immunodeficiency virus (HIV) infection. More recently, PML has attracted the attention of rheumatologists following reports of its being associated with the use of targeted therapies such as natalizumab and rituximab.1

A recent survey of rheumatologists’ knowledge of and attitudes towards PML revealed that concerns over PML affect decisions on the use of biologic agents. Further, rheumatologists have important real and perceived learning gaps regarding PML; for example, 41% of those surveyed could not identify the diagnostic test of choice for PML.2

PML IN RHEUMATIC DISEASES

The US Food and Drug Administration (FDA) issued an alert in December 2006 following documentation of two fatal cases of PML in patients with systemic lupus erythematosus (SLE), both of whom had been treated with rituximab.3 We subsequently performed a literature search to identify cases of PML associated with rheumatic diseases.1,4 Patients were included only if the information provided was sufficient to substantiate the diagnosis of PML and the rheumatic disease in question; patients were excluded if they had HIV or cancer or had undergone organ transplantation. The search revealed 50 patients with rheumatic diseases who had PML (Table 1); SLE was overrepresented (n = 32) despite a much lower population prevalence of SLE compared with rheumatoid arthritis.

Examination of the immunosuppressive therapies prescribed to these patients within 6 months of the onset of neurologic symptoms attributed to PML revealed that low-dose (≤ 15 mg/d) prednisone, with or without an antimalarial agent, was the only immunosuppressive therapy in 31% of patients with SLE and in 11% of patients with rheumatic diseases other than SLE. Three patients had no documented immunosuppressive therapy in the 6 months prior to the onset of PML. Two patients with SLE were prescribed rituximab; no cases were reported in association with biologic therapies other than rituximab.4

In order to circumvent reporting bias, a nationwide hospital discharge database representing nearly 300 million patient discharges was used to determine the relative frequency of PML in patients with rheumatic diseases.5 Because of the reliance on diagnostic coding, rheumatic diseases were likely underreported in this sample; information on therapies was unavailable. After excluding patients who had HIV or cancer or were organ transplant recipients, four cases of PML were identified per 100,000 SLE discharges. This rate was 10-fold higher than the rate associated with rheumatoid arthritis and 20-fold higher than that of the background population.

These data show that PML is a rare occurrence in patients with rheumatic diseases, and SLE appears to be associated with a predisposition to PML. This predisposition in patients with SLE does not appear to be proportional to the degree of iatrogenic immunosuppression, emphasizing the role of host factors.

DISEASE-MODIFYING DRUGS AND PML RISK

In addition to certain disease states, disease-modifying biologic drugs have recently been associated with rare instances of PML.

Rituximab

The first case of rituximab-associated PML in the setting of rheumatoid arthritis was recorded in September 2008.6 The patient had longstanding rheumatoid arthritis and Sjögren syndrome. She received four courses of rituximab and was diagnosed with PML 18 months after the last dose; she died 1 month later. Her therapy for rheumatoid arthritis included a tumor necrosis factor (TNF) antagonist prior to rituximab initiation and treatment with methotrexate and steroids before, during, and after rituximab therapy. Oropharyngeal cancer developed in this patient 9 months prior to the onset of PML and was treated with chemotherapy and radiation therapy.

Another case of PML in a patient with rheumatic disease who had been treated with rituximab was notable because it was the first in which the patient had not previously been treated with an anti-TNF agent.7

Ascertaining cause of PML in patients treated with rituximab is difficult because the potential pathogenic mechanism remains unknown. Humoral immunity is not protective against PML, leading to speculation that the loss of other B-cell functions, such as those of antigen-presenting cells or cytokine production, may lead to a defect in cell-mediated immunity. Another theory posits that reconstitution of naïve B cells with latent JCV infection following B-cell depletion from rituximab therapy may somehow facilitate the development of PML.

 

 

Efalizumab

Efalizumab is a monoclonal antibody that targets CD11a, the alpha subunit of lymphocyte function–associated antigen 1. Efalizumab blocks binding to intercellular adhesion molecule 1, and thereby blocks T-cell adhesion and migration. CD11a is also expressed on a variety of other leukocytes and lymphocytes such as B cells, monocytes, and natural killer cells.

Efalizumab was approved in 2003 by the FDA for the treatment of moderate to severe plaque psoriasis. It is estimated that 46,000 patients have been treated with efalizumab worldwide since its approval. In 2008, a black box warning was added to the efalizumab prescribing information following the occurrence of serious infections, including pulmonary tuberculosis, necrotizing fasciitis, and invasive fungal infections.8 Subsequently, four cases of PML, three of which were fatal, were reported in psoriasis patients treated with efalizumab. Of note, these were the first cases of PML reported in patients with psoriasis. Of more concern, the affected patients were among a group of approximately 1,100 patients who had been treated with efalizumab for more than 3 years. In February 2009, a public health advisory was issued by the FDA,9 and efalizumab was voluntarily withdrawn by its manufacturer 2 months later.

Belatacept

Belatacept is a recombinant soluble fusion protein of the extracellular domain of human cytotoxic T-lymphocyte antigen-4 with a fragment of a modified Fc domain of immunoglobulin G1. Recently approved by the FDA for prophylaxis of renal transplant rejection, it is a second-generation, higher-avidity version of abatacept. Abatacept is licensed for the treatment of rheumatoid arthritis and is under investigation for the treatment of vasculitis and SLE. Belatacept differs from abatacept by only two amino acids.

Two cases of PML have been reported in association with belatacept, one in a patient following renal transplantation and the other in a patient following liver transplantation. Both patients had been treated with other standard immunosuppressive therapies for prophylaxis of organ transplant rejection, including mycophenolate mofetil.

Mycophenolate mofetil

Mycophenolate mofetil is the prodrug of mycophenolic acid. Both have been the subjects of FDA alerts regarding PML, based on a January 2008 report of 10 definite and 7 possible cases of PML occurring with mycophenolate mofetil. The patients affected included four with SLE, none of whom underwent a renal transplant.10

In a retrospective cohort study of 32,757 renal transplant patients, Neff et al11 found 14 cases of PML per 100,000 person-years among patients treated with mycophenolate mofetil following kidney transplant compared with none in patients who did not receive mycophenolate mofetil. It is difficult to ascertain risk with mycophenolate mofetil because it is standard therapy among renal transplant patients, leaving few patients in these groups unexposed.

Given the FDA alert with respect to mycophenolate mofetil and PML,10 the frequent use of mycophenolate mofetil in the setting of SLE, and the concerns about possible predisposition to PML among patients with SLE, it will be important to clarify the level of risk in patients with SLE who are treated with mycophenolate mofetil.

AGGREGATE EXPERIENCE: REVIEW OF FEDERAL DATABASE

We examined the aggregate experience of PML in association with autoimmune disorders and biologic disease-modifying antirheumatic drug (DMARD) exposures reported in the FDA Adverse Event Reporting System (AERS) database.12 A total of 19 confirmed cases of PML in patients with rheumatic diseases were uncovered: 10 in patients with SLE, 5 in patients with rheumatoid arthritis, 3 in patients with vasculitis, and 1 in a patient with dermatomyositis. The patients with PML included six who received rituximab for the management of rheumatic diseases (Table 2). In all six patients, rituximab was the most recently prescribed DMARD. Four cases were identified in patients treated with anti-TNF therapy, but three of these had received anti-TNF therapy prior to rituximab, and the other was receiving concomitant cyclophosphamide for rheumatoid vasculitis.

Ten cases of PML were confirmed with cyclophosphamide treatment, and cyclophosphamide was the most recent DMARD prescribed in two of these cases. Five cases were confirmed with mycophenolate mofetil (in four of which it was the most recently prescribed DMARD) and six with azathioprine (in three of which it was the most recently prescribed DMARD).

Risk of PML with DMARD therapy

Rituximab. The confirmation of six cases of PML among rituximab-treated rheumatoid arthritis patients is a source of concern. Nevertheless, PML is a rare adverse event. It occurs in fewer than 1 in 10,000 rituximab-treated patients who have rheumatoid arthritis, among a total of approximately 130,000 such patients. A better understanding of the potential mechanism responsible for the increased risk of developing PML may help in risk prediction and to guide patient selection for this agent.

Anti-TNF therapy. A paucity of confirmed cases in patients treated with anti-TNF therapy argues against a significant risk of PML associated with this therapy, especially considering the estimated 2 to 3 million rheumatoid arthritis patients who are receiving treatment with anti-TNF agents. A note of caution is sounded by a recent case report of PML in a rheumatoid arthritis patient. The patient had been treated with infliximab, with the only background therapy being methotrexate.13 Ongoing vigilance is therefore necessary.

Mycophenolate mofetil. All five confirmed cases of PML in mycophenolate mofetil-treated patients had earlier received treatment with cyclophosphamide. These data indicate no clear signal of excess risk with mycophenolate mofetil above that seen with other nonbiologic immunosuppressive agents, such as cyclophosphamide or azathioprine.

CONCLUSION AND RECOMMENDATIONS

PML has been reported in association with a variety of disease states, although a predisposition in patients with SLE has become apparent. Synthetic and biologic immunosuppressive therapies have also been implicated, but PML may also occur in the setting of minimal iatrogenic immunosuppression.

Until greater clarity can be achieved, all patients with systemic rheumatic diseases should be considered at risk for PML, regardless of the nature or intensity of their immunosuppressive therapy. In this context, differentiating PML from neurologic syndromes related to the underlying rheumatic disease (eg, neuropsychiatric SLE, cerebral vasculitis) is critical, particularly given the markedly different approaches to management.

PML should be considered in patients with unexplained subacute progressive focal and diffuse neurologic deficits, especially if their clinical or radiologic status worsens in the face of increased intensity of immunosuppressive therapy. Spinal cord or optic nerve involvement argues against PML. A normal magnetic resonance image (MRI) has a high negative predictive value, and frank infarction is not a feature of PML. In classic PML, contrast enhancement is typically absent and routine cerebrospinal fluid (CSF) analysis is typically normal. However, contrast enhancement and edema on MRI, lymphocytic CSF pleocytosis, and elevated CSF protein may be seen in the more recently described “inflammatory PML,” in which case the distinction from cerebral vasculitis or neuropsychiatric SLE may be more difficult. Angiography appears normal in patients with PML.

The diagnostic test of choice is a polymerase chain reaction (PCR) assay for JCV in CSF. If the PCR is repeatedly negative, then a brain biopsy should be considered, especially in the setting of progressive neurologic decline in patients receiving immunosuppressive therapy.

 

 

DISCUSSION

Dr. Simpson: To what extent are these lesions in the brain being attributed to the underlying vasculitis, particularly in SLE, as opposed to pursuing a PML diagnosis, and how might this result in dramatic underreporting of the complication?

Dr. Molloy: We found that PML is almost certainly underdiagnosed, particularly in SLE patients. If a patient succumbs to assumed neuropsychiatric SLE, how often is an autopsy undertaken? One telling paper from Sweden documented four cases of PML in SLE patients.14 In one of these, the diagnosis was made retrospectively from autopsy tissue that had been banked 20 years previously. It undoubtedly is underdiagnosed.

Dr. Calabrese: Even in the most recent rituximab-associated cases of PML, several patients were empirically given additional immunosuppressive therapy because it was presumed that they had a comorbid neuropsychiatric rheumatic complication. The presence of neuropsychiatric complications ascribed to an autoinflammatory disease generally warrants escalation of immunosuppressive therapy. It has always been standard practice for us to rule out opportunistic infection, but JCV infection has not been on the radar screen until very recently.

Dr. Molloy: I’d like to emphasize that, in our literature review, 50% of the rheumatic disease patients diagnosed with PML had been treated with more intensive immunosuppressive therapy. It was only after they continued to deteriorate that JCV infection was suspected and PML ultimately diagnosed.

Dr. Berger: Is it fair to say that the incidence of PML in SLE is about 10 times that in rheumatoid arthritis?

Dr. Molloy: In the hospital discharge database, it was 10-fold higher in SLE than in rheumatoid arthritis, but we can’t draw a conclusion from the AERS database because we don’t have a denominator. The database consists of voluntary submission of cases.

Dr. Calabrese: The information that we can expect to glean from the database is profoundly limited, for all the reasons that you enumerated. Despite the flaws, we’re obligated to continuously examine it because sometimes a case or two may provide some special insight.

Dr. Simpson: As neurologists, we often lag behind rheumatologists in the use of new treatments, including intravenous immune globulin (IVIG) and now rituximab. Rituximab is becoming the go-to drug for a number of neurologic diseases. I’m using it quite a bit and have observed some dramatic responses in patients with chronic inflammatory demyelinating polyneuropathy, for example, in whom IVIG or plasmapheresis was failing. Anecdotally, some of the turnarounds in polymyositis and even myasthenia gravis are remarkable as well. I’m not sure to what extent neurologists—particularly peripheral neurologists—who use rituximab are recognizing PML.

Dr. Fox: The index of suspicion is probably vastly different among multiple sclerosis (MS) specialists and general neurologists. Neurologists who treat MS will be acutely aware of PML because of its association with natalizumab.

Dr. Berger: Yes, but you’re talking about possibly two orders of magnitude difference between natalizumab and rituximab. In fact, PML is rarely reported in the setting of neurologic disease. It’s mostly reported in the setting of rheumatologic disease.

Dr. Rudick: I don’t necessarily agree with you. Ascertaining the true incidence of PML with agents other than natalizumab is difficult. One is unlikely to miss a case of PML in an MS patient treated with natalizumab, but most cases stemming from the use of these other disease-modifying drugs are probably being missed.

Dr. Calabrese: I get two messages out of this body of work. Number one is that while PML is rare, it is seen across the spectrum of immunosuppressive agents, including biologic and nonbiologic drugs. Number two is that PML is seriously underreported and underrecognized, which is probably leading to suboptimal patient care. Rituximab was recently approved for treatment of Wegener granulomatosis, and this disease is heavily pretreated with cyclophosphamide. You would expect that PML is on the radar among clinicians caring for patients whose diseases warrant the use of increasingly complex, potent, and novel immunosuppressives.

Dr. Berger: There is one other biologic agent you left out—alemtuzumab. It wipes out all of the B cells and T cells; the B cells repopulate but the T cells remain suppressed for a long period. If ever there was a drug whose action mirrors what happens in HIV, alemtuzumab is that drug. Yet, PML is rarely seen with alemtuzumab. Alemtuzumab-associated PML has not been reported in the MS population, and it has only been seen in two transplantations that I’m aware of. I’m not saying that it doesn’t occur, but we’re not seeing it with the same frequency that one would predict given its profile.

Progressive multifocal leukoencephalopathy (PML) is a rare, typically fatal, opportunistic infection caused by the JC virus (JCV). Formerly an example of neurologic arcana, PML became an important clinical concern when it developed in patients with human immunodeficiency virus (HIV) infection. More recently, PML has attracted the attention of rheumatologists following reports of its being associated with the use of targeted therapies such as natalizumab and rituximab.1

A recent survey of rheumatologists’ knowledge of and attitudes towards PML revealed that concerns over PML affect decisions on the use of biologic agents. Further, rheumatologists have important real and perceived learning gaps regarding PML; for example, 41% of those surveyed could not identify the diagnostic test of choice for PML.2

PML IN RHEUMATIC DISEASES

The US Food and Drug Administration (FDA) issued an alert in December 2006 following documentation of two fatal cases of PML in patients with systemic lupus erythematosus (SLE), both of whom had been treated with rituximab.3 We subsequently performed a literature search to identify cases of PML associated with rheumatic diseases.1,4 Patients were included only if the information provided was sufficient to substantiate the diagnosis of PML and the rheumatic disease in question; patients were excluded if they had HIV or cancer or had undergone organ transplantation. The search revealed 50 patients with rheumatic diseases who had PML (Table 1); SLE was overrepresented (n = 32) despite a much lower population prevalence of SLE compared with rheumatoid arthritis.

Examination of the immunosuppressive therapies prescribed to these patients within 6 months of the onset of neurologic symptoms attributed to PML revealed that low-dose (≤ 15 mg/d) prednisone, with or without an antimalarial agent, was the only immunosuppressive therapy in 31% of patients with SLE and in 11% of patients with rheumatic diseases other than SLE. Three patients had no documented immunosuppressive therapy in the 6 months prior to the onset of PML. Two patients with SLE were prescribed rituximab; no cases were reported in association with biologic therapies other than rituximab.4

In order to circumvent reporting bias, a nationwide hospital discharge database representing nearly 300 million patient discharges was used to determine the relative frequency of PML in patients with rheumatic diseases.5 Because of the reliance on diagnostic coding, rheumatic diseases were likely underreported in this sample; information on therapies was unavailable. After excluding patients who had HIV or cancer or were organ transplant recipients, four cases of PML were identified per 100,000 SLE discharges. This rate was 10-fold higher than the rate associated with rheumatoid arthritis and 20-fold higher than that of the background population.

These data show that PML is a rare occurrence in patients with rheumatic diseases, and SLE appears to be associated with a predisposition to PML. This predisposition in patients with SLE does not appear to be proportional to the degree of iatrogenic immunosuppression, emphasizing the role of host factors.

DISEASE-MODIFYING DRUGS AND PML RISK

In addition to certain disease states, disease-modifying biologic drugs have recently been associated with rare instances of PML.

Rituximab

The first case of rituximab-associated PML in the setting of rheumatoid arthritis was recorded in September 2008.6 The patient had longstanding rheumatoid arthritis and Sjögren syndrome. She received four courses of rituximab and was diagnosed with PML 18 months after the last dose; she died 1 month later. Her therapy for rheumatoid arthritis included a tumor necrosis factor (TNF) antagonist prior to rituximab initiation and treatment with methotrexate and steroids before, during, and after rituximab therapy. Oropharyngeal cancer developed in this patient 9 months prior to the onset of PML and was treated with chemotherapy and radiation therapy.

Another case of PML in a patient with rheumatic disease who had been treated with rituximab was notable because it was the first in which the patient had not previously been treated with an anti-TNF agent.7

Ascertaining cause of PML in patients treated with rituximab is difficult because the potential pathogenic mechanism remains unknown. Humoral immunity is not protective against PML, leading to speculation that the loss of other B-cell functions, such as those of antigen-presenting cells or cytokine production, may lead to a defect in cell-mediated immunity. Another theory posits that reconstitution of naïve B cells with latent JCV infection following B-cell depletion from rituximab therapy may somehow facilitate the development of PML.

 

 

Efalizumab

Efalizumab is a monoclonal antibody that targets CD11a, the alpha subunit of lymphocyte function–associated antigen 1. Efalizumab blocks binding to intercellular adhesion molecule 1, and thereby blocks T-cell adhesion and migration. CD11a is also expressed on a variety of other leukocytes and lymphocytes such as B cells, monocytes, and natural killer cells.

Efalizumab was approved in 2003 by the FDA for the treatment of moderate to severe plaque psoriasis. It is estimated that 46,000 patients have been treated with efalizumab worldwide since its approval. In 2008, a black box warning was added to the efalizumab prescribing information following the occurrence of serious infections, including pulmonary tuberculosis, necrotizing fasciitis, and invasive fungal infections.8 Subsequently, four cases of PML, three of which were fatal, were reported in psoriasis patients treated with efalizumab. Of note, these were the first cases of PML reported in patients with psoriasis. Of more concern, the affected patients were among a group of approximately 1,100 patients who had been treated with efalizumab for more than 3 years. In February 2009, a public health advisory was issued by the FDA,9 and efalizumab was voluntarily withdrawn by its manufacturer 2 months later.

Belatacept

Belatacept is a recombinant soluble fusion protein of the extracellular domain of human cytotoxic T-lymphocyte antigen-4 with a fragment of a modified Fc domain of immunoglobulin G1. Recently approved by the FDA for prophylaxis of renal transplant rejection, it is a second-generation, higher-avidity version of abatacept. Abatacept is licensed for the treatment of rheumatoid arthritis and is under investigation for the treatment of vasculitis and SLE. Belatacept differs from abatacept by only two amino acids.

Two cases of PML have been reported in association with belatacept, one in a patient following renal transplantation and the other in a patient following liver transplantation. Both patients had been treated with other standard immunosuppressive therapies for prophylaxis of organ transplant rejection, including mycophenolate mofetil.

Mycophenolate mofetil

Mycophenolate mofetil is the prodrug of mycophenolic acid. Both have been the subjects of FDA alerts regarding PML, based on a January 2008 report of 10 definite and 7 possible cases of PML occurring with mycophenolate mofetil. The patients affected included four with SLE, none of whom underwent a renal transplant.10

In a retrospective cohort study of 32,757 renal transplant patients, Neff et al11 found 14 cases of PML per 100,000 person-years among patients treated with mycophenolate mofetil following kidney transplant compared with none in patients who did not receive mycophenolate mofetil. It is difficult to ascertain risk with mycophenolate mofetil because it is standard therapy among renal transplant patients, leaving few patients in these groups unexposed.

Given the FDA alert with respect to mycophenolate mofetil and PML,10 the frequent use of mycophenolate mofetil in the setting of SLE, and the concerns about possible predisposition to PML among patients with SLE, it will be important to clarify the level of risk in patients with SLE who are treated with mycophenolate mofetil.

AGGREGATE EXPERIENCE: REVIEW OF FEDERAL DATABASE

We examined the aggregate experience of PML in association with autoimmune disorders and biologic disease-modifying antirheumatic drug (DMARD) exposures reported in the FDA Adverse Event Reporting System (AERS) database.12 A total of 19 confirmed cases of PML in patients with rheumatic diseases were uncovered: 10 in patients with SLE, 5 in patients with rheumatoid arthritis, 3 in patients with vasculitis, and 1 in a patient with dermatomyositis. The patients with PML included six who received rituximab for the management of rheumatic diseases (Table 2). In all six patients, rituximab was the most recently prescribed DMARD. Four cases were identified in patients treated with anti-TNF therapy, but three of these had received anti-TNF therapy prior to rituximab, and the other was receiving concomitant cyclophosphamide for rheumatoid vasculitis.

Ten cases of PML were confirmed with cyclophosphamide treatment, and cyclophosphamide was the most recent DMARD prescribed in two of these cases. Five cases were confirmed with mycophenolate mofetil (in four of which it was the most recently prescribed DMARD) and six with azathioprine (in three of which it was the most recently prescribed DMARD).

Risk of PML with DMARD therapy

Rituximab. The confirmation of six cases of PML among rituximab-treated rheumatoid arthritis patients is a source of concern. Nevertheless, PML is a rare adverse event. It occurs in fewer than 1 in 10,000 rituximab-treated patients who have rheumatoid arthritis, among a total of approximately 130,000 such patients. A better understanding of the potential mechanism responsible for the increased risk of developing PML may help in risk prediction and to guide patient selection for this agent.

Anti-TNF therapy. A paucity of confirmed cases in patients treated with anti-TNF therapy argues against a significant risk of PML associated with this therapy, especially considering the estimated 2 to 3 million rheumatoid arthritis patients who are receiving treatment with anti-TNF agents. A note of caution is sounded by a recent case report of PML in a rheumatoid arthritis patient. The patient had been treated with infliximab, with the only background therapy being methotrexate.13 Ongoing vigilance is therefore necessary.

Mycophenolate mofetil. All five confirmed cases of PML in mycophenolate mofetil-treated patients had earlier received treatment with cyclophosphamide. These data indicate no clear signal of excess risk with mycophenolate mofetil above that seen with other nonbiologic immunosuppressive agents, such as cyclophosphamide or azathioprine.

CONCLUSION AND RECOMMENDATIONS

PML has been reported in association with a variety of disease states, although a predisposition in patients with SLE has become apparent. Synthetic and biologic immunosuppressive therapies have also been implicated, but PML may also occur in the setting of minimal iatrogenic immunosuppression.

Until greater clarity can be achieved, all patients with systemic rheumatic diseases should be considered at risk for PML, regardless of the nature or intensity of their immunosuppressive therapy. In this context, differentiating PML from neurologic syndromes related to the underlying rheumatic disease (eg, neuropsychiatric SLE, cerebral vasculitis) is critical, particularly given the markedly different approaches to management.

PML should be considered in patients with unexplained subacute progressive focal and diffuse neurologic deficits, especially if their clinical or radiologic status worsens in the face of increased intensity of immunosuppressive therapy. Spinal cord or optic nerve involvement argues against PML. A normal magnetic resonance image (MRI) has a high negative predictive value, and frank infarction is not a feature of PML. In classic PML, contrast enhancement is typically absent and routine cerebrospinal fluid (CSF) analysis is typically normal. However, contrast enhancement and edema on MRI, lymphocytic CSF pleocytosis, and elevated CSF protein may be seen in the more recently described “inflammatory PML,” in which case the distinction from cerebral vasculitis or neuropsychiatric SLE may be more difficult. Angiography appears normal in patients with PML.

The diagnostic test of choice is a polymerase chain reaction (PCR) assay for JCV in CSF. If the PCR is repeatedly negative, then a brain biopsy should be considered, especially in the setting of progressive neurologic decline in patients receiving immunosuppressive therapy.

 

 

DISCUSSION

Dr. Simpson: To what extent are these lesions in the brain being attributed to the underlying vasculitis, particularly in SLE, as opposed to pursuing a PML diagnosis, and how might this result in dramatic underreporting of the complication?

Dr. Molloy: We found that PML is almost certainly underdiagnosed, particularly in SLE patients. If a patient succumbs to assumed neuropsychiatric SLE, how often is an autopsy undertaken? One telling paper from Sweden documented four cases of PML in SLE patients.14 In one of these, the diagnosis was made retrospectively from autopsy tissue that had been banked 20 years previously. It undoubtedly is underdiagnosed.

Dr. Calabrese: Even in the most recent rituximab-associated cases of PML, several patients were empirically given additional immunosuppressive therapy because it was presumed that they had a comorbid neuropsychiatric rheumatic complication. The presence of neuropsychiatric complications ascribed to an autoinflammatory disease generally warrants escalation of immunosuppressive therapy. It has always been standard practice for us to rule out opportunistic infection, but JCV infection has not been on the radar screen until very recently.

Dr. Molloy: I’d like to emphasize that, in our literature review, 50% of the rheumatic disease patients diagnosed with PML had been treated with more intensive immunosuppressive therapy. It was only after they continued to deteriorate that JCV infection was suspected and PML ultimately diagnosed.

Dr. Berger: Is it fair to say that the incidence of PML in SLE is about 10 times that in rheumatoid arthritis?

Dr. Molloy: In the hospital discharge database, it was 10-fold higher in SLE than in rheumatoid arthritis, but we can’t draw a conclusion from the AERS database because we don’t have a denominator. The database consists of voluntary submission of cases.

Dr. Calabrese: The information that we can expect to glean from the database is profoundly limited, for all the reasons that you enumerated. Despite the flaws, we’re obligated to continuously examine it because sometimes a case or two may provide some special insight.

Dr. Simpson: As neurologists, we often lag behind rheumatologists in the use of new treatments, including intravenous immune globulin (IVIG) and now rituximab. Rituximab is becoming the go-to drug for a number of neurologic diseases. I’m using it quite a bit and have observed some dramatic responses in patients with chronic inflammatory demyelinating polyneuropathy, for example, in whom IVIG or plasmapheresis was failing. Anecdotally, some of the turnarounds in polymyositis and even myasthenia gravis are remarkable as well. I’m not sure to what extent neurologists—particularly peripheral neurologists—who use rituximab are recognizing PML.

Dr. Fox: The index of suspicion is probably vastly different among multiple sclerosis (MS) specialists and general neurologists. Neurologists who treat MS will be acutely aware of PML because of its association with natalizumab.

Dr. Berger: Yes, but you’re talking about possibly two orders of magnitude difference between natalizumab and rituximab. In fact, PML is rarely reported in the setting of neurologic disease. It’s mostly reported in the setting of rheumatologic disease.

Dr. Rudick: I don’t necessarily agree with you. Ascertaining the true incidence of PML with agents other than natalizumab is difficult. One is unlikely to miss a case of PML in an MS patient treated with natalizumab, but most cases stemming from the use of these other disease-modifying drugs are probably being missed.

Dr. Calabrese: I get two messages out of this body of work. Number one is that while PML is rare, it is seen across the spectrum of immunosuppressive agents, including biologic and nonbiologic drugs. Number two is that PML is seriously underreported and underrecognized, which is probably leading to suboptimal patient care. Rituximab was recently approved for treatment of Wegener granulomatosis, and this disease is heavily pretreated with cyclophosphamide. You would expect that PML is on the radar among clinicians caring for patients whose diseases warrant the use of increasingly complex, potent, and novel immunosuppressives.

Dr. Berger: There is one other biologic agent you left out—alemtuzumab. It wipes out all of the B cells and T cells; the B cells repopulate but the T cells remain suppressed for a long period. If ever there was a drug whose action mirrors what happens in HIV, alemtuzumab is that drug. Yet, PML is rarely seen with alemtuzumab. Alemtuzumab-associated PML has not been reported in the MS population, and it has only been seen in two transplantations that I’m aware of. I’m not saying that it doesn’t occur, but we’re not seeing it with the same frequency that one would predict given its profile.

References
  1. Calabrese LH, Molloy ES, Huang D, Ransohoff RM. Progressive multifocal leukoencephalopathy in rheumatic diseases: evolving clinical and pathologic patterns of disease. Arthritis Rheum 2007; 6:21162128.
  2. Calabrese LH, Molloy ES, Taege AJ. Rheumatologists’ knowledge, attitudes and concerns regarding progressive multifocal encephalopathy (PML) [abstract]. Arthritis Rheum 2009; 60 (suppl 10):130.
  3. Rituxan (rituximab). U.S. Food and Drug Administration Web site. http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsforHumanMedicalProducts/ucm150747.htm. Updated June 19, 2009. Accessed September 15, 2011.
  4. Molloy ES, Calabrese LH. Progressive multifocal leukoencephalopathy in patients with rheumatic diseases: are patients with systemic lupus erythematosus at particular risk? Autoimmun Rev 2008; 8:144146.
  5. Molloy ES, Calabrese LH. Progressive multifocal leukoencephalopathy: a national estimate of frequency in systemic lupus erythematosus and other rheumatic diseases. Arthritis Rheum 2009; 60:37613765.
  6. Fleischmann RM. Progressive multifocal leukoencephalopathy following rituximab treatment in a patient with rheumatoid arthritis. Arthritis Rheum 2009; 60:32253228.
  7. Rituxan (rituximab) – PML. U.S. Food and Drug Administration Web site. http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsforHumanMedicalProducts/ucm187791.htm. Updated October 23, 2009. Accessed September 15, 2011.
  8. Raptiva (efalizumab) October 2008. U.S. Food and Drug Administration Web site. http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsforHumanMedicalProducts/ucm092089.htm. Updated June 19, 2009. Accessed September 15, 2011.
  9. Raptiva (efalizumab) Feb 2009. U.S. Food and Drug Administration Web site. http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsforHumanMedicalProducts/ucm149675.htm. Updated June 16, 2009. Accessed September 15, 2011.
  10. Communication about an ongoing safety review of CellCept (mycophenolate mofetil) and Myfortic (mycophenolic acid). U.S. Food and Drug Administration Web site. http://www.fda.gov/Drugs/DrugSafety/PostmarketDrugSafetyInformationforPatientsandProviders/DrugSafetyInformationforHeathcareProfessionals/ucm072438.htm. Updated October 30, 2009. Accessed September 15, 2011.
  11. Neff RT, Hurst FP, Falta EM, et al. Progressive multifocal leukoencephalopathy and use of mycophenolate mofetil after kidney transplantation. Transplantation 2008; 86:14741478.
  12. Molloy ES, Calabrese LH. Progressive multifocal leukoencephalopathy and biologic therapy in rheumatic diseases: an analysis of the FDA AERS database [ACR abstract 700]. Arthritis Rheum 2010; 62 (suppl):S292.
  13. Kumar D, Bouldin TW, Berger RG. A case of progressive multifocal leukoencephalopathy in a patient treated with infliximab. Arthritis Rheum 2010; 62:31913195.
  14. Nived O, Bengtsson AA, Jonsen A, Sturfelt G. Progressive multifocal leukoencephalopathy—the importance of early diagnosis illustrated in four cases. Lupus 2008; 17:10361041.
References
  1. Calabrese LH, Molloy ES, Huang D, Ransohoff RM. Progressive multifocal leukoencephalopathy in rheumatic diseases: evolving clinical and pathologic patterns of disease. Arthritis Rheum 2007; 6:21162128.
  2. Calabrese LH, Molloy ES, Taege AJ. Rheumatologists’ knowledge, attitudes and concerns regarding progressive multifocal encephalopathy (PML) [abstract]. Arthritis Rheum 2009; 60 (suppl 10):130.
  3. Rituxan (rituximab). U.S. Food and Drug Administration Web site. http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsforHumanMedicalProducts/ucm150747.htm. Updated June 19, 2009. Accessed September 15, 2011.
  4. Molloy ES, Calabrese LH. Progressive multifocal leukoencephalopathy in patients with rheumatic diseases: are patients with systemic lupus erythematosus at particular risk? Autoimmun Rev 2008; 8:144146.
  5. Molloy ES, Calabrese LH. Progressive multifocal leukoencephalopathy: a national estimate of frequency in systemic lupus erythematosus and other rheumatic diseases. Arthritis Rheum 2009; 60:37613765.
  6. Fleischmann RM. Progressive multifocal leukoencephalopathy following rituximab treatment in a patient with rheumatoid arthritis. Arthritis Rheum 2009; 60:32253228.
  7. Rituxan (rituximab) – PML. U.S. Food and Drug Administration Web site. http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsforHumanMedicalProducts/ucm187791.htm. Updated October 23, 2009. Accessed September 15, 2011.
  8. Raptiva (efalizumab) October 2008. U.S. Food and Drug Administration Web site. http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsforHumanMedicalProducts/ucm092089.htm. Updated June 19, 2009. Accessed September 15, 2011.
  9. Raptiva (efalizumab) Feb 2009. U.S. Food and Drug Administration Web site. http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsforHumanMedicalProducts/ucm149675.htm. Updated June 16, 2009. Accessed September 15, 2011.
  10. Communication about an ongoing safety review of CellCept (mycophenolate mofetil) and Myfortic (mycophenolic acid). U.S. Food and Drug Administration Web site. http://www.fda.gov/Drugs/DrugSafety/PostmarketDrugSafetyInformationforPatientsandProviders/DrugSafetyInformationforHeathcareProfessionals/ucm072438.htm. Updated October 30, 2009. Accessed September 15, 2011.
  11. Neff RT, Hurst FP, Falta EM, et al. Progressive multifocal leukoencephalopathy and use of mycophenolate mofetil after kidney transplantation. Transplantation 2008; 86:14741478.
  12. Molloy ES, Calabrese LH. Progressive multifocal leukoencephalopathy and biologic therapy in rheumatic diseases: an analysis of the FDA AERS database [ACR abstract 700]. Arthritis Rheum 2010; 62 (suppl):S292.
  13. Kumar D, Bouldin TW, Berger RG. A case of progressive multifocal leukoencephalopathy in a patient treated with infliximab. Arthritis Rheum 2010; 62:31913195.
  14. Nived O, Bengtsson AA, Jonsen A, Sturfelt G. Progressive multifocal leukoencephalopathy—the importance of early diagnosis illustrated in four cases. Lupus 2008; 17:10361041.
Page Number
S28-S32
Page Number
S28-S32
Publications
Publications
Article Type
Display Headline
PML and rheumatology: The contribution of disease and drugs
Display Headline
PML and rheumatology: The contribution of disease and drugs
Citation Override
Cleveland Clinic Journal of Medicine 2011 November;78(suppl 2):S28-S32
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media