OBG Management is a leading publication in the ObGyn specialty addressing patient care and practice management under one cover.

Top Sections
Product Review
Expert Commentary
Clinical Review
obgm
Main menu
OBGM Main Menu
Explore menu
OBGM Explore Menu
Proclivity ID
18811001
Unpublish
Citation Name
OBG Manag
Specialty Focus
Obstetrics
Gynecology
Surgery
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
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
Clinical
Slot System
Top 25
Disable Sticky Ads
Disable Ad Block Mitigation
Featured Buckets Admin
Publication LayerRX Default ID
795
Non-Overridden Topics
Show Ads on this Publication's Homepage
Consolidated Pub
Show Article Page Numbers on TOC
Expire Announcement Bar
Thu, 08/01/2024 - 09:16
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
Challenge Center
Disable Inline Native ads
survey writer start date
Thu, 08/01/2024 - 09:16

Hormonal contraception and lactation: Reset your practices based on the evidence

Article Type
Changed
Tue, 02/28/2023 - 12:00

 

CASE Patient concerned about hormonal contraception’s impact on lactation

A 19-year-old woman (G2P1102) is postpartum day 1 after delivering a baby at 26 weeks’ gestation. When you see her on postpartum rounds, she states that she does not want any hormonal contraception because she heard that it will decrease her milk supply. What are your next steps?
 

The American Academy of Pediatrics recently updated its policy statement on breastfeeding and the use of human milk to recommend exclusive breastfeeding for 6 months and continued breastfeeding, with complementary foods, as mutually desired for 2 years or beyond given evidence of maternal health benefits with breastfeeding longer than 1 year.1

Breastfeeding prevalence—and challenges

Despite maternal and infant benefits associated with lactation, current breastfeeding prevalence in the United States remains suboptimal. In 2019, 24.9% of infants were exclusively breastfed through 6 months and 35.9% were breastfeeding at 12 months.2 Furthermore, disparities in breastfeeding exist, which contribute to health inequities. For example, non-Hispanic Black infants had lower rates of exclusive breastfeeding at 6 months (19.1%) and any breastfeeding at 12 months (24.1%) compared with non-Hispanic White infants (26.9% and 39.4%, respectively).3

While many new mothers intend to breastfeed and initiate breastfeeding in the hospital after delivery, overall and exclusive breastfeeding continuation rates are low, indicating that patients face challenges with breastfeeding after hospital discharge. Many structural and societal barriers to breastfeeding exist, including inadequate social support and parental leave policies.4 Suboptimal maternity care practices during the birth hospitalization may lead to challenges with breastfeeding initiation. Health care practitioners may present additional barriers to breastfeeding due to a lack of knowledge of available resources for patients or incomplete training in breastfeeding counseling and support.

To address our case patient’s concerns, clinicians should be aware of how exogenous progestins may affect breastfeeding physiology, risk factors for breastfeeding difficulty, and the available evidence for safety of hormonal contraception use while breastfeeding.

ILLUSTRATION: KIMBERLY MARTENS FOR OBG MANAGEMENT

Physiology of breastfeeding

During the second half of pregnancy, secretory differentiation (lactogenesis I) of mammary alveolar epithelial cells into secretory cells occurs to allow the mammary gland to eventually produce milk.5 After delivery of the placenta, progesterone withdrawal triggers secretory activation (lactogenesis II), which refers to the onset of copious milk production within 2 to 3 days postpartum.5 Most patients experience secretory activation within 72 hours; however, a delay in secretory activation past 72 hours is associated with cessation of any and exclusive breastfeeding at 4 weeks postpartum.6

Impaired lactation can be related to a delay in secretory activation or to insufficient lactation related to low milk supply. Maternal medical comorbidities (for example, diabetes mellitus, thyroid dysfunction, obesity), breast anatomy (such as insufficient glandular tissue, prior breast reduction surgery), pregnancy-related events (preeclampsia, retained placenta, postpartum hemorrhage), and infant conditions (such as multiple gestation, premature birth, congenital anomalies) all contribute to a risk of impaired lactation.7

 

Guidance on breastfeeding and hormonal contraception initiation

Early initiation of hormonal contraception poses theoretical concerns about breastfeeding difficulty if exogenous progestin interferes with endogenous signals for onset of milk production. The Centers for Disease Control and Prevention US Medical Eligibility Criteria (MEC) for Contraceptive Use provide recommendations on the safety of contraceptive use in the setting of various medical conditions or patient characteristics based on available data. The MEC uses 4 categories in assessing the safety of contraceptive method use for individuals with specific medical conditions or characteristics: 1, no restrictions exist for use of the contraceptive method; 2, advantages generally outweigh theoretical or proven risks; 3, theoretical or proven risks usually outweigh the advantages; and 4, conditions that represent an unacceptable health risk if the method is used.8

In the 2016 guidelines, combined hormonal contraceptives are considered category 4 at less than 21 days postpartum, regardless of breastfeeding status, due to the increased risk of venous thromboembolism in the immediate postpartum period (TABLE 1).8 Progestin-only contraception is considered category 1 in nonbreastfeeding individuals and category 2 in breastfeeding individuals based on overall evidence that found no adverse outcome with breastfeeding or infant outcomes with early initiation of progestin-only contraception (TABLE 1, TABLE 2).8

 

Since the publication of the 2016 MEC guidelines, several studies have continued to examine breastfeeding and infant outcomes with early initiation of hormonal contraception.

  • In a noninferiority randomized controlled trial of immediate versus delayed initiation of a levonorgestrel intrauterine device (LNG IUD), any breastfeeding at 8 weeks in the immediate group was 78% (95% confidence interval [CI], 70%–85%), which was lower than but within the specified noninferiority margin of the delayed breastfeeding group (83%; 95% CI, 75%–90%), indicating that breastfeeding outcomes with immediate initiation of an LNG IUD were not worse compared with delayed initiation.9
  • A secondary analysis of a randomized trial that compared intracesarean versus LNG IUD placement at 6 or more weeks postpartum showed no difference in breastfeeding at 6, 12, and 24 weeks after LNG IUD placement.10
  • A randomized trial of early (up to 48 hours postpartum) versus placement of an etonogestrel (ENG) implant at 6 or more weeks postpartum showed no difference between groups in infant weight at 12 months.11
  • A randomized trial of immediate (within 5 days of delivery) or interval placement of the 2-rod LNG implant (not approved in the United States) showed no difference in change in infant weight from birth to 6 months after delivery, onset of secretory activation, or breastfeeding continuation at 3 and 6 months postpartum.12
  • In a prospective cohort study that compared immediate postpartum initiation of ENG versus a 2-rod LNG implant (approved by the FDA but not marketed in the United States), there were no differences in breastfeeding continuation at 24 months and exclusive breastfeeding at 6 months postpartum.13
  • In a noninferiority randomized controlled trial that compared ENG implant initiation in the delivery room (0–2 hours postdelivery) versus delayed initiation (24–48 hours postdelivery), the time to secretory activation in those who initiated an ENG implant in the delivery room (66.8 [SD, 25.2] hours) was noninferior to delayed initiation (66.0 [SD, 35.3] hours). There also was no difference in ongoing breastfeeding over the first year after delivery and implant use at 12 months.14
  • A secondary analysis of a randomized controlled trial examined breastfeeding outcomes with receipt of depot medroxyprogesterone acetate (DMPA) prior to discharge in women who delivered infants who weighed 1,500 g or less at 32 weeks’ or less gestation. Time to secretory activation was longer in 29 women who received DMPA (103.7 hours) compared with 141 women who did not (88.6 hours; P = .028); however, there was no difference in daily milk production, lactation duration, or infant consumption of mother’s own milk.15

While the overall evidence suggests that early initiation of hormonal contraception does not affect breastfeeding or infant outcomes, it is important for clinicians to recognize the limitations of available data with regard to the populations included in these studies. Specifically, most studies did not include individuals with premature, low birth weight, or multiple gestation infants, who are at higher risk of impaired lactation, and individuals with a higher prevalence of breastfeeding were not included to determine whether early initiation of hormonal contraception would impact breastfeeding. Furthermore, while these studies enrolled participants who planned to breastfeed, data indicate that intentions to initiate and continue exclusive breastfeeding can vary.16 As the reported rates of any and exclusive breastfeeding are consistent with or lower than current US breastfeeding rates, any decrease in breastfeeding exclusivity or duration that may be attributable to hormonal contraception may be unacceptable to those who are strongly motivated to breastfeed.

Continue to: How can clinicians integrate evidence into contraception counseling?...

 

 

How can clinicians integrate evidence into contraception counseling?

The American College of Obstetricians and Gynecologists and the Academy of Breastfeeding Medicine offer guidance for how clinicians can address the use of hormonal contraception in breastfeeding patients. Both organizations recommend discussing the risks and benefits of hormonal contraception within the context of each person’s desire to breastfeed, potential for breastfeeding difficulty, and risk of pregnancy so that individuals can make their own informed decisions.17,18

Obstetric care clinicians have an important role in helping patients make informed infant feeding decisions without coercion or pressure. To start these discussions, clinicians can begin by assessing a patient’s breastfeeding goals by asking open-ended questions, such as:

  • What have you heard about breastfeeding?
  • What are your plans for returning to work or school after delivery?
  • How did breastfeeding go with older children?
  • What are your plans for feeding this baby?

In addition to gathering information about the patient’s priorities and goals, clinicians should identify any risk factors for breastfeeding challenges in the medical, surgical, or previous breastfeeding history. Clinicians can engage in a patient-centered approach to infant feeding decisions by anticipating any challenges and working together to develop strategies to address these challenges with the patient’s goals in mind.17

 

When counseling about contraception, a spectrum of approaches exists, from a nondirective information-sharing only model to directive counseling by the clinician. The shared decision-making model lies between these 2 approaches and recognizes the expertise of both the clinician and patient.19 To start these interactions, clinicians can ask about a patient’s reproductive goals by assessing the patient’s needs, values, and preferences for contraception. Potential questions include:

  • What kinds of contraceptive methods have you used in the past?
  • What is important to you in a contraceptive method?
  • How important is it to you to avoid another pregnancy right now?

Clinicians can then share information about different contraceptive methods based on the desired qualities that the patient has identified and how each method fits or does not fit into the patient’s goals and preferences. This collaborative approach facilitates an open dialogue and supports patient autonomy in contraceptive decision-making.

Lastly, clinicians should be cognizant of their own potential biases that could affect their counseling, such as encouraging contraceptive use because of a patient’s young age, parity, or premature delivery, as in our case presentation. Similarly, clinicians also should recognize that breastfeeding and contraceptive decisions are personal and are made with cultural, historical, and social contexts in mind.20 Ultimately, counseling should be patient centered and individualized for each person’s priorities related to infant feeding and pregnancy prevention. ●

References

 

  1. Meek JY, Noble L; Section on Breastfeeding. Policy statement: breastfeeding and the use of human milk. Pediatrics. 2022;150:e2022057988.
  2. Centers for Disease Control and Prevention. Breastfeeding report card, United States 2022. Accessed November 8, 2022. https://www.cdc.gov/breastfeeding/pdf/2022-Breast feeding-Report-Card-H.pdf
  3. Centers for Disease Control and Prevention. Rates of any and exclusive breastfeeding by sociodemographic characteristic among children born in 2019. Accessed November 8, 2022. https://www.cdc.gov/breastfeeding/data/nis_data/data-files/2019/rates-any-exclusive-bf-socio-dem-2019.html
  4. American College of Obstetricians and Gynecologists. Committee opinion no. 821: barriers to breastfeeding: supporting initiation and continuation of breastfeeding. Obstet Gynecol. 2021;137:e54-e62.
  5. Pang WW, Hartmann PE. Initiation of human lactation: secretory differentiation and secretory activation. J Mammary Gland Biol Neoplasia. 2007;12:211-221.
  6. Brownell E, Howard CR, Lawrence RA, et al. Delayed onset lactogenesis II predicts the cessation of any or exclusive breastfeeding. J Pediatr. 2012;161:608-614.
  7. American College of Obstetricians and Gynecologists. Committee opinion no. 820: breastfeeding challenges. Obstet Gynecol. 2021;137:e42-e53.
  8. Curtis KM, Tepper NK, Jatlaoui TC, et al. US Medical Eligibility Criteria for Contraceptive Use, 2016. MMWR Recomm Rep. 2016;65(RR-3):1-104.
  9. Turok DK, Leeman L, Sanders JN, et al. Immediate postpartum levonorgestrel intrauterine device insertion and breast-feeding outcomes: a noninferiority randomized controlled trial. Am J Obstet Gynecol. 2017;217:665.e1-665.e8.
  10. Levi EE, Findley MK, Avila K, et al. Placement of levonorgestrel intrauterine device at the time of cesarean delivery and the effect on breastfeeding duration. Breastfeed Med. 2018;13:674-679.
  11. Carmo LSMP, Braga GC, Ferriani RA, et al. Timing of etonogestrel-releasing implants and growth of breastfed infants: a randomized controlled trial. Obstet Gynecol. 2017;130:100-107.
  12. Averbach S, Kakaire O, McDiehl R, et al. The effect of immediate postpartum levonorgestrel contraceptive implant use on breastfeeding and infant growth: a randomized controlled trial. Contraception. 2019;99:87-93.
  13. Krashin JW, Lemani C, Nkambule J, et al. A comparison of breastfeeding exclusivity and duration rates between immediate postpartum levonorgestrel versus etonogestrel implant users: a prospective cohort study. Breastfeed Med. 2019;14:69-76.
  14. Henkel A, Lerma K, Reyes G, et al. Lactogenesis and breastfeeding after immediate vs delayed birth-hospitalization insertion of etonogestrel contraceptive implant: a noninferiority trial. Am J Obstet Gynecol. 2023; 228:55.e1-55.e9.
  15. Parker LA, Sullivan S, Cacho N, et al. Effect of postpartum depo medroxyprogesterone acetate on lactation in mothers of very low-birth-weight infants. Breastfeed Med. 2021;16:835-842.
  16. Nommsen-Rivers LA, Dewey KG. Development and validation of the infant feeding intentions scale. Matern Child Health J. 2009;13:334-342.
  17. American College of Obstetricians and Gynecologists. Committee opinion no. 756: optimizing support for breastfeeding as part of obstetric practice. Obstet Gynecol. 2018;132:e187-e196.
  18. Berens P, Labbok M; Academy of Breastfeeding Medicine. ABM Clinical Protocol #13: contraception during breastfeeding, revised 2015. Breastfeed Med. 2015;10:3-12.
  19. American College of Obstetricians and Gynecologists, Committee on Health Care for Underserved Women, Contraceptive Equity Expert Work Group, and Committee on Ethics. Committee statement no. 1: patient-centered contraceptive counseling. Obstet Gynecol. 2022;139:350-353.
  20. Bryant AG, Lyerly AD, DeVane-Johnson S, et al. Hormonal contraception, breastfeeding and bedside advocacy: the case for patient-centered care. Contraception. 2019;99:73-76.
Article PDF
Author and Disclosure Information

Dr. Chen is Associate Professor, Department of Obstetrics and Gynecology, University of California, Davis.

Dr. Crowe is Clinical Professor, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California.

The authors report no financial relationships relevant to this article.

Issue
OBG Management - 35(2)
Publications
Topics
Page Number
46-51
Sections
Author and Disclosure Information

Dr. Chen is Associate Professor, Department of Obstetrics and Gynecology, University of California, Davis.

Dr. Crowe is Clinical Professor, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California.

The authors report no financial relationships relevant to this article.

Author and Disclosure Information

Dr. Chen is Associate Professor, Department of Obstetrics and Gynecology, University of California, Davis.

Dr. Crowe is Clinical Professor, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California.

The authors report no financial relationships relevant to this article.

Article PDF
Article PDF

 

CASE Patient concerned about hormonal contraception’s impact on lactation

A 19-year-old woman (G2P1102) is postpartum day 1 after delivering a baby at 26 weeks’ gestation. When you see her on postpartum rounds, she states that she does not want any hormonal contraception because she heard that it will decrease her milk supply. What are your next steps?
 

The American Academy of Pediatrics recently updated its policy statement on breastfeeding and the use of human milk to recommend exclusive breastfeeding for 6 months and continued breastfeeding, with complementary foods, as mutually desired for 2 years or beyond given evidence of maternal health benefits with breastfeeding longer than 1 year.1

Breastfeeding prevalence—and challenges

Despite maternal and infant benefits associated with lactation, current breastfeeding prevalence in the United States remains suboptimal. In 2019, 24.9% of infants were exclusively breastfed through 6 months and 35.9% were breastfeeding at 12 months.2 Furthermore, disparities in breastfeeding exist, which contribute to health inequities. For example, non-Hispanic Black infants had lower rates of exclusive breastfeeding at 6 months (19.1%) and any breastfeeding at 12 months (24.1%) compared with non-Hispanic White infants (26.9% and 39.4%, respectively).3

While many new mothers intend to breastfeed and initiate breastfeeding in the hospital after delivery, overall and exclusive breastfeeding continuation rates are low, indicating that patients face challenges with breastfeeding after hospital discharge. Many structural and societal barriers to breastfeeding exist, including inadequate social support and parental leave policies.4 Suboptimal maternity care practices during the birth hospitalization may lead to challenges with breastfeeding initiation. Health care practitioners may present additional barriers to breastfeeding due to a lack of knowledge of available resources for patients or incomplete training in breastfeeding counseling and support.

To address our case patient’s concerns, clinicians should be aware of how exogenous progestins may affect breastfeeding physiology, risk factors for breastfeeding difficulty, and the available evidence for safety of hormonal contraception use while breastfeeding.

ILLUSTRATION: KIMBERLY MARTENS FOR OBG MANAGEMENT

Physiology of breastfeeding

During the second half of pregnancy, secretory differentiation (lactogenesis I) of mammary alveolar epithelial cells into secretory cells occurs to allow the mammary gland to eventually produce milk.5 After delivery of the placenta, progesterone withdrawal triggers secretory activation (lactogenesis II), which refers to the onset of copious milk production within 2 to 3 days postpartum.5 Most patients experience secretory activation within 72 hours; however, a delay in secretory activation past 72 hours is associated with cessation of any and exclusive breastfeeding at 4 weeks postpartum.6

Impaired lactation can be related to a delay in secretory activation or to insufficient lactation related to low milk supply. Maternal medical comorbidities (for example, diabetes mellitus, thyroid dysfunction, obesity), breast anatomy (such as insufficient glandular tissue, prior breast reduction surgery), pregnancy-related events (preeclampsia, retained placenta, postpartum hemorrhage), and infant conditions (such as multiple gestation, premature birth, congenital anomalies) all contribute to a risk of impaired lactation.7

 

Guidance on breastfeeding and hormonal contraception initiation

Early initiation of hormonal contraception poses theoretical concerns about breastfeeding difficulty if exogenous progestin interferes with endogenous signals for onset of milk production. The Centers for Disease Control and Prevention US Medical Eligibility Criteria (MEC) for Contraceptive Use provide recommendations on the safety of contraceptive use in the setting of various medical conditions or patient characteristics based on available data. The MEC uses 4 categories in assessing the safety of contraceptive method use for individuals with specific medical conditions or characteristics: 1, no restrictions exist for use of the contraceptive method; 2, advantages generally outweigh theoretical or proven risks; 3, theoretical or proven risks usually outweigh the advantages; and 4, conditions that represent an unacceptable health risk if the method is used.8

In the 2016 guidelines, combined hormonal contraceptives are considered category 4 at less than 21 days postpartum, regardless of breastfeeding status, due to the increased risk of venous thromboembolism in the immediate postpartum period (TABLE 1).8 Progestin-only contraception is considered category 1 in nonbreastfeeding individuals and category 2 in breastfeeding individuals based on overall evidence that found no adverse outcome with breastfeeding or infant outcomes with early initiation of progestin-only contraception (TABLE 1, TABLE 2).8

 

Since the publication of the 2016 MEC guidelines, several studies have continued to examine breastfeeding and infant outcomes with early initiation of hormonal contraception.

  • In a noninferiority randomized controlled trial of immediate versus delayed initiation of a levonorgestrel intrauterine device (LNG IUD), any breastfeeding at 8 weeks in the immediate group was 78% (95% confidence interval [CI], 70%–85%), which was lower than but within the specified noninferiority margin of the delayed breastfeeding group (83%; 95% CI, 75%–90%), indicating that breastfeeding outcomes with immediate initiation of an LNG IUD were not worse compared with delayed initiation.9
  • A secondary analysis of a randomized trial that compared intracesarean versus LNG IUD placement at 6 or more weeks postpartum showed no difference in breastfeeding at 6, 12, and 24 weeks after LNG IUD placement.10
  • A randomized trial of early (up to 48 hours postpartum) versus placement of an etonogestrel (ENG) implant at 6 or more weeks postpartum showed no difference between groups in infant weight at 12 months.11
  • A randomized trial of immediate (within 5 days of delivery) or interval placement of the 2-rod LNG implant (not approved in the United States) showed no difference in change in infant weight from birth to 6 months after delivery, onset of secretory activation, or breastfeeding continuation at 3 and 6 months postpartum.12
  • In a prospective cohort study that compared immediate postpartum initiation of ENG versus a 2-rod LNG implant (approved by the FDA but not marketed in the United States), there were no differences in breastfeeding continuation at 24 months and exclusive breastfeeding at 6 months postpartum.13
  • In a noninferiority randomized controlled trial that compared ENG implant initiation in the delivery room (0–2 hours postdelivery) versus delayed initiation (24–48 hours postdelivery), the time to secretory activation in those who initiated an ENG implant in the delivery room (66.8 [SD, 25.2] hours) was noninferior to delayed initiation (66.0 [SD, 35.3] hours). There also was no difference in ongoing breastfeeding over the first year after delivery and implant use at 12 months.14
  • A secondary analysis of a randomized controlled trial examined breastfeeding outcomes with receipt of depot medroxyprogesterone acetate (DMPA) prior to discharge in women who delivered infants who weighed 1,500 g or less at 32 weeks’ or less gestation. Time to secretory activation was longer in 29 women who received DMPA (103.7 hours) compared with 141 women who did not (88.6 hours; P = .028); however, there was no difference in daily milk production, lactation duration, or infant consumption of mother’s own milk.15

While the overall evidence suggests that early initiation of hormonal contraception does not affect breastfeeding or infant outcomes, it is important for clinicians to recognize the limitations of available data with regard to the populations included in these studies. Specifically, most studies did not include individuals with premature, low birth weight, or multiple gestation infants, who are at higher risk of impaired lactation, and individuals with a higher prevalence of breastfeeding were not included to determine whether early initiation of hormonal contraception would impact breastfeeding. Furthermore, while these studies enrolled participants who planned to breastfeed, data indicate that intentions to initiate and continue exclusive breastfeeding can vary.16 As the reported rates of any and exclusive breastfeeding are consistent with or lower than current US breastfeeding rates, any decrease in breastfeeding exclusivity or duration that may be attributable to hormonal contraception may be unacceptable to those who are strongly motivated to breastfeed.

Continue to: How can clinicians integrate evidence into contraception counseling?...

 

 

How can clinicians integrate evidence into contraception counseling?

The American College of Obstetricians and Gynecologists and the Academy of Breastfeeding Medicine offer guidance for how clinicians can address the use of hormonal contraception in breastfeeding patients. Both organizations recommend discussing the risks and benefits of hormonal contraception within the context of each person’s desire to breastfeed, potential for breastfeeding difficulty, and risk of pregnancy so that individuals can make their own informed decisions.17,18

Obstetric care clinicians have an important role in helping patients make informed infant feeding decisions without coercion or pressure. To start these discussions, clinicians can begin by assessing a patient’s breastfeeding goals by asking open-ended questions, such as:

  • What have you heard about breastfeeding?
  • What are your plans for returning to work or school after delivery?
  • How did breastfeeding go with older children?
  • What are your plans for feeding this baby?

In addition to gathering information about the patient’s priorities and goals, clinicians should identify any risk factors for breastfeeding challenges in the medical, surgical, or previous breastfeeding history. Clinicians can engage in a patient-centered approach to infant feeding decisions by anticipating any challenges and working together to develop strategies to address these challenges with the patient’s goals in mind.17

 

When counseling about contraception, a spectrum of approaches exists, from a nondirective information-sharing only model to directive counseling by the clinician. The shared decision-making model lies between these 2 approaches and recognizes the expertise of both the clinician and patient.19 To start these interactions, clinicians can ask about a patient’s reproductive goals by assessing the patient’s needs, values, and preferences for contraception. Potential questions include:

  • What kinds of contraceptive methods have you used in the past?
  • What is important to you in a contraceptive method?
  • How important is it to you to avoid another pregnancy right now?

Clinicians can then share information about different contraceptive methods based on the desired qualities that the patient has identified and how each method fits or does not fit into the patient’s goals and preferences. This collaborative approach facilitates an open dialogue and supports patient autonomy in contraceptive decision-making.

Lastly, clinicians should be cognizant of their own potential biases that could affect their counseling, such as encouraging contraceptive use because of a patient’s young age, parity, or premature delivery, as in our case presentation. Similarly, clinicians also should recognize that breastfeeding and contraceptive decisions are personal and are made with cultural, historical, and social contexts in mind.20 Ultimately, counseling should be patient centered and individualized for each person’s priorities related to infant feeding and pregnancy prevention. ●

 

CASE Patient concerned about hormonal contraception’s impact on lactation

A 19-year-old woman (G2P1102) is postpartum day 1 after delivering a baby at 26 weeks’ gestation. When you see her on postpartum rounds, she states that she does not want any hormonal contraception because she heard that it will decrease her milk supply. What are your next steps?
 

The American Academy of Pediatrics recently updated its policy statement on breastfeeding and the use of human milk to recommend exclusive breastfeeding for 6 months and continued breastfeeding, with complementary foods, as mutually desired for 2 years or beyond given evidence of maternal health benefits with breastfeeding longer than 1 year.1

Breastfeeding prevalence—and challenges

Despite maternal and infant benefits associated with lactation, current breastfeeding prevalence in the United States remains suboptimal. In 2019, 24.9% of infants were exclusively breastfed through 6 months and 35.9% were breastfeeding at 12 months.2 Furthermore, disparities in breastfeeding exist, which contribute to health inequities. For example, non-Hispanic Black infants had lower rates of exclusive breastfeeding at 6 months (19.1%) and any breastfeeding at 12 months (24.1%) compared with non-Hispanic White infants (26.9% and 39.4%, respectively).3

While many new mothers intend to breastfeed and initiate breastfeeding in the hospital after delivery, overall and exclusive breastfeeding continuation rates are low, indicating that patients face challenges with breastfeeding after hospital discharge. Many structural and societal barriers to breastfeeding exist, including inadequate social support and parental leave policies.4 Suboptimal maternity care practices during the birth hospitalization may lead to challenges with breastfeeding initiation. Health care practitioners may present additional barriers to breastfeeding due to a lack of knowledge of available resources for patients or incomplete training in breastfeeding counseling and support.

To address our case patient’s concerns, clinicians should be aware of how exogenous progestins may affect breastfeeding physiology, risk factors for breastfeeding difficulty, and the available evidence for safety of hormonal contraception use while breastfeeding.

ILLUSTRATION: KIMBERLY MARTENS FOR OBG MANAGEMENT

Physiology of breastfeeding

During the second half of pregnancy, secretory differentiation (lactogenesis I) of mammary alveolar epithelial cells into secretory cells occurs to allow the mammary gland to eventually produce milk.5 After delivery of the placenta, progesterone withdrawal triggers secretory activation (lactogenesis II), which refers to the onset of copious milk production within 2 to 3 days postpartum.5 Most patients experience secretory activation within 72 hours; however, a delay in secretory activation past 72 hours is associated with cessation of any and exclusive breastfeeding at 4 weeks postpartum.6

Impaired lactation can be related to a delay in secretory activation or to insufficient lactation related to low milk supply. Maternal medical comorbidities (for example, diabetes mellitus, thyroid dysfunction, obesity), breast anatomy (such as insufficient glandular tissue, prior breast reduction surgery), pregnancy-related events (preeclampsia, retained placenta, postpartum hemorrhage), and infant conditions (such as multiple gestation, premature birth, congenital anomalies) all contribute to a risk of impaired lactation.7

 

Guidance on breastfeeding and hormonal contraception initiation

Early initiation of hormonal contraception poses theoretical concerns about breastfeeding difficulty if exogenous progestin interferes with endogenous signals for onset of milk production. The Centers for Disease Control and Prevention US Medical Eligibility Criteria (MEC) for Contraceptive Use provide recommendations on the safety of contraceptive use in the setting of various medical conditions or patient characteristics based on available data. The MEC uses 4 categories in assessing the safety of contraceptive method use for individuals with specific medical conditions or characteristics: 1, no restrictions exist for use of the contraceptive method; 2, advantages generally outweigh theoretical or proven risks; 3, theoretical or proven risks usually outweigh the advantages; and 4, conditions that represent an unacceptable health risk if the method is used.8

In the 2016 guidelines, combined hormonal contraceptives are considered category 4 at less than 21 days postpartum, regardless of breastfeeding status, due to the increased risk of venous thromboembolism in the immediate postpartum period (TABLE 1).8 Progestin-only contraception is considered category 1 in nonbreastfeeding individuals and category 2 in breastfeeding individuals based on overall evidence that found no adverse outcome with breastfeeding or infant outcomes with early initiation of progestin-only contraception (TABLE 1, TABLE 2).8

 

Since the publication of the 2016 MEC guidelines, several studies have continued to examine breastfeeding and infant outcomes with early initiation of hormonal contraception.

  • In a noninferiority randomized controlled trial of immediate versus delayed initiation of a levonorgestrel intrauterine device (LNG IUD), any breastfeeding at 8 weeks in the immediate group was 78% (95% confidence interval [CI], 70%–85%), which was lower than but within the specified noninferiority margin of the delayed breastfeeding group (83%; 95% CI, 75%–90%), indicating that breastfeeding outcomes with immediate initiation of an LNG IUD were not worse compared with delayed initiation.9
  • A secondary analysis of a randomized trial that compared intracesarean versus LNG IUD placement at 6 or more weeks postpartum showed no difference in breastfeeding at 6, 12, and 24 weeks after LNG IUD placement.10
  • A randomized trial of early (up to 48 hours postpartum) versus placement of an etonogestrel (ENG) implant at 6 or more weeks postpartum showed no difference between groups in infant weight at 12 months.11
  • A randomized trial of immediate (within 5 days of delivery) or interval placement of the 2-rod LNG implant (not approved in the United States) showed no difference in change in infant weight from birth to 6 months after delivery, onset of secretory activation, or breastfeeding continuation at 3 and 6 months postpartum.12
  • In a prospective cohort study that compared immediate postpartum initiation of ENG versus a 2-rod LNG implant (approved by the FDA but not marketed in the United States), there were no differences in breastfeeding continuation at 24 months and exclusive breastfeeding at 6 months postpartum.13
  • In a noninferiority randomized controlled trial that compared ENG implant initiation in the delivery room (0–2 hours postdelivery) versus delayed initiation (24–48 hours postdelivery), the time to secretory activation in those who initiated an ENG implant in the delivery room (66.8 [SD, 25.2] hours) was noninferior to delayed initiation (66.0 [SD, 35.3] hours). There also was no difference in ongoing breastfeeding over the first year after delivery and implant use at 12 months.14
  • A secondary analysis of a randomized controlled trial examined breastfeeding outcomes with receipt of depot medroxyprogesterone acetate (DMPA) prior to discharge in women who delivered infants who weighed 1,500 g or less at 32 weeks’ or less gestation. Time to secretory activation was longer in 29 women who received DMPA (103.7 hours) compared with 141 women who did not (88.6 hours; P = .028); however, there was no difference in daily milk production, lactation duration, or infant consumption of mother’s own milk.15

While the overall evidence suggests that early initiation of hormonal contraception does not affect breastfeeding or infant outcomes, it is important for clinicians to recognize the limitations of available data with regard to the populations included in these studies. Specifically, most studies did not include individuals with premature, low birth weight, or multiple gestation infants, who are at higher risk of impaired lactation, and individuals with a higher prevalence of breastfeeding were not included to determine whether early initiation of hormonal contraception would impact breastfeeding. Furthermore, while these studies enrolled participants who planned to breastfeed, data indicate that intentions to initiate and continue exclusive breastfeeding can vary.16 As the reported rates of any and exclusive breastfeeding are consistent with or lower than current US breastfeeding rates, any decrease in breastfeeding exclusivity or duration that may be attributable to hormonal contraception may be unacceptable to those who are strongly motivated to breastfeed.

Continue to: How can clinicians integrate evidence into contraception counseling?...

 

 

How can clinicians integrate evidence into contraception counseling?

The American College of Obstetricians and Gynecologists and the Academy of Breastfeeding Medicine offer guidance for how clinicians can address the use of hormonal contraception in breastfeeding patients. Both organizations recommend discussing the risks and benefits of hormonal contraception within the context of each person’s desire to breastfeed, potential for breastfeeding difficulty, and risk of pregnancy so that individuals can make their own informed decisions.17,18

Obstetric care clinicians have an important role in helping patients make informed infant feeding decisions without coercion or pressure. To start these discussions, clinicians can begin by assessing a patient’s breastfeeding goals by asking open-ended questions, such as:

  • What have you heard about breastfeeding?
  • What are your plans for returning to work or school after delivery?
  • How did breastfeeding go with older children?
  • What are your plans for feeding this baby?

In addition to gathering information about the patient’s priorities and goals, clinicians should identify any risk factors for breastfeeding challenges in the medical, surgical, or previous breastfeeding history. Clinicians can engage in a patient-centered approach to infant feeding decisions by anticipating any challenges and working together to develop strategies to address these challenges with the patient’s goals in mind.17

 

When counseling about contraception, a spectrum of approaches exists, from a nondirective information-sharing only model to directive counseling by the clinician. The shared decision-making model lies between these 2 approaches and recognizes the expertise of both the clinician and patient.19 To start these interactions, clinicians can ask about a patient’s reproductive goals by assessing the patient’s needs, values, and preferences for contraception. Potential questions include:

  • What kinds of contraceptive methods have you used in the past?
  • What is important to you in a contraceptive method?
  • How important is it to you to avoid another pregnancy right now?

Clinicians can then share information about different contraceptive methods based on the desired qualities that the patient has identified and how each method fits or does not fit into the patient’s goals and preferences. This collaborative approach facilitates an open dialogue and supports patient autonomy in contraceptive decision-making.

Lastly, clinicians should be cognizant of their own potential biases that could affect their counseling, such as encouraging contraceptive use because of a patient’s young age, parity, or premature delivery, as in our case presentation. Similarly, clinicians also should recognize that breastfeeding and contraceptive decisions are personal and are made with cultural, historical, and social contexts in mind.20 Ultimately, counseling should be patient centered and individualized for each person’s priorities related to infant feeding and pregnancy prevention. ●

References

 

  1. Meek JY, Noble L; Section on Breastfeeding. Policy statement: breastfeeding and the use of human milk. Pediatrics. 2022;150:e2022057988.
  2. Centers for Disease Control and Prevention. Breastfeeding report card, United States 2022. Accessed November 8, 2022. https://www.cdc.gov/breastfeeding/pdf/2022-Breast feeding-Report-Card-H.pdf
  3. Centers for Disease Control and Prevention. Rates of any and exclusive breastfeeding by sociodemographic characteristic among children born in 2019. Accessed November 8, 2022. https://www.cdc.gov/breastfeeding/data/nis_data/data-files/2019/rates-any-exclusive-bf-socio-dem-2019.html
  4. American College of Obstetricians and Gynecologists. Committee opinion no. 821: barriers to breastfeeding: supporting initiation and continuation of breastfeeding. Obstet Gynecol. 2021;137:e54-e62.
  5. Pang WW, Hartmann PE. Initiation of human lactation: secretory differentiation and secretory activation. J Mammary Gland Biol Neoplasia. 2007;12:211-221.
  6. Brownell E, Howard CR, Lawrence RA, et al. Delayed onset lactogenesis II predicts the cessation of any or exclusive breastfeeding. J Pediatr. 2012;161:608-614.
  7. American College of Obstetricians and Gynecologists. Committee opinion no. 820: breastfeeding challenges. Obstet Gynecol. 2021;137:e42-e53.
  8. Curtis KM, Tepper NK, Jatlaoui TC, et al. US Medical Eligibility Criteria for Contraceptive Use, 2016. MMWR Recomm Rep. 2016;65(RR-3):1-104.
  9. Turok DK, Leeman L, Sanders JN, et al. Immediate postpartum levonorgestrel intrauterine device insertion and breast-feeding outcomes: a noninferiority randomized controlled trial. Am J Obstet Gynecol. 2017;217:665.e1-665.e8.
  10. Levi EE, Findley MK, Avila K, et al. Placement of levonorgestrel intrauterine device at the time of cesarean delivery and the effect on breastfeeding duration. Breastfeed Med. 2018;13:674-679.
  11. Carmo LSMP, Braga GC, Ferriani RA, et al. Timing of etonogestrel-releasing implants and growth of breastfed infants: a randomized controlled trial. Obstet Gynecol. 2017;130:100-107.
  12. Averbach S, Kakaire O, McDiehl R, et al. The effect of immediate postpartum levonorgestrel contraceptive implant use on breastfeeding and infant growth: a randomized controlled trial. Contraception. 2019;99:87-93.
  13. Krashin JW, Lemani C, Nkambule J, et al. A comparison of breastfeeding exclusivity and duration rates between immediate postpartum levonorgestrel versus etonogestrel implant users: a prospective cohort study. Breastfeed Med. 2019;14:69-76.
  14. Henkel A, Lerma K, Reyes G, et al. Lactogenesis and breastfeeding after immediate vs delayed birth-hospitalization insertion of etonogestrel contraceptive implant: a noninferiority trial. Am J Obstet Gynecol. 2023; 228:55.e1-55.e9.
  15. Parker LA, Sullivan S, Cacho N, et al. Effect of postpartum depo medroxyprogesterone acetate on lactation in mothers of very low-birth-weight infants. Breastfeed Med. 2021;16:835-842.
  16. Nommsen-Rivers LA, Dewey KG. Development and validation of the infant feeding intentions scale. Matern Child Health J. 2009;13:334-342.
  17. American College of Obstetricians and Gynecologists. Committee opinion no. 756: optimizing support for breastfeeding as part of obstetric practice. Obstet Gynecol. 2018;132:e187-e196.
  18. Berens P, Labbok M; Academy of Breastfeeding Medicine. ABM Clinical Protocol #13: contraception during breastfeeding, revised 2015. Breastfeed Med. 2015;10:3-12.
  19. American College of Obstetricians and Gynecologists, Committee on Health Care for Underserved Women, Contraceptive Equity Expert Work Group, and Committee on Ethics. Committee statement no. 1: patient-centered contraceptive counseling. Obstet Gynecol. 2022;139:350-353.
  20. Bryant AG, Lyerly AD, DeVane-Johnson S, et al. Hormonal contraception, breastfeeding and bedside advocacy: the case for patient-centered care. Contraception. 2019;99:73-76.
References

 

  1. Meek JY, Noble L; Section on Breastfeeding. Policy statement: breastfeeding and the use of human milk. Pediatrics. 2022;150:e2022057988.
  2. Centers for Disease Control and Prevention. Breastfeeding report card, United States 2022. Accessed November 8, 2022. https://www.cdc.gov/breastfeeding/pdf/2022-Breast feeding-Report-Card-H.pdf
  3. Centers for Disease Control and Prevention. Rates of any and exclusive breastfeeding by sociodemographic characteristic among children born in 2019. Accessed November 8, 2022. https://www.cdc.gov/breastfeeding/data/nis_data/data-files/2019/rates-any-exclusive-bf-socio-dem-2019.html
  4. American College of Obstetricians and Gynecologists. Committee opinion no. 821: barriers to breastfeeding: supporting initiation and continuation of breastfeeding. Obstet Gynecol. 2021;137:e54-e62.
  5. Pang WW, Hartmann PE. Initiation of human lactation: secretory differentiation and secretory activation. J Mammary Gland Biol Neoplasia. 2007;12:211-221.
  6. Brownell E, Howard CR, Lawrence RA, et al. Delayed onset lactogenesis II predicts the cessation of any or exclusive breastfeeding. J Pediatr. 2012;161:608-614.
  7. American College of Obstetricians and Gynecologists. Committee opinion no. 820: breastfeeding challenges. Obstet Gynecol. 2021;137:e42-e53.
  8. Curtis KM, Tepper NK, Jatlaoui TC, et al. US Medical Eligibility Criteria for Contraceptive Use, 2016. MMWR Recomm Rep. 2016;65(RR-3):1-104.
  9. Turok DK, Leeman L, Sanders JN, et al. Immediate postpartum levonorgestrel intrauterine device insertion and breast-feeding outcomes: a noninferiority randomized controlled trial. Am J Obstet Gynecol. 2017;217:665.e1-665.e8.
  10. Levi EE, Findley MK, Avila K, et al. Placement of levonorgestrel intrauterine device at the time of cesarean delivery and the effect on breastfeeding duration. Breastfeed Med. 2018;13:674-679.
  11. Carmo LSMP, Braga GC, Ferriani RA, et al. Timing of etonogestrel-releasing implants and growth of breastfed infants: a randomized controlled trial. Obstet Gynecol. 2017;130:100-107.
  12. Averbach S, Kakaire O, McDiehl R, et al. The effect of immediate postpartum levonorgestrel contraceptive implant use on breastfeeding and infant growth: a randomized controlled trial. Contraception. 2019;99:87-93.
  13. Krashin JW, Lemani C, Nkambule J, et al. A comparison of breastfeeding exclusivity and duration rates between immediate postpartum levonorgestrel versus etonogestrel implant users: a prospective cohort study. Breastfeed Med. 2019;14:69-76.
  14. Henkel A, Lerma K, Reyes G, et al. Lactogenesis and breastfeeding after immediate vs delayed birth-hospitalization insertion of etonogestrel contraceptive implant: a noninferiority trial. Am J Obstet Gynecol. 2023; 228:55.e1-55.e9.
  15. Parker LA, Sullivan S, Cacho N, et al. Effect of postpartum depo medroxyprogesterone acetate on lactation in mothers of very low-birth-weight infants. Breastfeed Med. 2021;16:835-842.
  16. Nommsen-Rivers LA, Dewey KG. Development and validation of the infant feeding intentions scale. Matern Child Health J. 2009;13:334-342.
  17. American College of Obstetricians and Gynecologists. Committee opinion no. 756: optimizing support for breastfeeding as part of obstetric practice. Obstet Gynecol. 2018;132:e187-e196.
  18. Berens P, Labbok M; Academy of Breastfeeding Medicine. ABM Clinical Protocol #13: contraception during breastfeeding, revised 2015. Breastfeed Med. 2015;10:3-12.
  19. American College of Obstetricians and Gynecologists, Committee on Health Care for Underserved Women, Contraceptive Equity Expert Work Group, and Committee on Ethics. Committee statement no. 1: patient-centered contraceptive counseling. Obstet Gynecol. 2022;139:350-353.
  20. Bryant AG, Lyerly AD, DeVane-Johnson S, et al. Hormonal contraception, breastfeeding and bedside advocacy: the case for patient-centered care. Contraception. 2019;99:73-76.
Issue
OBG Management - 35(2)
Issue
OBG Management - 35(2)
Page Number
46-51
Page Number
46-51
Publications
Publications
Topics
Article Type
Sections
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.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Progress in breast cancer screening over the past 50 years: A remarkable story, but still work to do

Article Type
Changed
Tue, 02/28/2023 - 12:05

 

Meaningful progress has been made in reducing deaths due to breast cancer over the last half century, with a 43% decrease in mortality rate (breast cancer deaths per 100,000 population).1 Screening mammography (SM) has contributed greatly to that success, accounting for 30% to 70% of the reduced mortality rate, with the remainder due to advancements in breast cancer treatment.2 Despite these improvements, invasive breast cancer remains the highest incident cancer in the United States and in the world, is the second leading cause of cancer death in the United States, and results in more years of life lost than any other cancer (TABLE 1).1,3

While the benefits and harms of SM are reasonably well understood, different guidelines groups have approached the relative value of the risks and benefits differently, which has led to challenges in implementation of shared decision making, particularly around the age to initiate routine screening.4-6 In this article, we will focus on the data behind the controversy, current gaps in knowledge, challenges related to breast density and screening in diverse groups, and emerging technologies to address these gaps and provide a construct for appropriate counseling of the patient across the risk spectrum.

New series on cancer screening

In recognition of 35 years of publication of OBG Management, this article on breast cancer screening by Mark D. Pearlman, MD, kicks off a series that focuses on various cancer screening modalities and expert recommendations.

Stay tuned for articles on the future of cervical cancer screening and genetic testing for cancer risk beyond BRCA testing.

We look forward to continuing OBG Management’s mission of enhancing the quality of reproductive health care and the professional development of ObGyns and all women’s health care clinicians.

 

Breast cancer risk

Variables that affect risk

While female sex and older age are the 2 greatest risks for the development of breast cancer, many other factors can either increase or decrease breast cancer risk in a person’s lifetime. The importance of identifying risk factors is 3-fold:

  1. to perform risk assessment to determine if individuals would benefit from average-risk versus high-risk breast cancer surveillance
  2. to identify persons who might benefit from BRCA genetic counseling and screening, risk reduction medications or procedures, and
  3. to allow patients to determine whether any modification in their lifestyle or reproductive choices would make sense to them to reduce their future breast cancer risk.

Most of these risk variables are largely inalterable (for example, family history of breast cancer, carriage of genetic pathogenic variants such as BRCA1 and BRCA2, age of menarche and menopause), but some are potentially modifiable, such as parity, age at first birth, lactation and duration, and dietary factors, among others. TABLE 2 lists common breast cancer risk factors.

Breast cancer risk assessment

Several validated tools have been developed to estimate a person’s breast cancer risk (TABLE 3). These tools combine known risk factors and, depending on the specific tool, can provide estimates of 5-year, 10-year, or lifetime risk of breast cancer. Patients at highest risk can benefit from earlier screening, supplemental screening with breast magnetic resonance imaging (MRI), or risk reduction (see the section, “High-risk screening”). Ideally, a risk assessment should be done by age 30 so that patients at high risk can be identified for earlier or more intensive screening and for possible genetic testing in those at risk for carriage of the BRCA or other breast cancer gene pathogenic variants.5,7

Continue to: Breast cancer screening: Efficacy and harms...

 

 

Breast cancer screening: Efficacy and harms

The earliest studies of breast cancer screening with mammography were randomized controlled trials (RCTs) that compared screened and unscreened patients aged 40 to 74. Nearly all the RCTs and numerous well-designed incidence-based and case-control studies have demonstrated that SM results in a clinically and statistically significant reduction in breast cancer mortality (TABLE 4).4,6,8 Since the mid-1980s and continuing to the current day, SM programs are routinely recommended in the United States. In addition to the mortality benefit outlined in TABLE 4, SM also is associated with a need for less invasive treatments if breast cancer is diagnosed.9,10

With several decades of experience, SM programs have demonstrated that multiple harms are associated with SM, including callbacks, false-positive mammograms that result in a benign biopsy, and overdiagnosis of breast cancer (TABLE 4). Overdiagnosis is a mammographic detection of a breast cancer that would not have harmed that woman in her lifetime. Overdiagnosis leads to overtreatment of breast cancers with its attendant side effects, the emotional harms of a breast cancer diagnosis, and the substantial financial cost of cancer treatment. Estimates of overdiagnosis range from 0% to 50%, with the most likely estimate of invasive breast cancer overdiagnosis from SM between 5% and 15%.11-13 Some of these overdiagnosed cancers are due to very slow growing cancers or breast cancers that may even regress. However, the higher rates of overdiagnosis occur in older persons who are screened and in whom competing causes of mortality become more prevalent. It is estimated that overdiagnosis of invasive breast cancer in patients younger than age 60 is less than 1%, but it exceeds 14% in those older than age 80 (TABLE 4).14

A structured approach is needed to counsel patients about SM so that they understand both the substantial benefit (earlier-stage diagnosis, reduced need for treatment, reduced breast cancer and all-cause mortality) and the potential harms (callback, false-positive results, and overdiagnosis). Moreover, the relative balance of the benefits and harms are influenced throughout their lifetime by both aging and changes in their personal and family medical history.

 


Counseling should consider factors beyond just the performance of mammography (sensitivity and specificity), such as the patient’s current health and age (competing causes of mortality), likelihood of developing breast cancer based on risk assessment (more benefit in higher-risk persons), and the individual patient’s values on the importance of the benefits and harms. The differing emphases on mammography performance and the relative value of the benefits and harms have led experts to produce disparate national guideline recommendations (TABLE 5).

Should SM start at age 40, 45, or 50 in average-risk persons?

There is not clear consensus about the age at which to begin to recommend routine SM in patients at average risk. The National Comprehensive Cancer Network (NCCN),7 American Cancer Society (ACS),4 and the US Preventive Services Task Force (USPSTF)5 recommend that those at average risk start SM at age 40, 45, and 50, respectively (TABLE 5). While the guideline groups listed in TABLE 5 agree that there is level 1 evidence that SM reduces breast cancer mortality in the general population for persons starting at age 40, because the incidence of breast cancer is lower in younger persons (TABLE 6),4 the net population-based screening benefit is lower in this group, and the number needed to invite to screening to save a single life due to breast cancer varies.

For patients in their 40s, it is estimated that 1,904 individuals need to be invited to SM to save 1 life, whereas for patients in their 50s, it is 1,339.15 However, for patients in their 40s, the number needed to screen to save 1 life due to breast cancer decreases from 1 in 1,904 if invited to be screened to 1 in 588 if they are actually screened.16 Furthermore, if a patient is diagnosed with breast cancer at age 40–50, the likelihood of dying is reduced at least 22% and perhaps as high as 48% if her cancer was diagnosed on SM compared with an unscreened individual with a symptomatic presentation (for example, palpable mass).4,15,17,18 Another benefit of SM in the fifth decade of life (40s) is the decreased need for more extensive treatment, including a higher risk of need for chemotherapy (odds ratio [OR], 2.81; 95% confidence interval [CI], 1.16–6.84); need for mastectomy (OR, 3.41; 95% CI, 1.36–8.52); and need for axillary lymph node dissection (OR, 5.76; 95% CI, 2.40–13.82) in unscreened (compared with screened) patients diagnosed with breast cancer.10

The harms associated with SM are not inconsequential and include callbacks (approximately 1 in 10), false-positive biopsy (approximately 1 in 100), and overdiagnosis (likely <1% of all breast cancers in persons younger than age 50). Because most patients in their 40s will not develop breast cancer (TABLE 6), the benefit of reduced breast cancer mortality will not be experienced by most in this decade of life, but they are still just as likely to experience a callback, false-positive biopsy, or the possibility of overdiagnosis. Interpretation of this balance on a population level is the crux of the various guideline groups’ development of differing recommendations as to when screening should start. Despite this seeming disagreement, all the guideline groups listed in TABLE 5 concur that persons at average risk for breast cancer should be offered SM if they desire starting at age 40 after a shared decision-making conversation that incorporates the patient’s view on the relative value of the benefits and risks.

Continue to: High-risk screening...

 

 

High-risk screening

Unlike in screening average-risk patients, there is less disagreement about screening in high-risk groups. TABLE 7 outlines the various categories and recommended strategies that qualify for screening at younger ages or more intensive screening. Adding breast MRI to SM in high-risk individuals results in both higher cancer detection rates and less interval breast cancers (cancers diagnosed between screening rounds) diagnosed compared with SM alone.19,20 Interval breast cancer tends to be more aggressive and is used as a surrogate marker for more recognized factors, such as breast cancer mortality. In addition to less interval breast cancers, high-risk patients are more likely to be diagnosed with node-negative disease if screening breast MRI is added to SM.

Long-term mortality benefit studies using MRI have not been conducted due to the prolonged follow-up times needed. Expense, lower specificity compared with mammography (that is, more false-positive results), and need for the use of gadolinium limit more widespread use of breast MRI screening in average-risk persons.

 

Screening in patients with dense breasts

Half of patients undergoing SM in the United States have dense breasts (heterogeneously dense breasts, 40%; extremely dense breasts, 10%). Importantly, increasing breast density is associated with a lower cancer detection rate with SM and is an independent risk factor for developing breast cancer. While most states already require patients to be notified if they have dense breasts identified on SM, the US Food and Drug Administration will soon make breast density patient notification a national standard (see: https://delauro.house.gov/media-center/press-releases/delauro-secures-timeline-fda-rollout-breast-density-notification-rule).

Most of the risk assessment tools listed in TABLE 3 incorporate breast density into their calculation of breast cancer risk. If that calculation places a patient into one of the highest-risk groups (based on additional factors like strong family history of breast cancer, reproductive risk factors, BRCA carriage, and so on), more intensive surveillance should be recommended (TABLE 7).7 However, once these risk calculations are done, most persons with dense breasts will remain in an average-risk category.

Because of the frequency and risks associated with dense breasts, different and alternative strategies have been recommended for screening persons who are at average risk with dense breasts. Supplemental screening with MRI, ultrasonography, contrast-enhanced mammography, and molecular breast imaging are all being considered but have not been studied sufficiently to demonstrate mortality benefit or cost-effectiveness.

Of all the supplemental modalities used to screen patients with dense breasts, MRI has been the best studied. A large RCT in the Netherlands evaluated supplemental MRI screening in persons with extremely dense breasts after a negative mammogram.21 Compared with no supplemental screening, the MRI group had 17 additional cancers detected per 1,000 screened and a 50% reduction in interval breast cancers; in addition, MRI was associated with a positive predictive value of 26% for biopsies. At present, high cost and limited access to standard breast MRI has not allowed its routine use for persons with dense breasts in the United States, but this may change with more experience and more widespread introduction and experience with abbreviated (or rapid) breast MRI in the future (TABLE 8).

Equitable screening

Black persons who are diagnosed with breast cancer have a 40% higher risk of dying than White patients due to multiple factors, including systemic racial factors (implicit and unconscious bias), reduced access to care, and a lower likelihood of receiving standard of care once diagnosed.22-24 In addition, Black patients have twice the likelihood of being diagnosed with triple-negative breast cancers, a biologically more aggressive tumor.22-24 Among Black, Asian, and Hispanic persons diagnosed with breast cancer, one-third are diagnosed younger than age 50, which is higher than for non-Hispanic White persons. Prior to the age of 50, Black, Asian, and Hispanic patients also have a 72% more likelihood of being diagnosed with invasive breast cancer, have a 58% greater risk of advanced-stage disease, and have a 127% higher risk of dying from breast cancer compared with White patients.25,26 Based on all of these factors, delaying SM until age 50 may adversely affect the Black, Asian, and Hispanic populations.

Persons in the LGBTQ+ community do not present for SM as frequently as the general population, often because they feel threatened or unwelcome.27 Clinicians and breast imaging units should review their inclusivity policies and training to provide a welcoming and respectful environment to all persons in an effort to reduce these barriers. While data are limited and largely depend on expert opinion, current recommendations for screening in the transgender patient depend on sex assigned at birth, the type and duration of hormone use, and surgical history. In patients assigned female sex at birth, average-risk and high-risk screening recommendations are similar to those for the general population unless bilateral mastectomy has been performed.28 In transfeminine patients who have used hormones for longer than 5 years, some groups recommend annual screening starting at age 40, although well-designed studies are lacking.29

Continue to: We have done well, can we do better?...

 

 

We have done well, can we do better?

Screening mammography clearly has been an important and effective tool in the effort to reduce breast cancer mortality, but there are clear limitations. These include moderate sensitivity of mammography, particularly in patients with dense breasts, and a specificity that results in either callbacks (10%), breast biopsies for benign disease (1%), or the reality of overdiagnosis, which becomes increasingly important in older patients.

With the introduction of mammography in the mid-1980s, a one-size-fits-all approach has proved challenging more recently due to an increased recognition of the harms of screening. As a result of this evolving understanding, different recommendations for average-risk screening have emerged. With the advent of breast MRI, risk-based screening is an important but underutilized tool to identify highest-risk individuals, which is associated with improved cancer detection rates, reduced node-positive disease, and fewer diagnosed interval breast cancers. Assuring that nearly all of this highest-risk group is identified through routine breast cancer risk assessment remains a challenge for clinicians.

But what SM recommendations should be offered to persons who fall into an intermediate-risk group (15%–20%), very low-risk groups (<5%), or patients with dense breasts? These are challenges that could be met through novel and individualized approaches (for example, polygenic risk scoring, further research on newer modalities of screening [TABLE 8]), improved screening algorithms for persons with dense breasts, and enhanced clinician engagement to achieve universal breast cancer and BRCA risk assessment of patients by age 25 to 30.

In 2023, best practice and consensus guidelines for intermediate- and low-risk breast cancer groups remain unclear, and one of the many ongoing challenges is to further reduce the impact of breast cancer on the lives of persons affected and the recognized harms of SM.

In the meantime, there is consensus in average-risk patients to provide counseling about SM by age 40. My approach has been to counsel all average-risk patients on the risks and benefits of mammography using the acronym TIP-V:

  • Use a Tool to calculate breast cancer risk (TABLE 3). If they are at high risk, provide recommendations for high-risk management (TABLE 7).7
  • For average-risk patients, counsel that their Incidence of developing breast cancer in the next decade is approximately 1 in 70 (TABLE 6).4
  • Provide data and guidance on the benefits of SM for patients in their 40s (mortality improvement, decreased treatment) and the likelihood of harm from breast cancer screening (10% callback, 1% benign biopsy, and <1% likelihood of overdiagnosis [TABLE 4]).4,14,15
  • Engage the patient to better understand their relative Values of the benefits and harms and make a shared decision on screening starting at age 40, 45, or 50.
 

Looking forward

In summary, SM remains an important tool in the effort to decrease the risk of mortality due to breast cancer. Given the limitations of SM, however, newer tools and methods—abbreviated MRI, contrast-enhanced mammography, molecular breast imaging, customized screening intervals depending on individual risk/polygenic risk score, and customized counseling and screening based on risk factors (TABLES 2 and 7)—will play an increased role in recommendations for breast cancer screening in the future. ●

References
  1. Giaquinto AN, Sung H, Miller KD, et al. Breast cancer statistics, 2022. CA Cancer J Clin. 2022;72:524-541.
  2. Berry DA, Cronin KA, Plevritis SK, et al. Effect of screening and adjuvant therapy on mortality from breast cancer. N Engl J Med. 2005;353:1784-1792.
  3. Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209-249.
  4. Oeffinger KC, Fontham ET, Etzioni R, et al; American Cancer Society. Breast cancer screening for women at average risk: 2015 guideline update from the American Cancer Society. JAMA. 2015;314:1599-1614.
  5. US Preventive Services Task Force; Owens DK, Davidson KW, Drist AH, et al. Risk assessment, genetic counseling, and genetic testing for BRCA-related cancer: US Preventive Services Task Force Recommendation statement. JAMA. 2019;322:652-665.
  6. Nelson HD, Cantor A, Humphrey L, et al. Screening for breast cancer: a systematic review to update the 2009 US Preventive Services Task Force recommendation. Evidence synthesis no 124.  AHRQ publication no 14-05201-EF-1. Rockville, MD: Agency for Healthcare Research and Quality; 2016.
  7. Bevers TB, Helvie M, Bonaccio E, et al. Breast cancer screening and diagnosis, version 3.2018, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2018;16:1362-1389.
  8. Duffy SW, Vulkan D, Cuckle H, et al. Effect of mammographic screening from age 40 years on breast cancer mortality (UK Age trial): final results of a randomised, controlled trial. Lancet Oncol. 2020;21:1165-1172.
  9. Karzai S, Port E, Siderides C, et al. Impact of screening mammography on treatment in young women diagnosed with breast cancer. Ann Surg Oncol. 2022. doi:10.1245/ s10434-022-11581-6.
  10. Ahn S, Wooster M, Valente C, et al. Impact of screening mammography on treatment in women diagnosed with breast cancer. Ann Surg Oncol. 2018;25:2979-2986.
  11. Coldman A, Phillips N. Incidence of breast cancer and estimates of overdiagnosis after the initiation of a population-based mammography screening program. CMAJ. 2013;185:E492-E498.
  12. Etzioni R, Gulati R, Mallinger L, et al. Influence of study features and methods on overdiagnosis estimates in breast and prostate cancer screening. Ann Internal Med. 2013;158:831-838.
  13. Ryser MD, Lange J, Inoue LY, et al. Estimation of breast cancer overdiagnosis in a US breast screening cohort. Ann Intern Med. 2022;175:471-478.
  14. Monticciolo DL, Malak SF, Friedewald SM, et al. Breast cancer screening recommendations inclusive of all women at average risk: update from the ACR and Society of Breast Imaging. J Am Coll Radiol. 2021;18:1280-1288.
  15. Nelson HD, Fu R, Cantor A, Pappas M, et al. Effectiveness of breast cancer screening: systematic review and meta-analysis to update the 2009 US Preventive Services Task Force recommendation. Ann Internal Med. 2016;164:244-255.
  16. Hendrick RE, Helvie MA, Hardesty LA. Implications of CISNET modeling on number needed to screen and mortality reduction with digital mammography in women 40–49 years old. Am J Roentgenol. 2014;203:1379-1381.
  17. Broeders M, Moss S, Nyström L, et al; EUROSCREEN Working Group. The impact of mammographic screening on breast cancer mortality in Europe: a review of observational studies. J Med Screen. 2012;19(suppl 1):14-25.
  18. Tabár L, Yen AMF, Wu WYY, et al. Insights from the breast cancer screening trials: how screening affects the natural history of breast cancer and implications for evaluating service screening programs. Breast J. 2015;21:13-20.
  19. Kriege M, Brekelmans CTM, Boetes C, et al; Magnetic Resonance Imaging Screening Study Group. Efficacy of MRI and mammography for breast-cancer screening in women with a familial or genetic predisposition. N Engl J Med. 2004;351:427-437.
  20. Vreemann S, Gubern-Merida A, Lardenoije S, et al. The frequency of missed breast cancers in women participating in a high-risk MRI screening program. Breast Cancer Res Treat. 2018;169:323-331.
  21. Bakker MF, de Lange SV, Pijnappel RM, et al. Supplemental MRI screening for women with extremely dense breast tissue. N Engl J Med. 2019;381:2091-2102.
  22. Amirikia KC, Mills P, Bush J, et al. Higher population‐based incidence rates of triple‐negative breast cancer among young African‐American women: implications for breast cancer screening recommendations. Cancer. 2011;117:2747-2753.
  23. Kohler BA, Sherman RL, Howlader N, et al. Annual report to the nation on the status of cancer, 1975-2011, featuring incidence of breast cancer subtypes by race/ethnicity, poverty, and state. J Natl Cancer Inst. 2015;107:djv048.
  24. Newman LA, Kaljee LM. Health disparities and triple-negative breast cancer in African American women: a review. JAMA Surg. 2017;152:485-493.
  25. Stapleton SM, Oseni TO, Bababekov YJ, et al. Race/ethnicity and age distribution of breast cancer diagnosis in the United States. JAMA Surg. 2018;153:594-595.
  26. Hendrick RE, Monticciolo DL, Biggs KW, et al. Age distributions of breast cancer diagnosis and mortality by race and ethnicity in US women. Cancer. 2021;127:4384-4392.
  27. Perry H, Fang AJ, Tsai EM, et al. Imaging health and radiology care of transgender patients: a call to build evidence-based best practices. J Am Coll Radiol. 2021;18(3 pt B):475-480.
  28. Lockhart R, Kamaya A. Patient-friendly summary of the ACR Appropriateness Criteria: transgender breast cancer screening. J Am Coll Radiol. 2022;19:e19.
  29. Expert Panel on Breast Imaging; Brown A, Lourenco AP, Niell BL, et al. ACR Appropriateness Criteria transgender breast cancer screening. J Am Coll Radiol. 2021;18:S502-S515.
  30. Mørch LS, Skovlund CW, Hannaford PC, et al. Contemporary hormonal contraception and the risk of breast cancer. N Engl J Med. 2017;377:2228-2239.
  31. Siegel RL, Miller KD, Fuchs HE, et al. Cancer statistics, 2021. CA Cancer J Clin. 2021;71:7-33.
  32. Laws A, Katlin F, Hans M, et al. Screening MRI does not increase cancer detection or result in an earlier stage at diagnosis for patients with high-risk breast lesions: a propensity score analysis. Ann Surg Oncol. 2023;30;68-77.
  33. American College of Obstetricians and Gynecologists. Practice bulletin no 179: Breast cancer risk assessment and screening in average-risk women. Obstet Gynecol. 2017;130:e1-e16.
  34. Grimm LJ, Mango VL, Harvey JA, et al. Implementation of abbreviated breast MRI for screening: AJR expert panel narrative review. AJR Am J Roentgenol. 2022;218:202-212.
  35. Potsch N, Vatteroini G, Clauser P, et al. Contrast-enhanced mammography versus contrast-enhanced breast MRI: a systematic review and meta-analysis. Radiology. 2022;305:94-103.
  36. Covington MF, Parent EE, Dibble EH, et al. Advances and future directions in molecular breast imaging. J Nucl Med. 2022;63:17-21.
Article PDF
Author and Disclosure Information

Disclaimer: Gender-neutral terms (“persons,” “people,” “patients,” “individuals,” “they,” etc) are used throughout this article, but the use of screening mammography and other breast cancer screening tools generally references persons who were assigned female sex at birth.

Dr. Pearlman is Professor Emeritus, 
Departments of Obstetrics and 
Gynecology, Department of Surgery, 
University of Michigan Health 
System, Ann Arbor, Michigan.

The author reports no financial relationships relevant to  this article.

Issue
OBG Management - 35(2)
Publications
Topics
Page Number
33-44
Sections
Author and Disclosure Information

Disclaimer: Gender-neutral terms (“persons,” “people,” “patients,” “individuals,” “they,” etc) are used throughout this article, but the use of screening mammography and other breast cancer screening tools generally references persons who were assigned female sex at birth.

Dr. Pearlman is Professor Emeritus, 
Departments of Obstetrics and 
Gynecology, Department of Surgery, 
University of Michigan Health 
System, Ann Arbor, Michigan.

The author reports no financial relationships relevant to  this article.

Author and Disclosure Information

Disclaimer: Gender-neutral terms (“persons,” “people,” “patients,” “individuals,” “they,” etc) are used throughout this article, but the use of screening mammography and other breast cancer screening tools generally references persons who were assigned female sex at birth.

Dr. Pearlman is Professor Emeritus, 
Departments of Obstetrics and 
Gynecology, Department of Surgery, 
University of Michigan Health 
System, Ann Arbor, Michigan.

The author reports no financial relationships relevant to  this article.

Article PDF
Article PDF

 

Meaningful progress has been made in reducing deaths due to breast cancer over the last half century, with a 43% decrease in mortality rate (breast cancer deaths per 100,000 population).1 Screening mammography (SM) has contributed greatly to that success, accounting for 30% to 70% of the reduced mortality rate, with the remainder due to advancements in breast cancer treatment.2 Despite these improvements, invasive breast cancer remains the highest incident cancer in the United States and in the world, is the second leading cause of cancer death in the United States, and results in more years of life lost than any other cancer (TABLE 1).1,3

While the benefits and harms of SM are reasonably well understood, different guidelines groups have approached the relative value of the risks and benefits differently, which has led to challenges in implementation of shared decision making, particularly around the age to initiate routine screening.4-6 In this article, we will focus on the data behind the controversy, current gaps in knowledge, challenges related to breast density and screening in diverse groups, and emerging technologies to address these gaps and provide a construct for appropriate counseling of the patient across the risk spectrum.

New series on cancer screening

In recognition of 35 years of publication of OBG Management, this article on breast cancer screening by Mark D. Pearlman, MD, kicks off a series that focuses on various cancer screening modalities and expert recommendations.

Stay tuned for articles on the future of cervical cancer screening and genetic testing for cancer risk beyond BRCA testing.

We look forward to continuing OBG Management’s mission of enhancing the quality of reproductive health care and the professional development of ObGyns and all women’s health care clinicians.

 

Breast cancer risk

Variables that affect risk

While female sex and older age are the 2 greatest risks for the development of breast cancer, many other factors can either increase or decrease breast cancer risk in a person’s lifetime. The importance of identifying risk factors is 3-fold:

  1. to perform risk assessment to determine if individuals would benefit from average-risk versus high-risk breast cancer surveillance
  2. to identify persons who might benefit from BRCA genetic counseling and screening, risk reduction medications or procedures, and
  3. to allow patients to determine whether any modification in their lifestyle or reproductive choices would make sense to them to reduce their future breast cancer risk.

Most of these risk variables are largely inalterable (for example, family history of breast cancer, carriage of genetic pathogenic variants such as BRCA1 and BRCA2, age of menarche and menopause), but some are potentially modifiable, such as parity, age at first birth, lactation and duration, and dietary factors, among others. TABLE 2 lists common breast cancer risk factors.

Breast cancer risk assessment

Several validated tools have been developed to estimate a person’s breast cancer risk (TABLE 3). These tools combine known risk factors and, depending on the specific tool, can provide estimates of 5-year, 10-year, or lifetime risk of breast cancer. Patients at highest risk can benefit from earlier screening, supplemental screening with breast magnetic resonance imaging (MRI), or risk reduction (see the section, “High-risk screening”). Ideally, a risk assessment should be done by age 30 so that patients at high risk can be identified for earlier or more intensive screening and for possible genetic testing in those at risk for carriage of the BRCA or other breast cancer gene pathogenic variants.5,7

Continue to: Breast cancer screening: Efficacy and harms...

 

 

Breast cancer screening: Efficacy and harms

The earliest studies of breast cancer screening with mammography were randomized controlled trials (RCTs) that compared screened and unscreened patients aged 40 to 74. Nearly all the RCTs and numerous well-designed incidence-based and case-control studies have demonstrated that SM results in a clinically and statistically significant reduction in breast cancer mortality (TABLE 4).4,6,8 Since the mid-1980s and continuing to the current day, SM programs are routinely recommended in the United States. In addition to the mortality benefit outlined in TABLE 4, SM also is associated with a need for less invasive treatments if breast cancer is diagnosed.9,10

With several decades of experience, SM programs have demonstrated that multiple harms are associated with SM, including callbacks, false-positive mammograms that result in a benign biopsy, and overdiagnosis of breast cancer (TABLE 4). Overdiagnosis is a mammographic detection of a breast cancer that would not have harmed that woman in her lifetime. Overdiagnosis leads to overtreatment of breast cancers with its attendant side effects, the emotional harms of a breast cancer diagnosis, and the substantial financial cost of cancer treatment. Estimates of overdiagnosis range from 0% to 50%, with the most likely estimate of invasive breast cancer overdiagnosis from SM between 5% and 15%.11-13 Some of these overdiagnosed cancers are due to very slow growing cancers or breast cancers that may even regress. However, the higher rates of overdiagnosis occur in older persons who are screened and in whom competing causes of mortality become more prevalent. It is estimated that overdiagnosis of invasive breast cancer in patients younger than age 60 is less than 1%, but it exceeds 14% in those older than age 80 (TABLE 4).14

A structured approach is needed to counsel patients about SM so that they understand both the substantial benefit (earlier-stage diagnosis, reduced need for treatment, reduced breast cancer and all-cause mortality) and the potential harms (callback, false-positive results, and overdiagnosis). Moreover, the relative balance of the benefits and harms are influenced throughout their lifetime by both aging and changes in their personal and family medical history.

 


Counseling should consider factors beyond just the performance of mammography (sensitivity and specificity), such as the patient’s current health and age (competing causes of mortality), likelihood of developing breast cancer based on risk assessment (more benefit in higher-risk persons), and the individual patient’s values on the importance of the benefits and harms. The differing emphases on mammography performance and the relative value of the benefits and harms have led experts to produce disparate national guideline recommendations (TABLE 5).

Should SM start at age 40, 45, or 50 in average-risk persons?

There is not clear consensus about the age at which to begin to recommend routine SM in patients at average risk. The National Comprehensive Cancer Network (NCCN),7 American Cancer Society (ACS),4 and the US Preventive Services Task Force (USPSTF)5 recommend that those at average risk start SM at age 40, 45, and 50, respectively (TABLE 5). While the guideline groups listed in TABLE 5 agree that there is level 1 evidence that SM reduces breast cancer mortality in the general population for persons starting at age 40, because the incidence of breast cancer is lower in younger persons (TABLE 6),4 the net population-based screening benefit is lower in this group, and the number needed to invite to screening to save a single life due to breast cancer varies.

For patients in their 40s, it is estimated that 1,904 individuals need to be invited to SM to save 1 life, whereas for patients in their 50s, it is 1,339.15 However, for patients in their 40s, the number needed to screen to save 1 life due to breast cancer decreases from 1 in 1,904 if invited to be screened to 1 in 588 if they are actually screened.16 Furthermore, if a patient is diagnosed with breast cancer at age 40–50, the likelihood of dying is reduced at least 22% and perhaps as high as 48% if her cancer was diagnosed on SM compared with an unscreened individual with a symptomatic presentation (for example, palpable mass).4,15,17,18 Another benefit of SM in the fifth decade of life (40s) is the decreased need for more extensive treatment, including a higher risk of need for chemotherapy (odds ratio [OR], 2.81; 95% confidence interval [CI], 1.16–6.84); need for mastectomy (OR, 3.41; 95% CI, 1.36–8.52); and need for axillary lymph node dissection (OR, 5.76; 95% CI, 2.40–13.82) in unscreened (compared with screened) patients diagnosed with breast cancer.10

The harms associated with SM are not inconsequential and include callbacks (approximately 1 in 10), false-positive biopsy (approximately 1 in 100), and overdiagnosis (likely <1% of all breast cancers in persons younger than age 50). Because most patients in their 40s will not develop breast cancer (TABLE 6), the benefit of reduced breast cancer mortality will not be experienced by most in this decade of life, but they are still just as likely to experience a callback, false-positive biopsy, or the possibility of overdiagnosis. Interpretation of this balance on a population level is the crux of the various guideline groups’ development of differing recommendations as to when screening should start. Despite this seeming disagreement, all the guideline groups listed in TABLE 5 concur that persons at average risk for breast cancer should be offered SM if they desire starting at age 40 after a shared decision-making conversation that incorporates the patient’s view on the relative value of the benefits and risks.

Continue to: High-risk screening...

 

 

High-risk screening

Unlike in screening average-risk patients, there is less disagreement about screening in high-risk groups. TABLE 7 outlines the various categories and recommended strategies that qualify for screening at younger ages or more intensive screening. Adding breast MRI to SM in high-risk individuals results in both higher cancer detection rates and less interval breast cancers (cancers diagnosed between screening rounds) diagnosed compared with SM alone.19,20 Interval breast cancer tends to be more aggressive and is used as a surrogate marker for more recognized factors, such as breast cancer mortality. In addition to less interval breast cancers, high-risk patients are more likely to be diagnosed with node-negative disease if screening breast MRI is added to SM.

Long-term mortality benefit studies using MRI have not been conducted due to the prolonged follow-up times needed. Expense, lower specificity compared with mammography (that is, more false-positive results), and need for the use of gadolinium limit more widespread use of breast MRI screening in average-risk persons.

 

Screening in patients with dense breasts

Half of patients undergoing SM in the United States have dense breasts (heterogeneously dense breasts, 40%; extremely dense breasts, 10%). Importantly, increasing breast density is associated with a lower cancer detection rate with SM and is an independent risk factor for developing breast cancer. While most states already require patients to be notified if they have dense breasts identified on SM, the US Food and Drug Administration will soon make breast density patient notification a national standard (see: https://delauro.house.gov/media-center/press-releases/delauro-secures-timeline-fda-rollout-breast-density-notification-rule).

Most of the risk assessment tools listed in TABLE 3 incorporate breast density into their calculation of breast cancer risk. If that calculation places a patient into one of the highest-risk groups (based on additional factors like strong family history of breast cancer, reproductive risk factors, BRCA carriage, and so on), more intensive surveillance should be recommended (TABLE 7).7 However, once these risk calculations are done, most persons with dense breasts will remain in an average-risk category.

Because of the frequency and risks associated with dense breasts, different and alternative strategies have been recommended for screening persons who are at average risk with dense breasts. Supplemental screening with MRI, ultrasonography, contrast-enhanced mammography, and molecular breast imaging are all being considered but have not been studied sufficiently to demonstrate mortality benefit or cost-effectiveness.

Of all the supplemental modalities used to screen patients with dense breasts, MRI has been the best studied. A large RCT in the Netherlands evaluated supplemental MRI screening in persons with extremely dense breasts after a negative mammogram.21 Compared with no supplemental screening, the MRI group had 17 additional cancers detected per 1,000 screened and a 50% reduction in interval breast cancers; in addition, MRI was associated with a positive predictive value of 26% for biopsies. At present, high cost and limited access to standard breast MRI has not allowed its routine use for persons with dense breasts in the United States, but this may change with more experience and more widespread introduction and experience with abbreviated (or rapid) breast MRI in the future (TABLE 8).

Equitable screening

Black persons who are diagnosed with breast cancer have a 40% higher risk of dying than White patients due to multiple factors, including systemic racial factors (implicit and unconscious bias), reduced access to care, and a lower likelihood of receiving standard of care once diagnosed.22-24 In addition, Black patients have twice the likelihood of being diagnosed with triple-negative breast cancers, a biologically more aggressive tumor.22-24 Among Black, Asian, and Hispanic persons diagnosed with breast cancer, one-third are diagnosed younger than age 50, which is higher than for non-Hispanic White persons. Prior to the age of 50, Black, Asian, and Hispanic patients also have a 72% more likelihood of being diagnosed with invasive breast cancer, have a 58% greater risk of advanced-stage disease, and have a 127% higher risk of dying from breast cancer compared with White patients.25,26 Based on all of these factors, delaying SM until age 50 may adversely affect the Black, Asian, and Hispanic populations.

Persons in the LGBTQ+ community do not present for SM as frequently as the general population, often because they feel threatened or unwelcome.27 Clinicians and breast imaging units should review their inclusivity policies and training to provide a welcoming and respectful environment to all persons in an effort to reduce these barriers. While data are limited and largely depend on expert opinion, current recommendations for screening in the transgender patient depend on sex assigned at birth, the type and duration of hormone use, and surgical history. In patients assigned female sex at birth, average-risk and high-risk screening recommendations are similar to those for the general population unless bilateral mastectomy has been performed.28 In transfeminine patients who have used hormones for longer than 5 years, some groups recommend annual screening starting at age 40, although well-designed studies are lacking.29

Continue to: We have done well, can we do better?...

 

 

We have done well, can we do better?

Screening mammography clearly has been an important and effective tool in the effort to reduce breast cancer mortality, but there are clear limitations. These include moderate sensitivity of mammography, particularly in patients with dense breasts, and a specificity that results in either callbacks (10%), breast biopsies for benign disease (1%), or the reality of overdiagnosis, which becomes increasingly important in older patients.

With the introduction of mammography in the mid-1980s, a one-size-fits-all approach has proved challenging more recently due to an increased recognition of the harms of screening. As a result of this evolving understanding, different recommendations for average-risk screening have emerged. With the advent of breast MRI, risk-based screening is an important but underutilized tool to identify highest-risk individuals, which is associated with improved cancer detection rates, reduced node-positive disease, and fewer diagnosed interval breast cancers. Assuring that nearly all of this highest-risk group is identified through routine breast cancer risk assessment remains a challenge for clinicians.

But what SM recommendations should be offered to persons who fall into an intermediate-risk group (15%–20%), very low-risk groups (<5%), or patients with dense breasts? These are challenges that could be met through novel and individualized approaches (for example, polygenic risk scoring, further research on newer modalities of screening [TABLE 8]), improved screening algorithms for persons with dense breasts, and enhanced clinician engagement to achieve universal breast cancer and BRCA risk assessment of patients by age 25 to 30.

In 2023, best practice and consensus guidelines for intermediate- and low-risk breast cancer groups remain unclear, and one of the many ongoing challenges is to further reduce the impact of breast cancer on the lives of persons affected and the recognized harms of SM.

In the meantime, there is consensus in average-risk patients to provide counseling about SM by age 40. My approach has been to counsel all average-risk patients on the risks and benefits of mammography using the acronym TIP-V:

  • Use a Tool to calculate breast cancer risk (TABLE 3). If they are at high risk, provide recommendations for high-risk management (TABLE 7).7
  • For average-risk patients, counsel that their Incidence of developing breast cancer in the next decade is approximately 1 in 70 (TABLE 6).4
  • Provide data and guidance on the benefits of SM for patients in their 40s (mortality improvement, decreased treatment) and the likelihood of harm from breast cancer screening (10% callback, 1% benign biopsy, and <1% likelihood of overdiagnosis [TABLE 4]).4,14,15
  • Engage the patient to better understand their relative Values of the benefits and harms and make a shared decision on screening starting at age 40, 45, or 50.
 

Looking forward

In summary, SM remains an important tool in the effort to decrease the risk of mortality due to breast cancer. Given the limitations of SM, however, newer tools and methods—abbreviated MRI, contrast-enhanced mammography, molecular breast imaging, customized screening intervals depending on individual risk/polygenic risk score, and customized counseling and screening based on risk factors (TABLES 2 and 7)—will play an increased role in recommendations for breast cancer screening in the future. ●

 

Meaningful progress has been made in reducing deaths due to breast cancer over the last half century, with a 43% decrease in mortality rate (breast cancer deaths per 100,000 population).1 Screening mammography (SM) has contributed greatly to that success, accounting for 30% to 70% of the reduced mortality rate, with the remainder due to advancements in breast cancer treatment.2 Despite these improvements, invasive breast cancer remains the highest incident cancer in the United States and in the world, is the second leading cause of cancer death in the United States, and results in more years of life lost than any other cancer (TABLE 1).1,3

While the benefits and harms of SM are reasonably well understood, different guidelines groups have approached the relative value of the risks and benefits differently, which has led to challenges in implementation of shared decision making, particularly around the age to initiate routine screening.4-6 In this article, we will focus on the data behind the controversy, current gaps in knowledge, challenges related to breast density and screening in diverse groups, and emerging technologies to address these gaps and provide a construct for appropriate counseling of the patient across the risk spectrum.

New series on cancer screening

In recognition of 35 years of publication of OBG Management, this article on breast cancer screening by Mark D. Pearlman, MD, kicks off a series that focuses on various cancer screening modalities and expert recommendations.

Stay tuned for articles on the future of cervical cancer screening and genetic testing for cancer risk beyond BRCA testing.

We look forward to continuing OBG Management’s mission of enhancing the quality of reproductive health care and the professional development of ObGyns and all women’s health care clinicians.

 

Breast cancer risk

Variables that affect risk

While female sex and older age are the 2 greatest risks for the development of breast cancer, many other factors can either increase or decrease breast cancer risk in a person’s lifetime. The importance of identifying risk factors is 3-fold:

  1. to perform risk assessment to determine if individuals would benefit from average-risk versus high-risk breast cancer surveillance
  2. to identify persons who might benefit from BRCA genetic counseling and screening, risk reduction medications or procedures, and
  3. to allow patients to determine whether any modification in their lifestyle or reproductive choices would make sense to them to reduce their future breast cancer risk.

Most of these risk variables are largely inalterable (for example, family history of breast cancer, carriage of genetic pathogenic variants such as BRCA1 and BRCA2, age of menarche and menopause), but some are potentially modifiable, such as parity, age at first birth, lactation and duration, and dietary factors, among others. TABLE 2 lists common breast cancer risk factors.

Breast cancer risk assessment

Several validated tools have been developed to estimate a person’s breast cancer risk (TABLE 3). These tools combine known risk factors and, depending on the specific tool, can provide estimates of 5-year, 10-year, or lifetime risk of breast cancer. Patients at highest risk can benefit from earlier screening, supplemental screening with breast magnetic resonance imaging (MRI), or risk reduction (see the section, “High-risk screening”). Ideally, a risk assessment should be done by age 30 so that patients at high risk can be identified for earlier or more intensive screening and for possible genetic testing in those at risk for carriage of the BRCA or other breast cancer gene pathogenic variants.5,7

Continue to: Breast cancer screening: Efficacy and harms...

 

 

Breast cancer screening: Efficacy and harms

The earliest studies of breast cancer screening with mammography were randomized controlled trials (RCTs) that compared screened and unscreened patients aged 40 to 74. Nearly all the RCTs and numerous well-designed incidence-based and case-control studies have demonstrated that SM results in a clinically and statistically significant reduction in breast cancer mortality (TABLE 4).4,6,8 Since the mid-1980s and continuing to the current day, SM programs are routinely recommended in the United States. In addition to the mortality benefit outlined in TABLE 4, SM also is associated with a need for less invasive treatments if breast cancer is diagnosed.9,10

With several decades of experience, SM programs have demonstrated that multiple harms are associated with SM, including callbacks, false-positive mammograms that result in a benign biopsy, and overdiagnosis of breast cancer (TABLE 4). Overdiagnosis is a mammographic detection of a breast cancer that would not have harmed that woman in her lifetime. Overdiagnosis leads to overtreatment of breast cancers with its attendant side effects, the emotional harms of a breast cancer diagnosis, and the substantial financial cost of cancer treatment. Estimates of overdiagnosis range from 0% to 50%, with the most likely estimate of invasive breast cancer overdiagnosis from SM between 5% and 15%.11-13 Some of these overdiagnosed cancers are due to very slow growing cancers or breast cancers that may even regress. However, the higher rates of overdiagnosis occur in older persons who are screened and in whom competing causes of mortality become more prevalent. It is estimated that overdiagnosis of invasive breast cancer in patients younger than age 60 is less than 1%, but it exceeds 14% in those older than age 80 (TABLE 4).14

A structured approach is needed to counsel patients about SM so that they understand both the substantial benefit (earlier-stage diagnosis, reduced need for treatment, reduced breast cancer and all-cause mortality) and the potential harms (callback, false-positive results, and overdiagnosis). Moreover, the relative balance of the benefits and harms are influenced throughout their lifetime by both aging and changes in their personal and family medical history.

 


Counseling should consider factors beyond just the performance of mammography (sensitivity and specificity), such as the patient’s current health and age (competing causes of mortality), likelihood of developing breast cancer based on risk assessment (more benefit in higher-risk persons), and the individual patient’s values on the importance of the benefits and harms. The differing emphases on mammography performance and the relative value of the benefits and harms have led experts to produce disparate national guideline recommendations (TABLE 5).

Should SM start at age 40, 45, or 50 in average-risk persons?

There is not clear consensus about the age at which to begin to recommend routine SM in patients at average risk. The National Comprehensive Cancer Network (NCCN),7 American Cancer Society (ACS),4 and the US Preventive Services Task Force (USPSTF)5 recommend that those at average risk start SM at age 40, 45, and 50, respectively (TABLE 5). While the guideline groups listed in TABLE 5 agree that there is level 1 evidence that SM reduces breast cancer mortality in the general population for persons starting at age 40, because the incidence of breast cancer is lower in younger persons (TABLE 6),4 the net population-based screening benefit is lower in this group, and the number needed to invite to screening to save a single life due to breast cancer varies.

For patients in their 40s, it is estimated that 1,904 individuals need to be invited to SM to save 1 life, whereas for patients in their 50s, it is 1,339.15 However, for patients in their 40s, the number needed to screen to save 1 life due to breast cancer decreases from 1 in 1,904 if invited to be screened to 1 in 588 if they are actually screened.16 Furthermore, if a patient is diagnosed with breast cancer at age 40–50, the likelihood of dying is reduced at least 22% and perhaps as high as 48% if her cancer was diagnosed on SM compared with an unscreened individual with a symptomatic presentation (for example, palpable mass).4,15,17,18 Another benefit of SM in the fifth decade of life (40s) is the decreased need for more extensive treatment, including a higher risk of need for chemotherapy (odds ratio [OR], 2.81; 95% confidence interval [CI], 1.16–6.84); need for mastectomy (OR, 3.41; 95% CI, 1.36–8.52); and need for axillary lymph node dissection (OR, 5.76; 95% CI, 2.40–13.82) in unscreened (compared with screened) patients diagnosed with breast cancer.10

The harms associated with SM are not inconsequential and include callbacks (approximately 1 in 10), false-positive biopsy (approximately 1 in 100), and overdiagnosis (likely <1% of all breast cancers in persons younger than age 50). Because most patients in their 40s will not develop breast cancer (TABLE 6), the benefit of reduced breast cancer mortality will not be experienced by most in this decade of life, but they are still just as likely to experience a callback, false-positive biopsy, or the possibility of overdiagnosis. Interpretation of this balance on a population level is the crux of the various guideline groups’ development of differing recommendations as to when screening should start. Despite this seeming disagreement, all the guideline groups listed in TABLE 5 concur that persons at average risk for breast cancer should be offered SM if they desire starting at age 40 after a shared decision-making conversation that incorporates the patient’s view on the relative value of the benefits and risks.

Continue to: High-risk screening...

 

 

High-risk screening

Unlike in screening average-risk patients, there is less disagreement about screening in high-risk groups. TABLE 7 outlines the various categories and recommended strategies that qualify for screening at younger ages or more intensive screening. Adding breast MRI to SM in high-risk individuals results in both higher cancer detection rates and less interval breast cancers (cancers diagnosed between screening rounds) diagnosed compared with SM alone.19,20 Interval breast cancer tends to be more aggressive and is used as a surrogate marker for more recognized factors, such as breast cancer mortality. In addition to less interval breast cancers, high-risk patients are more likely to be diagnosed with node-negative disease if screening breast MRI is added to SM.

Long-term mortality benefit studies using MRI have not been conducted due to the prolonged follow-up times needed. Expense, lower specificity compared with mammography (that is, more false-positive results), and need for the use of gadolinium limit more widespread use of breast MRI screening in average-risk persons.

 

Screening in patients with dense breasts

Half of patients undergoing SM in the United States have dense breasts (heterogeneously dense breasts, 40%; extremely dense breasts, 10%). Importantly, increasing breast density is associated with a lower cancer detection rate with SM and is an independent risk factor for developing breast cancer. While most states already require patients to be notified if they have dense breasts identified on SM, the US Food and Drug Administration will soon make breast density patient notification a national standard (see: https://delauro.house.gov/media-center/press-releases/delauro-secures-timeline-fda-rollout-breast-density-notification-rule).

Most of the risk assessment tools listed in TABLE 3 incorporate breast density into their calculation of breast cancer risk. If that calculation places a patient into one of the highest-risk groups (based on additional factors like strong family history of breast cancer, reproductive risk factors, BRCA carriage, and so on), more intensive surveillance should be recommended (TABLE 7).7 However, once these risk calculations are done, most persons with dense breasts will remain in an average-risk category.

Because of the frequency and risks associated with dense breasts, different and alternative strategies have been recommended for screening persons who are at average risk with dense breasts. Supplemental screening with MRI, ultrasonography, contrast-enhanced mammography, and molecular breast imaging are all being considered but have not been studied sufficiently to demonstrate mortality benefit or cost-effectiveness.

Of all the supplemental modalities used to screen patients with dense breasts, MRI has been the best studied. A large RCT in the Netherlands evaluated supplemental MRI screening in persons with extremely dense breasts after a negative mammogram.21 Compared with no supplemental screening, the MRI group had 17 additional cancers detected per 1,000 screened and a 50% reduction in interval breast cancers; in addition, MRI was associated with a positive predictive value of 26% for biopsies. At present, high cost and limited access to standard breast MRI has not allowed its routine use for persons with dense breasts in the United States, but this may change with more experience and more widespread introduction and experience with abbreviated (or rapid) breast MRI in the future (TABLE 8).

Equitable screening

Black persons who are diagnosed with breast cancer have a 40% higher risk of dying than White patients due to multiple factors, including systemic racial factors (implicit and unconscious bias), reduced access to care, and a lower likelihood of receiving standard of care once diagnosed.22-24 In addition, Black patients have twice the likelihood of being diagnosed with triple-negative breast cancers, a biologically more aggressive tumor.22-24 Among Black, Asian, and Hispanic persons diagnosed with breast cancer, one-third are diagnosed younger than age 50, which is higher than for non-Hispanic White persons. Prior to the age of 50, Black, Asian, and Hispanic patients also have a 72% more likelihood of being diagnosed with invasive breast cancer, have a 58% greater risk of advanced-stage disease, and have a 127% higher risk of dying from breast cancer compared with White patients.25,26 Based on all of these factors, delaying SM until age 50 may adversely affect the Black, Asian, and Hispanic populations.

Persons in the LGBTQ+ community do not present for SM as frequently as the general population, often because they feel threatened or unwelcome.27 Clinicians and breast imaging units should review their inclusivity policies and training to provide a welcoming and respectful environment to all persons in an effort to reduce these barriers. While data are limited and largely depend on expert opinion, current recommendations for screening in the transgender patient depend on sex assigned at birth, the type and duration of hormone use, and surgical history. In patients assigned female sex at birth, average-risk and high-risk screening recommendations are similar to those for the general population unless bilateral mastectomy has been performed.28 In transfeminine patients who have used hormones for longer than 5 years, some groups recommend annual screening starting at age 40, although well-designed studies are lacking.29

Continue to: We have done well, can we do better?...

 

 

We have done well, can we do better?

Screening mammography clearly has been an important and effective tool in the effort to reduce breast cancer mortality, but there are clear limitations. These include moderate sensitivity of mammography, particularly in patients with dense breasts, and a specificity that results in either callbacks (10%), breast biopsies for benign disease (1%), or the reality of overdiagnosis, which becomes increasingly important in older patients.

With the introduction of mammography in the mid-1980s, a one-size-fits-all approach has proved challenging more recently due to an increased recognition of the harms of screening. As a result of this evolving understanding, different recommendations for average-risk screening have emerged. With the advent of breast MRI, risk-based screening is an important but underutilized tool to identify highest-risk individuals, which is associated with improved cancer detection rates, reduced node-positive disease, and fewer diagnosed interval breast cancers. Assuring that nearly all of this highest-risk group is identified through routine breast cancer risk assessment remains a challenge for clinicians.

But what SM recommendations should be offered to persons who fall into an intermediate-risk group (15%–20%), very low-risk groups (<5%), or patients with dense breasts? These are challenges that could be met through novel and individualized approaches (for example, polygenic risk scoring, further research on newer modalities of screening [TABLE 8]), improved screening algorithms for persons with dense breasts, and enhanced clinician engagement to achieve universal breast cancer and BRCA risk assessment of patients by age 25 to 30.

In 2023, best practice and consensus guidelines for intermediate- and low-risk breast cancer groups remain unclear, and one of the many ongoing challenges is to further reduce the impact of breast cancer on the lives of persons affected and the recognized harms of SM.

In the meantime, there is consensus in average-risk patients to provide counseling about SM by age 40. My approach has been to counsel all average-risk patients on the risks and benefits of mammography using the acronym TIP-V:

  • Use a Tool to calculate breast cancer risk (TABLE 3). If they are at high risk, provide recommendations for high-risk management (TABLE 7).7
  • For average-risk patients, counsel that their Incidence of developing breast cancer in the next decade is approximately 1 in 70 (TABLE 6).4
  • Provide data and guidance on the benefits of SM for patients in their 40s (mortality improvement, decreased treatment) and the likelihood of harm from breast cancer screening (10% callback, 1% benign biopsy, and <1% likelihood of overdiagnosis [TABLE 4]).4,14,15
  • Engage the patient to better understand their relative Values of the benefits and harms and make a shared decision on screening starting at age 40, 45, or 50.
 

Looking forward

In summary, SM remains an important tool in the effort to decrease the risk of mortality due to breast cancer. Given the limitations of SM, however, newer tools and methods—abbreviated MRI, contrast-enhanced mammography, molecular breast imaging, customized screening intervals depending on individual risk/polygenic risk score, and customized counseling and screening based on risk factors (TABLES 2 and 7)—will play an increased role in recommendations for breast cancer screening in the future. ●

References
  1. Giaquinto AN, Sung H, Miller KD, et al. Breast cancer statistics, 2022. CA Cancer J Clin. 2022;72:524-541.
  2. Berry DA, Cronin KA, Plevritis SK, et al. Effect of screening and adjuvant therapy on mortality from breast cancer. N Engl J Med. 2005;353:1784-1792.
  3. Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209-249.
  4. Oeffinger KC, Fontham ET, Etzioni R, et al; American Cancer Society. Breast cancer screening for women at average risk: 2015 guideline update from the American Cancer Society. JAMA. 2015;314:1599-1614.
  5. US Preventive Services Task Force; Owens DK, Davidson KW, Drist AH, et al. Risk assessment, genetic counseling, and genetic testing for BRCA-related cancer: US Preventive Services Task Force Recommendation statement. JAMA. 2019;322:652-665.
  6. Nelson HD, Cantor A, Humphrey L, et al. Screening for breast cancer: a systematic review to update the 2009 US Preventive Services Task Force recommendation. Evidence synthesis no 124.  AHRQ publication no 14-05201-EF-1. Rockville, MD: Agency for Healthcare Research and Quality; 2016.
  7. Bevers TB, Helvie M, Bonaccio E, et al. Breast cancer screening and diagnosis, version 3.2018, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2018;16:1362-1389.
  8. Duffy SW, Vulkan D, Cuckle H, et al. Effect of mammographic screening from age 40 years on breast cancer mortality (UK Age trial): final results of a randomised, controlled trial. Lancet Oncol. 2020;21:1165-1172.
  9. Karzai S, Port E, Siderides C, et al. Impact of screening mammography on treatment in young women diagnosed with breast cancer. Ann Surg Oncol. 2022. doi:10.1245/ s10434-022-11581-6.
  10. Ahn S, Wooster M, Valente C, et al. Impact of screening mammography on treatment in women diagnosed with breast cancer. Ann Surg Oncol. 2018;25:2979-2986.
  11. Coldman A, Phillips N. Incidence of breast cancer and estimates of overdiagnosis after the initiation of a population-based mammography screening program. CMAJ. 2013;185:E492-E498.
  12. Etzioni R, Gulati R, Mallinger L, et al. Influence of study features and methods on overdiagnosis estimates in breast and prostate cancer screening. Ann Internal Med. 2013;158:831-838.
  13. Ryser MD, Lange J, Inoue LY, et al. Estimation of breast cancer overdiagnosis in a US breast screening cohort. Ann Intern Med. 2022;175:471-478.
  14. Monticciolo DL, Malak SF, Friedewald SM, et al. Breast cancer screening recommendations inclusive of all women at average risk: update from the ACR and Society of Breast Imaging. J Am Coll Radiol. 2021;18:1280-1288.
  15. Nelson HD, Fu R, Cantor A, Pappas M, et al. Effectiveness of breast cancer screening: systematic review and meta-analysis to update the 2009 US Preventive Services Task Force recommendation. Ann Internal Med. 2016;164:244-255.
  16. Hendrick RE, Helvie MA, Hardesty LA. Implications of CISNET modeling on number needed to screen and mortality reduction with digital mammography in women 40–49 years old. Am J Roentgenol. 2014;203:1379-1381.
  17. Broeders M, Moss S, Nyström L, et al; EUROSCREEN Working Group. The impact of mammographic screening on breast cancer mortality in Europe: a review of observational studies. J Med Screen. 2012;19(suppl 1):14-25.
  18. Tabár L, Yen AMF, Wu WYY, et al. Insights from the breast cancer screening trials: how screening affects the natural history of breast cancer and implications for evaluating service screening programs. Breast J. 2015;21:13-20.
  19. Kriege M, Brekelmans CTM, Boetes C, et al; Magnetic Resonance Imaging Screening Study Group. Efficacy of MRI and mammography for breast-cancer screening in women with a familial or genetic predisposition. N Engl J Med. 2004;351:427-437.
  20. Vreemann S, Gubern-Merida A, Lardenoije S, et al. The frequency of missed breast cancers in women participating in a high-risk MRI screening program. Breast Cancer Res Treat. 2018;169:323-331.
  21. Bakker MF, de Lange SV, Pijnappel RM, et al. Supplemental MRI screening for women with extremely dense breast tissue. N Engl J Med. 2019;381:2091-2102.
  22. Amirikia KC, Mills P, Bush J, et al. Higher population‐based incidence rates of triple‐negative breast cancer among young African‐American women: implications for breast cancer screening recommendations. Cancer. 2011;117:2747-2753.
  23. Kohler BA, Sherman RL, Howlader N, et al. Annual report to the nation on the status of cancer, 1975-2011, featuring incidence of breast cancer subtypes by race/ethnicity, poverty, and state. J Natl Cancer Inst. 2015;107:djv048.
  24. Newman LA, Kaljee LM. Health disparities and triple-negative breast cancer in African American women: a review. JAMA Surg. 2017;152:485-493.
  25. Stapleton SM, Oseni TO, Bababekov YJ, et al. Race/ethnicity and age distribution of breast cancer diagnosis in the United States. JAMA Surg. 2018;153:594-595.
  26. Hendrick RE, Monticciolo DL, Biggs KW, et al. Age distributions of breast cancer diagnosis and mortality by race and ethnicity in US women. Cancer. 2021;127:4384-4392.
  27. Perry H, Fang AJ, Tsai EM, et al. Imaging health and radiology care of transgender patients: a call to build evidence-based best practices. J Am Coll Radiol. 2021;18(3 pt B):475-480.
  28. Lockhart R, Kamaya A. Patient-friendly summary of the ACR Appropriateness Criteria: transgender breast cancer screening. J Am Coll Radiol. 2022;19:e19.
  29. Expert Panel on Breast Imaging; Brown A, Lourenco AP, Niell BL, et al. ACR Appropriateness Criteria transgender breast cancer screening. J Am Coll Radiol. 2021;18:S502-S515.
  30. Mørch LS, Skovlund CW, Hannaford PC, et al. Contemporary hormonal contraception and the risk of breast cancer. N Engl J Med. 2017;377:2228-2239.
  31. Siegel RL, Miller KD, Fuchs HE, et al. Cancer statistics, 2021. CA Cancer J Clin. 2021;71:7-33.
  32. Laws A, Katlin F, Hans M, et al. Screening MRI does not increase cancer detection or result in an earlier stage at diagnosis for patients with high-risk breast lesions: a propensity score analysis. Ann Surg Oncol. 2023;30;68-77.
  33. American College of Obstetricians and Gynecologists. Practice bulletin no 179: Breast cancer risk assessment and screening in average-risk women. Obstet Gynecol. 2017;130:e1-e16.
  34. Grimm LJ, Mango VL, Harvey JA, et al. Implementation of abbreviated breast MRI for screening: AJR expert panel narrative review. AJR Am J Roentgenol. 2022;218:202-212.
  35. Potsch N, Vatteroini G, Clauser P, et al. Contrast-enhanced mammography versus contrast-enhanced breast MRI: a systematic review and meta-analysis. Radiology. 2022;305:94-103.
  36. Covington MF, Parent EE, Dibble EH, et al. Advances and future directions in molecular breast imaging. J Nucl Med. 2022;63:17-21.
References
  1. Giaquinto AN, Sung H, Miller KD, et al. Breast cancer statistics, 2022. CA Cancer J Clin. 2022;72:524-541.
  2. Berry DA, Cronin KA, Plevritis SK, et al. Effect of screening and adjuvant therapy on mortality from breast cancer. N Engl J Med. 2005;353:1784-1792.
  3. Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209-249.
  4. Oeffinger KC, Fontham ET, Etzioni R, et al; American Cancer Society. Breast cancer screening for women at average risk: 2015 guideline update from the American Cancer Society. JAMA. 2015;314:1599-1614.
  5. US Preventive Services Task Force; Owens DK, Davidson KW, Drist AH, et al. Risk assessment, genetic counseling, and genetic testing for BRCA-related cancer: US Preventive Services Task Force Recommendation statement. JAMA. 2019;322:652-665.
  6. Nelson HD, Cantor A, Humphrey L, et al. Screening for breast cancer: a systematic review to update the 2009 US Preventive Services Task Force recommendation. Evidence synthesis no 124.  AHRQ publication no 14-05201-EF-1. Rockville, MD: Agency for Healthcare Research and Quality; 2016.
  7. Bevers TB, Helvie M, Bonaccio E, et al. Breast cancer screening and diagnosis, version 3.2018, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2018;16:1362-1389.
  8. Duffy SW, Vulkan D, Cuckle H, et al. Effect of mammographic screening from age 40 years on breast cancer mortality (UK Age trial): final results of a randomised, controlled trial. Lancet Oncol. 2020;21:1165-1172.
  9. Karzai S, Port E, Siderides C, et al. Impact of screening mammography on treatment in young women diagnosed with breast cancer. Ann Surg Oncol. 2022. doi:10.1245/ s10434-022-11581-6.
  10. Ahn S, Wooster M, Valente C, et al. Impact of screening mammography on treatment in women diagnosed with breast cancer. Ann Surg Oncol. 2018;25:2979-2986.
  11. Coldman A, Phillips N. Incidence of breast cancer and estimates of overdiagnosis after the initiation of a population-based mammography screening program. CMAJ. 2013;185:E492-E498.
  12. Etzioni R, Gulati R, Mallinger L, et al. Influence of study features and methods on overdiagnosis estimates in breast and prostate cancer screening. Ann Internal Med. 2013;158:831-838.
  13. Ryser MD, Lange J, Inoue LY, et al. Estimation of breast cancer overdiagnosis in a US breast screening cohort. Ann Intern Med. 2022;175:471-478.
  14. Monticciolo DL, Malak SF, Friedewald SM, et al. Breast cancer screening recommendations inclusive of all women at average risk: update from the ACR and Society of Breast Imaging. J Am Coll Radiol. 2021;18:1280-1288.
  15. Nelson HD, Fu R, Cantor A, Pappas M, et al. Effectiveness of breast cancer screening: systematic review and meta-analysis to update the 2009 US Preventive Services Task Force recommendation. Ann Internal Med. 2016;164:244-255.
  16. Hendrick RE, Helvie MA, Hardesty LA. Implications of CISNET modeling on number needed to screen and mortality reduction with digital mammography in women 40–49 years old. Am J Roentgenol. 2014;203:1379-1381.
  17. Broeders M, Moss S, Nyström L, et al; EUROSCREEN Working Group. The impact of mammographic screening on breast cancer mortality in Europe: a review of observational studies. J Med Screen. 2012;19(suppl 1):14-25.
  18. Tabár L, Yen AMF, Wu WYY, et al. Insights from the breast cancer screening trials: how screening affects the natural history of breast cancer and implications for evaluating service screening programs. Breast J. 2015;21:13-20.
  19. Kriege M, Brekelmans CTM, Boetes C, et al; Magnetic Resonance Imaging Screening Study Group. Efficacy of MRI and mammography for breast-cancer screening in women with a familial or genetic predisposition. N Engl J Med. 2004;351:427-437.
  20. Vreemann S, Gubern-Merida A, Lardenoije S, et al. The frequency of missed breast cancers in women participating in a high-risk MRI screening program. Breast Cancer Res Treat. 2018;169:323-331.
  21. Bakker MF, de Lange SV, Pijnappel RM, et al. Supplemental MRI screening for women with extremely dense breast tissue. N Engl J Med. 2019;381:2091-2102.
  22. Amirikia KC, Mills P, Bush J, et al. Higher population‐based incidence rates of triple‐negative breast cancer among young African‐American women: implications for breast cancer screening recommendations. Cancer. 2011;117:2747-2753.
  23. Kohler BA, Sherman RL, Howlader N, et al. Annual report to the nation on the status of cancer, 1975-2011, featuring incidence of breast cancer subtypes by race/ethnicity, poverty, and state. J Natl Cancer Inst. 2015;107:djv048.
  24. Newman LA, Kaljee LM. Health disparities and triple-negative breast cancer in African American women: a review. JAMA Surg. 2017;152:485-493.
  25. Stapleton SM, Oseni TO, Bababekov YJ, et al. Race/ethnicity and age distribution of breast cancer diagnosis in the United States. JAMA Surg. 2018;153:594-595.
  26. Hendrick RE, Monticciolo DL, Biggs KW, et al. Age distributions of breast cancer diagnosis and mortality by race and ethnicity in US women. Cancer. 2021;127:4384-4392.
  27. Perry H, Fang AJ, Tsai EM, et al. Imaging health and radiology care of transgender patients: a call to build evidence-based best practices. J Am Coll Radiol. 2021;18(3 pt B):475-480.
  28. Lockhart R, Kamaya A. Patient-friendly summary of the ACR Appropriateness Criteria: transgender breast cancer screening. J Am Coll Radiol. 2022;19:e19.
  29. Expert Panel on Breast Imaging; Brown A, Lourenco AP, Niell BL, et al. ACR Appropriateness Criteria transgender breast cancer screening. J Am Coll Radiol. 2021;18:S502-S515.
  30. Mørch LS, Skovlund CW, Hannaford PC, et al. Contemporary hormonal contraception and the risk of breast cancer. N Engl J Med. 2017;377:2228-2239.
  31. Siegel RL, Miller KD, Fuchs HE, et al. Cancer statistics, 2021. CA Cancer J Clin. 2021;71:7-33.
  32. Laws A, Katlin F, Hans M, et al. Screening MRI does not increase cancer detection or result in an earlier stage at diagnosis for patients with high-risk breast lesions: a propensity score analysis. Ann Surg Oncol. 2023;30;68-77.
  33. American College of Obstetricians and Gynecologists. Practice bulletin no 179: Breast cancer risk assessment and screening in average-risk women. Obstet Gynecol. 2017;130:e1-e16.
  34. Grimm LJ, Mango VL, Harvey JA, et al. Implementation of abbreviated breast MRI for screening: AJR expert panel narrative review. AJR Am J Roentgenol. 2022;218:202-212.
  35. Potsch N, Vatteroini G, Clauser P, et al. Contrast-enhanced mammography versus contrast-enhanced breast MRI: a systematic review and meta-analysis. Radiology. 2022;305:94-103.
  36. Covington MF, Parent EE, Dibble EH, et al. Advances and future directions in molecular breast imaging. J Nucl Med. 2022;63:17-21.
Issue
OBG Management - 35(2)
Issue
OBG Management - 35(2)
Page Number
33-44
Page Number
33-44
Publications
Publications
Topics
Article Type
Sections
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.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

2023 Update on bone health

Article Type
Changed
Wed, 02/22/2023 - 11:46

 

 

I recently heard a lecture where the speaker quoted this statistic: “A 50-year-old woman who does not currently have heart disease or cancer has a life expectancy of 91.” Hopefully, anyone reading this article already is aware of the fact that as our patients age, hip fracture results in greater morbidity and mortality than early breast cancer. It should be well known to clinicians (and, ultimately, to our patients) that localized breast cancer has a survival rate of 99%,1 whereas hip fracture carries a 21% mortality in the first year after the event.2 In addition, approximately one-third of women who fracture their hip do not have osteoporosis.3 Furthermore, the role of muscle mass, strength, and performance in bone health has become well established.4

With this in mind, a recent encounter with a patient in my clinical practice illustrates what I believe is an increasing problem today. The patient had been on long-term prednisone systemically for polymyalgia rheumatica. Her dual energy x-ray absorptiometry (DXA) bone mass measurements were among the worst osteoporotic numbers I have witnessed. She related to me the “argument” that occurred between her rheumatologist and endocrinologist. One wanted her to use injectable parathyroid hormone analog daily, while the other advised yearly infusion of zoledronic acid. She chose the yearly infusion. I inquired if either physician had mentioned anything to her about using nonskid rugs in the bathroom, grab bars, being careful of black ice, a calcium-rich diet, vitamin D supplementation, good eyesight, illumination so she does not miss a step, mindful walking, and maintaining optimal balance, muscle mass, strength, and performance-enhancing exercise? She replied, “No, just which drug I should take.”

Realize that the goal for our patients should be to avoid the morbidity and mortality associated especially with hip fracture. The goal is not to have a better bone mass measurement on your DXA scan as you age. This is exactly why the name of this column, years ago, was changed from “Update on osteoporosis” to “Update on bone health.” Similarly, in 2021, the NOF (National Osteoporosis Foundation) became the BHOF (Bone Health and Osteoporosis Foundation). Thus, our understanding and interest in bone health should and must go beyond simply bone mass measurement with DXA technology. The articles highlighted in this year’s Update reflect the importance of this concept.

 

Know SERMs’ effects on bone health for appropriate prescribing

Goldstein SR. Selective estrogen receptor modulators and bone health. Climacteric. 2022;25:56-59.

Selective estrogen receptor modulators (SERMs) are synthetic molecules that bind to the estrogen receptor and can have agonistic activity in some tissues and antagonistic activity in others. In a recent article, I reviewed the known data regarding the effects of various SERMs on bone health.5

A rundown on 4 SERMs and their effects on bone

Tamoxifen is approved by the US Food and Drug Administration (FDA) for the prevention and treatment of breast cancer in women with estrogen receptor–positive tumors. The only prospective study of tamoxifen versus placebo in which fracture risk was studied in women at risk for but not diagnosed with breast cancer was the National Surgical Adjuvant Breast and Bowel Project (NSABP) P-1 trial. In this study, more than 13,000 women were randomly assigned to treatment with tamoxifen or placebo, with a primary objective of studying the incidence of invasive breast cancer in these high-risk women. With 7 years of follow-up, women receiving tamoxifen had significantly fewer fractures of the hip, radius, and spine (80 vs 116 in the placebo group), resulting in a combined relative risk (RR) of 0.68 (95% confidence interval [CI], 0.51–0.92).6

Raloxifene, another SERM, was extensively studied in the MORE (Multiple Outcomes of Raloxifene Evaluation) trial.7 This study involved more than 7,700 postmenopausal women with osteoporosis, average age 67. The incidence of first vertebral fracture was decreased from 4.3% with placebo to 1.9% with raloxifene (RR, 0.55; 95% CI, 0.29–0.71), and subsequent vertebral fractures were decreased from 20.2% with placebo to 14.1% with raloxifene (RR, 0.70; 95% CI, 0.60–0.90). In 2007, the FDA approved raloxifene for “reduction in risk of invasive breast cancer in postmenopausal women with osteoporosis” as well as for “postmenopausal women at high risk for invasive breast cancer” based on the Study of Tamoxifen and Raloxifene (STAR) trial that involved almost 20,000 postmenopausal women deemed at high risk for breast cancer.8

The concept of combining an estrogen with a SERM, known as a TSEC (tissue selective estrogen complex) was studied and brought to market as conjugated equine estrogen (CEE) 0.45 mg and bazedoxifene (BZA) 20 mg. CEE and BZA individually have been shown to prevent vertebral fracture.9,10 The combination of BZA and CEE has been shown to improve bone density compared with placebo.11 There are, however, no fracture prevention data for this combination therapy. This was the basis on which the combination agent received regulatory approval for prevention of osteoporosis in postmenopausal women. This combination drug is also FDA approved for treating moderate to severe vasomotor symptoms of menopause.

Ospemifene is yet another SERM that is clinically available, at an oral dose of 60 mg, and is indicated for the treatment of moderate to severe dyspareunia secondary to vulvovaginal atrophy, or genitourinary syndrome of menopause (GSM). Ospemifene effectively reduced bone loss in ovariectomized rats, with activity comparable to estradiol and raloxifene.12 Clinical data from three phase 1 or phase 2 clinical trials revealed that ospemifene 60 mg/day had a positive effect on biochemical markers for bone turnover in healthy postmenopausal women, with significant improvements relative to placebo and effects comparable to those of raloxifene.13 While actual fracture or bone mineral density (BMD) data in postmenopausal women are lacking, there is a good correlation between biochemical markers for bone turnover and occurrence of fracture.14 Women who need treatment for osteoporosis should not be treated with ospemifene, but women who use ospemifene for dyspareunia can expect positive activity on bone metabolism.

 

WHAT THIS EVIDENCE MEANS FOR PRACTICE

SERMs, unlike estrogen, have no class labeling. In fact, in the endometrium and vagina, they have variable effects. To date, however, in postmenopausal women, all SERMs have shown estrogenic activity in bone as well as being antiestrogenic in breast. Tamoxifen, well known for its use in estrogen receptor–positive breast cancer patients, demonstrates positive effects on bone and fracture reduction compared with placebo. Raloxifene is approved for prevention and treatment of osteoporosis and for breast cancer chemoprevention in high-risk patients. The TSEC combination of CEE and the SERM bazedoxifene is approved for treatment of moderate to severe vasomotor symptoms and prevention of osteoporosis. Finally, the SERM ospemifene, approved for treating moderate to severe dyspareunia or dryness due to vulvovaginal atrophy, or GSM, has demonstrated evidence of a positive effect on bone turnover and metabolism. Clinicians need to be aware of these effects when choosing medications for their patients.

 

Continue to: Gut microbiome constituents may influence the development of osteoporosis: A potential treatment target?...

 

 

Gut microbiome constituents may influence the development of osteoporosis: A potential treatment target?

Cronin O, Lanham-New SA, Corfe BM, et al. Role of the microbiome in regulating bone metabolism and susceptibility to osteoporosis. Calcif Tissue Int. 2022;110:273-284.

Yang X, Chang T, Yuan Q, et al. Changes in the composition of gut and vaginal microbiota in patients with postmenopausal osteoporosis. Front Immunol. 2022;13:930244.



The role of the microbiome in many arenas is rapidly emerging. Apparently, its relationship in bone metabolism is still in its infancy. A review of PubMed articles showed that 1 paper was published in 2012, none until 2 more in 2015, with a total of 221 published through November 1, 2022. A recent review by Cronin and colleagues on the microbiome’s role in regulating bone metabolism came out of a workshop held by the Osteoporosis and Bone Research Academy of the Royal Osteoporosis Society in the United Kingdom.15

 

The gut microbiome’s relationship with bone health

The authors noted that the human microbiota functions at the interface between diet, medication use, lifestyle, host immune development, and health. Hence, it is closely aligned with many of the recognized modifiable factors that influence bone mass accrual in the young and bone maintenance and skeletal decline in older populations. Microbiome research and discovery supports a role of the human gut microbiome in the regulation of bone metabolism and the pathogenesis of osteoporosis as well as its prevention and treatment.

Numerous factors which influence the gut microbiome and the development of osteoporosis overlap. These include body mass index (BMI), vitamin D, alcohol intake, diet, corticosteroid use, physical activity, sex hormone deficiency, genetic variability, and chronic inflammatory disorders.

Cronin and colleagues reviewed a number of clinical studies and concluded that “the available evidence suggests that probiotic supplements can attenuate bone loss in postmenopausal women, although the studies investigating this have been short term and individually have had small sample sizes. Moving forward, it will be important to conduct larger scale studies to evaluate if the skeletal response differs with different types of probiotic and also to determine if the effects are sustained in the longer term.”15

Composition of the microbiota

A recent study by Yang and colleagues focused on changes in gut and vaginal microbiota composition in patients with postmenopausal osteoporosis. They analyzed data from 132 postmenopausal women with osteoporosis (n = 34), osteopenia (n = 47), and controls (n = 51) based on their T-scores.16

Significant differences were observed in the microbial compositions of fecal samples between groups (P<.05), with some species enhanced in the control group whereas other species were higher in the osteoporosis group. Similar but less pronounced differences were seen in the vaginal microbiome but of different species.

The authors concluded that “The results show that changes in BMD in postmenopausal women are associated with the changes in gut microbiome and vaginal microbiome; however, changes in gut microbiome are more closely correlated with postmenopausal osteoporosis than vaginal microbiome.”16

 

WHAT THIS EVIDENCE MEANS FOR PRACTICE
While we are not yet ready to try to clinically alter the gut microbiome with various interventions, realizing that there is crosstalk between the gut microbiome and bone health is another factor to consider, and it begins with an appreciation of the various factors where the 2 overlap—BMI, vitamin D, alcohol intake, diet, corticosteroid use, physical activity, sex hormone deficiency, genetic variability, and chronic inflammatory disorders.

Continue to: Sarcopenia, osteoporosis, and frailty: A fracture risk triple play...

 

 

Sarcopenia, osteoporosis, and frailty: A fracture risk triple play

Laskou F, Fuggle NR, Patel HP, et al. Associations of osteoporosis and sarcopenia with frailty and multimorbidity among participants of the Hertfordshire Cohort Study. J Cachexia Sarcopenia Muscle. 2022;13:220-229.

Laskou and colleagues aimed to explore the relationship between sarcopenia, osteoporosis, and frailty in community-dwelling adults participating in a cohort study in the United Kingdom and to determine if the coexistence of osteoporosis and sarcopenia is associated with a significantly heavier health burden.17

 

Study details

The authors examined data from 206 women with an average age of 75.5 years. Sarcopenia was defined using the European Working Group on Sarcopenia in Older People (EWGSOP) criteria, which includes low grip strength or slow chair rise and low muscle quantity. Osteoporosis was defined by standard measurements as a T-score of less than or equal to -2.5 standard deviations at the femoral neck or use of any osteoporosis medications. Frailty was defined using the Fried definition, which includes having 3 or more of the following 5 domains: weakness, slowness, exhaustion, low physical activity, and unintentional weight loss. Having 1 or 2 domains is “prefrailty” and no domains signifies nonfrail.

Frailty confers additional risk

The study results showed that among the 206 women, the prevalence of frailty and prefrailty was 9.2% and 60.7%, respectively. Of the 5 Fried frailty components, low walking speed and low physical activity followed by self-reported exhaustion were the most prevalent (96.6%, 87.5%, and 75.8%, respectively) among frail participants. Having sarcopenia only was strongly associated with frailty (odds ratio [OR], 8.28; 95% CI, 1.27–54.03; P=.027]). The likelihood of being frail was substantially higher with the presence of coexisting sarcopenia and osteoporosis (OR, 26.15; 95% CI, 3.31–218.76; P=.003).

Thus, both these conditions confer a high health burden for the individual as well as for health care systems. Osteosarcopenia is the term given when low bone mass and sarcopenia occur in consort. Previous data have shown that when osteoporosis or even osteopenia is combined with sarcopenia, it can result in a 3-fold increase in the risk of falls and a 4-fold increase in the risk of fracture compared with women who have osteopenia or osteoporosis alone.18

 

WHAT THIS EVIDENCE MEANS FOR PRACTICE
Sarcopenia, osteoporosis, and frailty are highly prevalent in older adults but are frequently underrecognized. Sarcopenia is characterized by progressive and generalized decline in muscle strength, function, and muscle mass with increasing age. Sarcopenia increases the likelihood of falls and adversely impacts functional independence and quality of life. Osteoporosis predisposes to low energy, fragility fractures, and is associated with chronic pain, impaired physical function, loss of independence, and higher risk of institutionalization. Clinicians need to be aware that when sarcopenia coexists with any degree of low bone mass, it will significantly increase the risk of falls and fracture compared with having osteopenia or osteoporosis alone.

Continue to: Denosumab effective in reducing falls, strengthening muscle...

 

 

Denosumab effective in reducing falls, strengthening muscle

Rupp T, von Vopelius E, Strahl A, et al. Beneficial effects of denosumab on muscle performance in patients with low BMD: a retrospective, propensity score-matched study. Osteoporos Int. 2022;33:2177-2184.

Results of a previous study showed that denosumab treatment significantly decreased falls and resulted in significant improvement in all sarcopenic measures.19 Furthermore, 1 year after denosumab was discontinued, a significant worsening occurred in both falls and sarcopenic measures. In that study, the control group, treated with alendronate or zoledronate, also showed improvement on some tests of muscle performance but no improvement in the risk of falls.

Those results agreed with the outcomes of the FREEDOM (Fracture Reduction Evaluation of Denosumab in Osteoporosis) trial.20 This study revealed that denosumab treatment not only reduced the risk of vertebral, nonvertebral, and hip fracture over 36 months but also that the denosumab-treated group had fewer falls compared with the placebo-treated group (4.5% vs 5.7%; P = .02).

 

Denosumab found to increase muscle strength

More recently, Rupp and colleagues conducted a retrospective cohort study that included women with osteoporosis or osteopenia who received vitamin D only (n = 52), alendronate 70 mg/week (n = 26), or denosumab (n = 52).21

After a mean follow-up period of 17.6 (SD, 9.0) months, the authors observed a significantly higher increase in grip force in both the denosumab (P<.001) and bisphosphonate groups (P = .001) compared with the vitamin D group. In addition, the denosumab group showed a significantly higher increase in chair rising test performance compared with the bisphosphonate group (denosumab vs bisphosphonate, P = 0.03). They concluded that denosumab resulted in increased muscle strength in the upper and lower limbs, indicating systemic rather than site-specific effects as compared with the bisphosphonate.

The authors concluded that based on these findings, denosumab might be favored over other osteoporosis treatments in patients with low BMD coexisting with poor muscle strength. ●

WHAT THIS EVIDENCE MEANS FOR PRACTICE
Osteoporosis and sarcopenia may share similar underlying risk factors. Muscle-bone interactions are important to minimize the risk of falls, fractures, and hospitalizations. In previous studies, denosumab as well as various bisphosphonates improved measures of sarcopenia, although only denosumab was associated with a reduction in the risk of falls. The study by Rupp and colleagues suggests that denosumab treatment may result in increased muscle strength in upper and lower limbs, indicating some systemic effect and not simply site-specific activity. Thus, in choosing a bone-specific agent for patients with abnormal muscle strength, mass, or performance, clinicians may want to consider denosumab as a choice for these reasons.
References
  1. American Cancer Society. Cancer Facts & Figures 2020. Atlanta, Georgia: American Cancer Society; 2020. Accessed November 7, 2022. https://www.cancer.org/content /dam/cancer-org/research/cancer-facts-and-statistics /annual-cancer-facts-and-figures/2020/cancer-facts-and -figures-2020.pdf
  2. Downey C, Kelly M, Quinlan JF. Changing trends in the mortality rate at 1-year post hip fracture—a systematic review. World J Orthop. 2019;10:166-175.
  3. Schuit SC, van der Klift M, Weel AE, et al. Fracture incidence and association with bone mineral density in elderly men and women: the Rotterdam study. Bone. 2004;34:195-202.
  4. de Villiers TJ, Goldstein SR. Update on bone health: the International Menopause Society White Paper 2021. Climacteric. 2021;24:498-504.
  5. Goldstein SR. Selective estrogen receptor modulators and bone health. Climacteric. 2022;25:56-59.
  6. Fisher B, Costantino JP, Wickerham DL, et al. Tamoxifen for the prevention of breast cancer: current status of the National Surgical Adjuvant Breast and Bowel Project P-1 study. J Natl Cancer Inst. 2005;97:1652-1662.
  7. Ettinger B, Black DM, Mitlak BH, et al; for the Multiple Outcomes of Raloxifene Evaluation (MORE) Investigators. Reduction of vertebral fracture risk in postmenopausal women with osteoporosis treated with raloxifene: results from a 3-year randomized clinical trial. JAMA. 1999;282:637645.
  8. Vogel VG, Costantino JP, Wickerham DL, et al; National Surgical Adjuvant Breast and Bowel Project (NSABP). Effects of tamoxifen vs raloxifene on the risk of developing invasive breast cancer and other disease outcomes: the NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial. JAMA. 2006;295:2727-2741.
  9. Silverman SL, Christiansen C, Genant HK, et al. Efficacy of bazedoxifene in reducing new vertebral fracture risk in postmenopausal women with osteoporosis: results from a 3-year, randomized, placebo-, and active-controlled clinical trial. J Bone Miner Res. 2008;23:1923-1934.
  10. Anderson GL, Limacher M, Assaf AR, et al; Women’s Health Initiative Steering Committee. Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women’s Health Initiative randomized controlled trial. JAMA. 2004:291:1701-1712.
  11. Lindsay R, Gallagher JC, Kagan R, et al. Efficacy of tissue-selective estrogen complex of bazedoxifene/conjugated estrogens for osteoporosis prevention in at-risk postmenopausal women. Fertil Steril. 2009;92:1045-1052.
  12. Kangas L, Härkönen P, Väänänen K, et al. Effects of the selective estrogen receptor modulator ospemifene on bone in rats. Horm Metab Res. 2014;46:27-35. 
  13. Constantine GD, Kagan R, Miller PD. Effects of ospemifene on bone parameters including clinical biomarkers in postmenopausal women. Menopause. 2016;23:638-644.
  14. Gerdhem P, Ivaska KK, Alatalo SL, et al. Biochemical markers of bone metabolism and prediction of fracture in elderly women. J Bone Miner Res. 2004;19:386-393.
  15. Cronin O, Lanham-New SA, Corfe BM, et al. Role of the microbiome in regulating bone metabolism and susceptibility to osteoporosis. Calcif Tissue Int. 2022;110:273-284.
  16. Yang X, Chang T, Yuan Q, et al. Changes in the composition of gut and vaginal microbiota in patients with postmenopausal osteoporosis. Front Immunol. 2022;13:930244.
  17. Laskou F, Fuggle NR, Patel HP, et al. Associations of osteoporosis and sarcopenia with frailty and multimorbidity among participants of the Hertfordshire Cohort Study. J Cachexia Sarcopenia Muscle. 2022;13:220-229.
  18. Hida T, Shimokata H, Sakai Y, et al. Sarcopenia and sarcopenic leg as potential risk factors for acute osteoporotic vertebral fracture among older women. Eur Spine J. 2016;25:3424-3431.
  19. El Miedany Y, El Gaafary M, Toth M, et al; Egyptian Academy of Bone Health, Metabolic Bone Diseases. Is there a potential dual effect of denosumab for treatment of osteoporosis and sarcopenia? Clin Rheumatol. 2021;40:4225-4232.
  20. Cummings SR, Martin JS, McClung MR, et al; FREEDOM trial. Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med. 2009;361:756-765.
  21. Rupp T, von Vopelius E, Strahl A, et al. Beneficial effects of denosumab on muscle performance in patients with low BMD: a retrospective, propensity score-matched study. Osteoporos Int. 2022;33:2177-2184.
Article PDF
Author and Disclosure Information

Steven R. Goldstein, MD, NCMP, CCD

Dr. Goldstein is Professor of Obstetrics and Gynecology, New York University Grossman School of Medicine, New York. He serves on the OBG Management Board of Editors.

Dr. Goldstein reports serving as a consultant to Astellas Pharma, Cook Ob/Gyn, Myovant Sciences, and Scynexis.

 

Issue
OBG Management - 35(2)
Publications
Topics
Page Number
12-17
Sections
Author and Disclosure Information

Steven R. Goldstein, MD, NCMP, CCD

Dr. Goldstein is Professor of Obstetrics and Gynecology, New York University Grossman School of Medicine, New York. He serves on the OBG Management Board of Editors.

Dr. Goldstein reports serving as a consultant to Astellas Pharma, Cook Ob/Gyn, Myovant Sciences, and Scynexis.

 

Author and Disclosure Information

Steven R. Goldstein, MD, NCMP, CCD

Dr. Goldstein is Professor of Obstetrics and Gynecology, New York University Grossman School of Medicine, New York. He serves on the OBG Management Board of Editors.

Dr. Goldstein reports serving as a consultant to Astellas Pharma, Cook Ob/Gyn, Myovant Sciences, and Scynexis.

 

Article PDF
Article PDF

 

 

I recently heard a lecture where the speaker quoted this statistic: “A 50-year-old woman who does not currently have heart disease or cancer has a life expectancy of 91.” Hopefully, anyone reading this article already is aware of the fact that as our patients age, hip fracture results in greater morbidity and mortality than early breast cancer. It should be well known to clinicians (and, ultimately, to our patients) that localized breast cancer has a survival rate of 99%,1 whereas hip fracture carries a 21% mortality in the first year after the event.2 In addition, approximately one-third of women who fracture their hip do not have osteoporosis.3 Furthermore, the role of muscle mass, strength, and performance in bone health has become well established.4

With this in mind, a recent encounter with a patient in my clinical practice illustrates what I believe is an increasing problem today. The patient had been on long-term prednisone systemically for polymyalgia rheumatica. Her dual energy x-ray absorptiometry (DXA) bone mass measurements were among the worst osteoporotic numbers I have witnessed. She related to me the “argument” that occurred between her rheumatologist and endocrinologist. One wanted her to use injectable parathyroid hormone analog daily, while the other advised yearly infusion of zoledronic acid. She chose the yearly infusion. I inquired if either physician had mentioned anything to her about using nonskid rugs in the bathroom, grab bars, being careful of black ice, a calcium-rich diet, vitamin D supplementation, good eyesight, illumination so she does not miss a step, mindful walking, and maintaining optimal balance, muscle mass, strength, and performance-enhancing exercise? She replied, “No, just which drug I should take.”

Realize that the goal for our patients should be to avoid the morbidity and mortality associated especially with hip fracture. The goal is not to have a better bone mass measurement on your DXA scan as you age. This is exactly why the name of this column, years ago, was changed from “Update on osteoporosis” to “Update on bone health.” Similarly, in 2021, the NOF (National Osteoporosis Foundation) became the BHOF (Bone Health and Osteoporosis Foundation). Thus, our understanding and interest in bone health should and must go beyond simply bone mass measurement with DXA technology. The articles highlighted in this year’s Update reflect the importance of this concept.

 

Know SERMs’ effects on bone health for appropriate prescribing

Goldstein SR. Selective estrogen receptor modulators and bone health. Climacteric. 2022;25:56-59.

Selective estrogen receptor modulators (SERMs) are synthetic molecules that bind to the estrogen receptor and can have agonistic activity in some tissues and antagonistic activity in others. In a recent article, I reviewed the known data regarding the effects of various SERMs on bone health.5

A rundown on 4 SERMs and their effects on bone

Tamoxifen is approved by the US Food and Drug Administration (FDA) for the prevention and treatment of breast cancer in women with estrogen receptor–positive tumors. The only prospective study of tamoxifen versus placebo in which fracture risk was studied in women at risk for but not diagnosed with breast cancer was the National Surgical Adjuvant Breast and Bowel Project (NSABP) P-1 trial. In this study, more than 13,000 women were randomly assigned to treatment with tamoxifen or placebo, with a primary objective of studying the incidence of invasive breast cancer in these high-risk women. With 7 years of follow-up, women receiving tamoxifen had significantly fewer fractures of the hip, radius, and spine (80 vs 116 in the placebo group), resulting in a combined relative risk (RR) of 0.68 (95% confidence interval [CI], 0.51–0.92).6

Raloxifene, another SERM, was extensively studied in the MORE (Multiple Outcomes of Raloxifene Evaluation) trial.7 This study involved more than 7,700 postmenopausal women with osteoporosis, average age 67. The incidence of first vertebral fracture was decreased from 4.3% with placebo to 1.9% with raloxifene (RR, 0.55; 95% CI, 0.29–0.71), and subsequent vertebral fractures were decreased from 20.2% with placebo to 14.1% with raloxifene (RR, 0.70; 95% CI, 0.60–0.90). In 2007, the FDA approved raloxifene for “reduction in risk of invasive breast cancer in postmenopausal women with osteoporosis” as well as for “postmenopausal women at high risk for invasive breast cancer” based on the Study of Tamoxifen and Raloxifene (STAR) trial that involved almost 20,000 postmenopausal women deemed at high risk for breast cancer.8

The concept of combining an estrogen with a SERM, known as a TSEC (tissue selective estrogen complex) was studied and brought to market as conjugated equine estrogen (CEE) 0.45 mg and bazedoxifene (BZA) 20 mg. CEE and BZA individually have been shown to prevent vertebral fracture.9,10 The combination of BZA and CEE has been shown to improve bone density compared with placebo.11 There are, however, no fracture prevention data for this combination therapy. This was the basis on which the combination agent received regulatory approval for prevention of osteoporosis in postmenopausal women. This combination drug is also FDA approved for treating moderate to severe vasomotor symptoms of menopause.

Ospemifene is yet another SERM that is clinically available, at an oral dose of 60 mg, and is indicated for the treatment of moderate to severe dyspareunia secondary to vulvovaginal atrophy, or genitourinary syndrome of menopause (GSM). Ospemifene effectively reduced bone loss in ovariectomized rats, with activity comparable to estradiol and raloxifene.12 Clinical data from three phase 1 or phase 2 clinical trials revealed that ospemifene 60 mg/day had a positive effect on biochemical markers for bone turnover in healthy postmenopausal women, with significant improvements relative to placebo and effects comparable to those of raloxifene.13 While actual fracture or bone mineral density (BMD) data in postmenopausal women are lacking, there is a good correlation between biochemical markers for bone turnover and occurrence of fracture.14 Women who need treatment for osteoporosis should not be treated with ospemifene, but women who use ospemifene for dyspareunia can expect positive activity on bone metabolism.

 

WHAT THIS EVIDENCE MEANS FOR PRACTICE

SERMs, unlike estrogen, have no class labeling. In fact, in the endometrium and vagina, they have variable effects. To date, however, in postmenopausal women, all SERMs have shown estrogenic activity in bone as well as being antiestrogenic in breast. Tamoxifen, well known for its use in estrogen receptor–positive breast cancer patients, demonstrates positive effects on bone and fracture reduction compared with placebo. Raloxifene is approved for prevention and treatment of osteoporosis and for breast cancer chemoprevention in high-risk patients. The TSEC combination of CEE and the SERM bazedoxifene is approved for treatment of moderate to severe vasomotor symptoms and prevention of osteoporosis. Finally, the SERM ospemifene, approved for treating moderate to severe dyspareunia or dryness due to vulvovaginal atrophy, or GSM, has demonstrated evidence of a positive effect on bone turnover and metabolism. Clinicians need to be aware of these effects when choosing medications for their patients.

 

Continue to: Gut microbiome constituents may influence the development of osteoporosis: A potential treatment target?...

 

 

Gut microbiome constituents may influence the development of osteoporosis: A potential treatment target?

Cronin O, Lanham-New SA, Corfe BM, et al. Role of the microbiome in regulating bone metabolism and susceptibility to osteoporosis. Calcif Tissue Int. 2022;110:273-284.

Yang X, Chang T, Yuan Q, et al. Changes in the composition of gut and vaginal microbiota in patients with postmenopausal osteoporosis. Front Immunol. 2022;13:930244.



The role of the microbiome in many arenas is rapidly emerging. Apparently, its relationship in bone metabolism is still in its infancy. A review of PubMed articles showed that 1 paper was published in 2012, none until 2 more in 2015, with a total of 221 published through November 1, 2022. A recent review by Cronin and colleagues on the microbiome’s role in regulating bone metabolism came out of a workshop held by the Osteoporosis and Bone Research Academy of the Royal Osteoporosis Society in the United Kingdom.15

 

The gut microbiome’s relationship with bone health

The authors noted that the human microbiota functions at the interface between diet, medication use, lifestyle, host immune development, and health. Hence, it is closely aligned with many of the recognized modifiable factors that influence bone mass accrual in the young and bone maintenance and skeletal decline in older populations. Microbiome research and discovery supports a role of the human gut microbiome in the regulation of bone metabolism and the pathogenesis of osteoporosis as well as its prevention and treatment.

Numerous factors which influence the gut microbiome and the development of osteoporosis overlap. These include body mass index (BMI), vitamin D, alcohol intake, diet, corticosteroid use, physical activity, sex hormone deficiency, genetic variability, and chronic inflammatory disorders.

Cronin and colleagues reviewed a number of clinical studies and concluded that “the available evidence suggests that probiotic supplements can attenuate bone loss in postmenopausal women, although the studies investigating this have been short term and individually have had small sample sizes. Moving forward, it will be important to conduct larger scale studies to evaluate if the skeletal response differs with different types of probiotic and also to determine if the effects are sustained in the longer term.”15

Composition of the microbiota

A recent study by Yang and colleagues focused on changes in gut and vaginal microbiota composition in patients with postmenopausal osteoporosis. They analyzed data from 132 postmenopausal women with osteoporosis (n = 34), osteopenia (n = 47), and controls (n = 51) based on their T-scores.16

Significant differences were observed in the microbial compositions of fecal samples between groups (P<.05), with some species enhanced in the control group whereas other species were higher in the osteoporosis group. Similar but less pronounced differences were seen in the vaginal microbiome but of different species.

The authors concluded that “The results show that changes in BMD in postmenopausal women are associated with the changes in gut microbiome and vaginal microbiome; however, changes in gut microbiome are more closely correlated with postmenopausal osteoporosis than vaginal microbiome.”16

 

WHAT THIS EVIDENCE MEANS FOR PRACTICE
While we are not yet ready to try to clinically alter the gut microbiome with various interventions, realizing that there is crosstalk between the gut microbiome and bone health is another factor to consider, and it begins with an appreciation of the various factors where the 2 overlap—BMI, vitamin D, alcohol intake, diet, corticosteroid use, physical activity, sex hormone deficiency, genetic variability, and chronic inflammatory disorders.

Continue to: Sarcopenia, osteoporosis, and frailty: A fracture risk triple play...

 

 

Sarcopenia, osteoporosis, and frailty: A fracture risk triple play

Laskou F, Fuggle NR, Patel HP, et al. Associations of osteoporosis and sarcopenia with frailty and multimorbidity among participants of the Hertfordshire Cohort Study. J Cachexia Sarcopenia Muscle. 2022;13:220-229.

Laskou and colleagues aimed to explore the relationship between sarcopenia, osteoporosis, and frailty in community-dwelling adults participating in a cohort study in the United Kingdom and to determine if the coexistence of osteoporosis and sarcopenia is associated with a significantly heavier health burden.17

 

Study details

The authors examined data from 206 women with an average age of 75.5 years. Sarcopenia was defined using the European Working Group on Sarcopenia in Older People (EWGSOP) criteria, which includes low grip strength or slow chair rise and low muscle quantity. Osteoporosis was defined by standard measurements as a T-score of less than or equal to -2.5 standard deviations at the femoral neck or use of any osteoporosis medications. Frailty was defined using the Fried definition, which includes having 3 or more of the following 5 domains: weakness, slowness, exhaustion, low physical activity, and unintentional weight loss. Having 1 or 2 domains is “prefrailty” and no domains signifies nonfrail.

Frailty confers additional risk

The study results showed that among the 206 women, the prevalence of frailty and prefrailty was 9.2% and 60.7%, respectively. Of the 5 Fried frailty components, low walking speed and low physical activity followed by self-reported exhaustion were the most prevalent (96.6%, 87.5%, and 75.8%, respectively) among frail participants. Having sarcopenia only was strongly associated with frailty (odds ratio [OR], 8.28; 95% CI, 1.27–54.03; P=.027]). The likelihood of being frail was substantially higher with the presence of coexisting sarcopenia and osteoporosis (OR, 26.15; 95% CI, 3.31–218.76; P=.003).

Thus, both these conditions confer a high health burden for the individual as well as for health care systems. Osteosarcopenia is the term given when low bone mass and sarcopenia occur in consort. Previous data have shown that when osteoporosis or even osteopenia is combined with sarcopenia, it can result in a 3-fold increase in the risk of falls and a 4-fold increase in the risk of fracture compared with women who have osteopenia or osteoporosis alone.18

 

WHAT THIS EVIDENCE MEANS FOR PRACTICE
Sarcopenia, osteoporosis, and frailty are highly prevalent in older adults but are frequently underrecognized. Sarcopenia is characterized by progressive and generalized decline in muscle strength, function, and muscle mass with increasing age. Sarcopenia increases the likelihood of falls and adversely impacts functional independence and quality of life. Osteoporosis predisposes to low energy, fragility fractures, and is associated with chronic pain, impaired physical function, loss of independence, and higher risk of institutionalization. Clinicians need to be aware that when sarcopenia coexists with any degree of low bone mass, it will significantly increase the risk of falls and fracture compared with having osteopenia or osteoporosis alone.

Continue to: Denosumab effective in reducing falls, strengthening muscle...

 

 

Denosumab effective in reducing falls, strengthening muscle

Rupp T, von Vopelius E, Strahl A, et al. Beneficial effects of denosumab on muscle performance in patients with low BMD: a retrospective, propensity score-matched study. Osteoporos Int. 2022;33:2177-2184.

Results of a previous study showed that denosumab treatment significantly decreased falls and resulted in significant improvement in all sarcopenic measures.19 Furthermore, 1 year after denosumab was discontinued, a significant worsening occurred in both falls and sarcopenic measures. In that study, the control group, treated with alendronate or zoledronate, also showed improvement on some tests of muscle performance but no improvement in the risk of falls.

Those results agreed with the outcomes of the FREEDOM (Fracture Reduction Evaluation of Denosumab in Osteoporosis) trial.20 This study revealed that denosumab treatment not only reduced the risk of vertebral, nonvertebral, and hip fracture over 36 months but also that the denosumab-treated group had fewer falls compared with the placebo-treated group (4.5% vs 5.7%; P = .02).

 

Denosumab found to increase muscle strength

More recently, Rupp and colleagues conducted a retrospective cohort study that included women with osteoporosis or osteopenia who received vitamin D only (n = 52), alendronate 70 mg/week (n = 26), or denosumab (n = 52).21

After a mean follow-up period of 17.6 (SD, 9.0) months, the authors observed a significantly higher increase in grip force in both the denosumab (P<.001) and bisphosphonate groups (P = .001) compared with the vitamin D group. In addition, the denosumab group showed a significantly higher increase in chair rising test performance compared with the bisphosphonate group (denosumab vs bisphosphonate, P = 0.03). They concluded that denosumab resulted in increased muscle strength in the upper and lower limbs, indicating systemic rather than site-specific effects as compared with the bisphosphonate.

The authors concluded that based on these findings, denosumab might be favored over other osteoporosis treatments in patients with low BMD coexisting with poor muscle strength. ●

WHAT THIS EVIDENCE MEANS FOR PRACTICE
Osteoporosis and sarcopenia may share similar underlying risk factors. Muscle-bone interactions are important to minimize the risk of falls, fractures, and hospitalizations. In previous studies, denosumab as well as various bisphosphonates improved measures of sarcopenia, although only denosumab was associated with a reduction in the risk of falls. The study by Rupp and colleagues suggests that denosumab treatment may result in increased muscle strength in upper and lower limbs, indicating some systemic effect and not simply site-specific activity. Thus, in choosing a bone-specific agent for patients with abnormal muscle strength, mass, or performance, clinicians may want to consider denosumab as a choice for these reasons.

 

 

I recently heard a lecture where the speaker quoted this statistic: “A 50-year-old woman who does not currently have heart disease or cancer has a life expectancy of 91.” Hopefully, anyone reading this article already is aware of the fact that as our patients age, hip fracture results in greater morbidity and mortality than early breast cancer. It should be well known to clinicians (and, ultimately, to our patients) that localized breast cancer has a survival rate of 99%,1 whereas hip fracture carries a 21% mortality in the first year after the event.2 In addition, approximately one-third of women who fracture their hip do not have osteoporosis.3 Furthermore, the role of muscle mass, strength, and performance in bone health has become well established.4

With this in mind, a recent encounter with a patient in my clinical practice illustrates what I believe is an increasing problem today. The patient had been on long-term prednisone systemically for polymyalgia rheumatica. Her dual energy x-ray absorptiometry (DXA) bone mass measurements were among the worst osteoporotic numbers I have witnessed. She related to me the “argument” that occurred between her rheumatologist and endocrinologist. One wanted her to use injectable parathyroid hormone analog daily, while the other advised yearly infusion of zoledronic acid. She chose the yearly infusion. I inquired if either physician had mentioned anything to her about using nonskid rugs in the bathroom, grab bars, being careful of black ice, a calcium-rich diet, vitamin D supplementation, good eyesight, illumination so she does not miss a step, mindful walking, and maintaining optimal balance, muscle mass, strength, and performance-enhancing exercise? She replied, “No, just which drug I should take.”

Realize that the goal for our patients should be to avoid the morbidity and mortality associated especially with hip fracture. The goal is not to have a better bone mass measurement on your DXA scan as you age. This is exactly why the name of this column, years ago, was changed from “Update on osteoporosis” to “Update on bone health.” Similarly, in 2021, the NOF (National Osteoporosis Foundation) became the BHOF (Bone Health and Osteoporosis Foundation). Thus, our understanding and interest in bone health should and must go beyond simply bone mass measurement with DXA technology. The articles highlighted in this year’s Update reflect the importance of this concept.

 

Know SERMs’ effects on bone health for appropriate prescribing

Goldstein SR. Selective estrogen receptor modulators and bone health. Climacteric. 2022;25:56-59.

Selective estrogen receptor modulators (SERMs) are synthetic molecules that bind to the estrogen receptor and can have agonistic activity in some tissues and antagonistic activity in others. In a recent article, I reviewed the known data regarding the effects of various SERMs on bone health.5

A rundown on 4 SERMs and their effects on bone

Tamoxifen is approved by the US Food and Drug Administration (FDA) for the prevention and treatment of breast cancer in women with estrogen receptor–positive tumors. The only prospective study of tamoxifen versus placebo in which fracture risk was studied in women at risk for but not diagnosed with breast cancer was the National Surgical Adjuvant Breast and Bowel Project (NSABP) P-1 trial. In this study, more than 13,000 women were randomly assigned to treatment with tamoxifen or placebo, with a primary objective of studying the incidence of invasive breast cancer in these high-risk women. With 7 years of follow-up, women receiving tamoxifen had significantly fewer fractures of the hip, radius, and spine (80 vs 116 in the placebo group), resulting in a combined relative risk (RR) of 0.68 (95% confidence interval [CI], 0.51–0.92).6

Raloxifene, another SERM, was extensively studied in the MORE (Multiple Outcomes of Raloxifene Evaluation) trial.7 This study involved more than 7,700 postmenopausal women with osteoporosis, average age 67. The incidence of first vertebral fracture was decreased from 4.3% with placebo to 1.9% with raloxifene (RR, 0.55; 95% CI, 0.29–0.71), and subsequent vertebral fractures were decreased from 20.2% with placebo to 14.1% with raloxifene (RR, 0.70; 95% CI, 0.60–0.90). In 2007, the FDA approved raloxifene for “reduction in risk of invasive breast cancer in postmenopausal women with osteoporosis” as well as for “postmenopausal women at high risk for invasive breast cancer” based on the Study of Tamoxifen and Raloxifene (STAR) trial that involved almost 20,000 postmenopausal women deemed at high risk for breast cancer.8

The concept of combining an estrogen with a SERM, known as a TSEC (tissue selective estrogen complex) was studied and brought to market as conjugated equine estrogen (CEE) 0.45 mg and bazedoxifene (BZA) 20 mg. CEE and BZA individually have been shown to prevent vertebral fracture.9,10 The combination of BZA and CEE has been shown to improve bone density compared with placebo.11 There are, however, no fracture prevention data for this combination therapy. This was the basis on which the combination agent received regulatory approval for prevention of osteoporosis in postmenopausal women. This combination drug is also FDA approved for treating moderate to severe vasomotor symptoms of menopause.

Ospemifene is yet another SERM that is clinically available, at an oral dose of 60 mg, and is indicated for the treatment of moderate to severe dyspareunia secondary to vulvovaginal atrophy, or genitourinary syndrome of menopause (GSM). Ospemifene effectively reduced bone loss in ovariectomized rats, with activity comparable to estradiol and raloxifene.12 Clinical data from three phase 1 or phase 2 clinical trials revealed that ospemifene 60 mg/day had a positive effect on biochemical markers for bone turnover in healthy postmenopausal women, with significant improvements relative to placebo and effects comparable to those of raloxifene.13 While actual fracture or bone mineral density (BMD) data in postmenopausal women are lacking, there is a good correlation between biochemical markers for bone turnover and occurrence of fracture.14 Women who need treatment for osteoporosis should not be treated with ospemifene, but women who use ospemifene for dyspareunia can expect positive activity on bone metabolism.

 

WHAT THIS EVIDENCE MEANS FOR PRACTICE

SERMs, unlike estrogen, have no class labeling. In fact, in the endometrium and vagina, they have variable effects. To date, however, in postmenopausal women, all SERMs have shown estrogenic activity in bone as well as being antiestrogenic in breast. Tamoxifen, well known for its use in estrogen receptor–positive breast cancer patients, demonstrates positive effects on bone and fracture reduction compared with placebo. Raloxifene is approved for prevention and treatment of osteoporosis and for breast cancer chemoprevention in high-risk patients. The TSEC combination of CEE and the SERM bazedoxifene is approved for treatment of moderate to severe vasomotor symptoms and prevention of osteoporosis. Finally, the SERM ospemifene, approved for treating moderate to severe dyspareunia or dryness due to vulvovaginal atrophy, or GSM, has demonstrated evidence of a positive effect on bone turnover and metabolism. Clinicians need to be aware of these effects when choosing medications for their patients.

 

Continue to: Gut microbiome constituents may influence the development of osteoporosis: A potential treatment target?...

 

 

Gut microbiome constituents may influence the development of osteoporosis: A potential treatment target?

Cronin O, Lanham-New SA, Corfe BM, et al. Role of the microbiome in regulating bone metabolism and susceptibility to osteoporosis. Calcif Tissue Int. 2022;110:273-284.

Yang X, Chang T, Yuan Q, et al. Changes in the composition of gut and vaginal microbiota in patients with postmenopausal osteoporosis. Front Immunol. 2022;13:930244.



The role of the microbiome in many arenas is rapidly emerging. Apparently, its relationship in bone metabolism is still in its infancy. A review of PubMed articles showed that 1 paper was published in 2012, none until 2 more in 2015, with a total of 221 published through November 1, 2022. A recent review by Cronin and colleagues on the microbiome’s role in regulating bone metabolism came out of a workshop held by the Osteoporosis and Bone Research Academy of the Royal Osteoporosis Society in the United Kingdom.15

 

The gut microbiome’s relationship with bone health

The authors noted that the human microbiota functions at the interface between diet, medication use, lifestyle, host immune development, and health. Hence, it is closely aligned with many of the recognized modifiable factors that influence bone mass accrual in the young and bone maintenance and skeletal decline in older populations. Microbiome research and discovery supports a role of the human gut microbiome in the regulation of bone metabolism and the pathogenesis of osteoporosis as well as its prevention and treatment.

Numerous factors which influence the gut microbiome and the development of osteoporosis overlap. These include body mass index (BMI), vitamin D, alcohol intake, diet, corticosteroid use, physical activity, sex hormone deficiency, genetic variability, and chronic inflammatory disorders.

Cronin and colleagues reviewed a number of clinical studies and concluded that “the available evidence suggests that probiotic supplements can attenuate bone loss in postmenopausal women, although the studies investigating this have been short term and individually have had small sample sizes. Moving forward, it will be important to conduct larger scale studies to evaluate if the skeletal response differs with different types of probiotic and also to determine if the effects are sustained in the longer term.”15

Composition of the microbiota

A recent study by Yang and colleagues focused on changes in gut and vaginal microbiota composition in patients with postmenopausal osteoporosis. They analyzed data from 132 postmenopausal women with osteoporosis (n = 34), osteopenia (n = 47), and controls (n = 51) based on their T-scores.16

Significant differences were observed in the microbial compositions of fecal samples between groups (P<.05), with some species enhanced in the control group whereas other species were higher in the osteoporosis group. Similar but less pronounced differences were seen in the vaginal microbiome but of different species.

The authors concluded that “The results show that changes in BMD in postmenopausal women are associated with the changes in gut microbiome and vaginal microbiome; however, changes in gut microbiome are more closely correlated with postmenopausal osteoporosis than vaginal microbiome.”16

 

WHAT THIS EVIDENCE MEANS FOR PRACTICE
While we are not yet ready to try to clinically alter the gut microbiome with various interventions, realizing that there is crosstalk between the gut microbiome and bone health is another factor to consider, and it begins with an appreciation of the various factors where the 2 overlap—BMI, vitamin D, alcohol intake, diet, corticosteroid use, physical activity, sex hormone deficiency, genetic variability, and chronic inflammatory disorders.

Continue to: Sarcopenia, osteoporosis, and frailty: A fracture risk triple play...

 

 

Sarcopenia, osteoporosis, and frailty: A fracture risk triple play

Laskou F, Fuggle NR, Patel HP, et al. Associations of osteoporosis and sarcopenia with frailty and multimorbidity among participants of the Hertfordshire Cohort Study. J Cachexia Sarcopenia Muscle. 2022;13:220-229.

Laskou and colleagues aimed to explore the relationship between sarcopenia, osteoporosis, and frailty in community-dwelling adults participating in a cohort study in the United Kingdom and to determine if the coexistence of osteoporosis and sarcopenia is associated with a significantly heavier health burden.17

 

Study details

The authors examined data from 206 women with an average age of 75.5 years. Sarcopenia was defined using the European Working Group on Sarcopenia in Older People (EWGSOP) criteria, which includes low grip strength or slow chair rise and low muscle quantity. Osteoporosis was defined by standard measurements as a T-score of less than or equal to -2.5 standard deviations at the femoral neck or use of any osteoporosis medications. Frailty was defined using the Fried definition, which includes having 3 or more of the following 5 domains: weakness, slowness, exhaustion, low physical activity, and unintentional weight loss. Having 1 or 2 domains is “prefrailty” and no domains signifies nonfrail.

Frailty confers additional risk

The study results showed that among the 206 women, the prevalence of frailty and prefrailty was 9.2% and 60.7%, respectively. Of the 5 Fried frailty components, low walking speed and low physical activity followed by self-reported exhaustion were the most prevalent (96.6%, 87.5%, and 75.8%, respectively) among frail participants. Having sarcopenia only was strongly associated with frailty (odds ratio [OR], 8.28; 95% CI, 1.27–54.03; P=.027]). The likelihood of being frail was substantially higher with the presence of coexisting sarcopenia and osteoporosis (OR, 26.15; 95% CI, 3.31–218.76; P=.003).

Thus, both these conditions confer a high health burden for the individual as well as for health care systems. Osteosarcopenia is the term given when low bone mass and sarcopenia occur in consort. Previous data have shown that when osteoporosis or even osteopenia is combined with sarcopenia, it can result in a 3-fold increase in the risk of falls and a 4-fold increase in the risk of fracture compared with women who have osteopenia or osteoporosis alone.18

 

WHAT THIS EVIDENCE MEANS FOR PRACTICE
Sarcopenia, osteoporosis, and frailty are highly prevalent in older adults but are frequently underrecognized. Sarcopenia is characterized by progressive and generalized decline in muscle strength, function, and muscle mass with increasing age. Sarcopenia increases the likelihood of falls and adversely impacts functional independence and quality of life. Osteoporosis predisposes to low energy, fragility fractures, and is associated with chronic pain, impaired physical function, loss of independence, and higher risk of institutionalization. Clinicians need to be aware that when sarcopenia coexists with any degree of low bone mass, it will significantly increase the risk of falls and fracture compared with having osteopenia or osteoporosis alone.

Continue to: Denosumab effective in reducing falls, strengthening muscle...

 

 

Denosumab effective in reducing falls, strengthening muscle

Rupp T, von Vopelius E, Strahl A, et al. Beneficial effects of denosumab on muscle performance in patients with low BMD: a retrospective, propensity score-matched study. Osteoporos Int. 2022;33:2177-2184.

Results of a previous study showed that denosumab treatment significantly decreased falls and resulted in significant improvement in all sarcopenic measures.19 Furthermore, 1 year after denosumab was discontinued, a significant worsening occurred in both falls and sarcopenic measures. In that study, the control group, treated with alendronate or zoledronate, also showed improvement on some tests of muscle performance but no improvement in the risk of falls.

Those results agreed with the outcomes of the FREEDOM (Fracture Reduction Evaluation of Denosumab in Osteoporosis) trial.20 This study revealed that denosumab treatment not only reduced the risk of vertebral, nonvertebral, and hip fracture over 36 months but also that the denosumab-treated group had fewer falls compared with the placebo-treated group (4.5% vs 5.7%; P = .02).

 

Denosumab found to increase muscle strength

More recently, Rupp and colleagues conducted a retrospective cohort study that included women with osteoporosis or osteopenia who received vitamin D only (n = 52), alendronate 70 mg/week (n = 26), or denosumab (n = 52).21

After a mean follow-up period of 17.6 (SD, 9.0) months, the authors observed a significantly higher increase in grip force in both the denosumab (P<.001) and bisphosphonate groups (P = .001) compared with the vitamin D group. In addition, the denosumab group showed a significantly higher increase in chair rising test performance compared with the bisphosphonate group (denosumab vs bisphosphonate, P = 0.03). They concluded that denosumab resulted in increased muscle strength in the upper and lower limbs, indicating systemic rather than site-specific effects as compared with the bisphosphonate.

The authors concluded that based on these findings, denosumab might be favored over other osteoporosis treatments in patients with low BMD coexisting with poor muscle strength. ●

WHAT THIS EVIDENCE MEANS FOR PRACTICE
Osteoporosis and sarcopenia may share similar underlying risk factors. Muscle-bone interactions are important to minimize the risk of falls, fractures, and hospitalizations. In previous studies, denosumab as well as various bisphosphonates improved measures of sarcopenia, although only denosumab was associated with a reduction in the risk of falls. The study by Rupp and colleagues suggests that denosumab treatment may result in increased muscle strength in upper and lower limbs, indicating some systemic effect and not simply site-specific activity. Thus, in choosing a bone-specific agent for patients with abnormal muscle strength, mass, or performance, clinicians may want to consider denosumab as a choice for these reasons.
References
  1. American Cancer Society. Cancer Facts & Figures 2020. Atlanta, Georgia: American Cancer Society; 2020. Accessed November 7, 2022. https://www.cancer.org/content /dam/cancer-org/research/cancer-facts-and-statistics /annual-cancer-facts-and-figures/2020/cancer-facts-and -figures-2020.pdf
  2. Downey C, Kelly M, Quinlan JF. Changing trends in the mortality rate at 1-year post hip fracture—a systematic review. World J Orthop. 2019;10:166-175.
  3. Schuit SC, van der Klift M, Weel AE, et al. Fracture incidence and association with bone mineral density in elderly men and women: the Rotterdam study. Bone. 2004;34:195-202.
  4. de Villiers TJ, Goldstein SR. Update on bone health: the International Menopause Society White Paper 2021. Climacteric. 2021;24:498-504.
  5. Goldstein SR. Selective estrogen receptor modulators and bone health. Climacteric. 2022;25:56-59.
  6. Fisher B, Costantino JP, Wickerham DL, et al. Tamoxifen for the prevention of breast cancer: current status of the National Surgical Adjuvant Breast and Bowel Project P-1 study. J Natl Cancer Inst. 2005;97:1652-1662.
  7. Ettinger B, Black DM, Mitlak BH, et al; for the Multiple Outcomes of Raloxifene Evaluation (MORE) Investigators. Reduction of vertebral fracture risk in postmenopausal women with osteoporosis treated with raloxifene: results from a 3-year randomized clinical trial. JAMA. 1999;282:637645.
  8. Vogel VG, Costantino JP, Wickerham DL, et al; National Surgical Adjuvant Breast and Bowel Project (NSABP). Effects of tamoxifen vs raloxifene on the risk of developing invasive breast cancer and other disease outcomes: the NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial. JAMA. 2006;295:2727-2741.
  9. Silverman SL, Christiansen C, Genant HK, et al. Efficacy of bazedoxifene in reducing new vertebral fracture risk in postmenopausal women with osteoporosis: results from a 3-year, randomized, placebo-, and active-controlled clinical trial. J Bone Miner Res. 2008;23:1923-1934.
  10. Anderson GL, Limacher M, Assaf AR, et al; Women’s Health Initiative Steering Committee. Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women’s Health Initiative randomized controlled trial. JAMA. 2004:291:1701-1712.
  11. Lindsay R, Gallagher JC, Kagan R, et al. Efficacy of tissue-selective estrogen complex of bazedoxifene/conjugated estrogens for osteoporosis prevention in at-risk postmenopausal women. Fertil Steril. 2009;92:1045-1052.
  12. Kangas L, Härkönen P, Väänänen K, et al. Effects of the selective estrogen receptor modulator ospemifene on bone in rats. Horm Metab Res. 2014;46:27-35. 
  13. Constantine GD, Kagan R, Miller PD. Effects of ospemifene on bone parameters including clinical biomarkers in postmenopausal women. Menopause. 2016;23:638-644.
  14. Gerdhem P, Ivaska KK, Alatalo SL, et al. Biochemical markers of bone metabolism and prediction of fracture in elderly women. J Bone Miner Res. 2004;19:386-393.
  15. Cronin O, Lanham-New SA, Corfe BM, et al. Role of the microbiome in regulating bone metabolism and susceptibility to osteoporosis. Calcif Tissue Int. 2022;110:273-284.
  16. Yang X, Chang T, Yuan Q, et al. Changes in the composition of gut and vaginal microbiota in patients with postmenopausal osteoporosis. Front Immunol. 2022;13:930244.
  17. Laskou F, Fuggle NR, Patel HP, et al. Associations of osteoporosis and sarcopenia with frailty and multimorbidity among participants of the Hertfordshire Cohort Study. J Cachexia Sarcopenia Muscle. 2022;13:220-229.
  18. Hida T, Shimokata H, Sakai Y, et al. Sarcopenia and sarcopenic leg as potential risk factors for acute osteoporotic vertebral fracture among older women. Eur Spine J. 2016;25:3424-3431.
  19. El Miedany Y, El Gaafary M, Toth M, et al; Egyptian Academy of Bone Health, Metabolic Bone Diseases. Is there a potential dual effect of denosumab for treatment of osteoporosis and sarcopenia? Clin Rheumatol. 2021;40:4225-4232.
  20. Cummings SR, Martin JS, McClung MR, et al; FREEDOM trial. Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med. 2009;361:756-765.
  21. Rupp T, von Vopelius E, Strahl A, et al. Beneficial effects of denosumab on muscle performance in patients with low BMD: a retrospective, propensity score-matched study. Osteoporos Int. 2022;33:2177-2184.
References
  1. American Cancer Society. Cancer Facts & Figures 2020. Atlanta, Georgia: American Cancer Society; 2020. Accessed November 7, 2022. https://www.cancer.org/content /dam/cancer-org/research/cancer-facts-and-statistics /annual-cancer-facts-and-figures/2020/cancer-facts-and -figures-2020.pdf
  2. Downey C, Kelly M, Quinlan JF. Changing trends in the mortality rate at 1-year post hip fracture—a systematic review. World J Orthop. 2019;10:166-175.
  3. Schuit SC, van der Klift M, Weel AE, et al. Fracture incidence and association with bone mineral density in elderly men and women: the Rotterdam study. Bone. 2004;34:195-202.
  4. de Villiers TJ, Goldstein SR. Update on bone health: the International Menopause Society White Paper 2021. Climacteric. 2021;24:498-504.
  5. Goldstein SR. Selective estrogen receptor modulators and bone health. Climacteric. 2022;25:56-59.
  6. Fisher B, Costantino JP, Wickerham DL, et al. Tamoxifen for the prevention of breast cancer: current status of the National Surgical Adjuvant Breast and Bowel Project P-1 study. J Natl Cancer Inst. 2005;97:1652-1662.
  7. Ettinger B, Black DM, Mitlak BH, et al; for the Multiple Outcomes of Raloxifene Evaluation (MORE) Investigators. Reduction of vertebral fracture risk in postmenopausal women with osteoporosis treated with raloxifene: results from a 3-year randomized clinical trial. JAMA. 1999;282:637645.
  8. Vogel VG, Costantino JP, Wickerham DL, et al; National Surgical Adjuvant Breast and Bowel Project (NSABP). Effects of tamoxifen vs raloxifene on the risk of developing invasive breast cancer and other disease outcomes: the NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial. JAMA. 2006;295:2727-2741.
  9. Silverman SL, Christiansen C, Genant HK, et al. Efficacy of bazedoxifene in reducing new vertebral fracture risk in postmenopausal women with osteoporosis: results from a 3-year, randomized, placebo-, and active-controlled clinical trial. J Bone Miner Res. 2008;23:1923-1934.
  10. Anderson GL, Limacher M, Assaf AR, et al; Women’s Health Initiative Steering Committee. Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women’s Health Initiative randomized controlled trial. JAMA. 2004:291:1701-1712.
  11. Lindsay R, Gallagher JC, Kagan R, et al. Efficacy of tissue-selective estrogen complex of bazedoxifene/conjugated estrogens for osteoporosis prevention in at-risk postmenopausal women. Fertil Steril. 2009;92:1045-1052.
  12. Kangas L, Härkönen P, Väänänen K, et al. Effects of the selective estrogen receptor modulator ospemifene on bone in rats. Horm Metab Res. 2014;46:27-35. 
  13. Constantine GD, Kagan R, Miller PD. Effects of ospemifene on bone parameters including clinical biomarkers in postmenopausal women. Menopause. 2016;23:638-644.
  14. Gerdhem P, Ivaska KK, Alatalo SL, et al. Biochemical markers of bone metabolism and prediction of fracture in elderly women. J Bone Miner Res. 2004;19:386-393.
  15. Cronin O, Lanham-New SA, Corfe BM, et al. Role of the microbiome in regulating bone metabolism and susceptibility to osteoporosis. Calcif Tissue Int. 2022;110:273-284.
  16. Yang X, Chang T, Yuan Q, et al. Changes in the composition of gut and vaginal microbiota in patients with postmenopausal osteoporosis. Front Immunol. 2022;13:930244.
  17. Laskou F, Fuggle NR, Patel HP, et al. Associations of osteoporosis and sarcopenia with frailty and multimorbidity among participants of the Hertfordshire Cohort Study. J Cachexia Sarcopenia Muscle. 2022;13:220-229.
  18. Hida T, Shimokata H, Sakai Y, et al. Sarcopenia and sarcopenic leg as potential risk factors for acute osteoporotic vertebral fracture among older women. Eur Spine J. 2016;25:3424-3431.
  19. El Miedany Y, El Gaafary M, Toth M, et al; Egyptian Academy of Bone Health, Metabolic Bone Diseases. Is there a potential dual effect of denosumab for treatment of osteoporosis and sarcopenia? Clin Rheumatol. 2021;40:4225-4232.
  20. Cummings SR, Martin JS, McClung MR, et al; FREEDOM trial. Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med. 2009;361:756-765.
  21. Rupp T, von Vopelius E, Strahl A, et al. Beneficial effects of denosumab on muscle performance in patients with low BMD: a retrospective, propensity score-matched study. Osteoporos Int. 2022;33:2177-2184.
Issue
OBG Management - 35(2)
Issue
OBG Management - 35(2)
Page Number
12-17
Page Number
12-17
Publications
Publications
Topics
Article Type
Sections
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.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Nonsurgical options for stress urinary incontinence

Article Type
Changed
Sun, 02/12/2023 - 09:52

In the article, “Nonsurgical treatments for patients with urinary incontinence” (OBG Manag. September 2022;34:36- 42.), the authors, Ashley J. Murillo, MD, and Halina M. Zyczynski, MD, discuss the successful nonsurgical management of urge urinary incontinence, stress urinary incontinence, and mixed urinary incontinence, presenting the case of a 39-year-old woman with urine leakage during exercise. As a follow-up for readers, OBG Management posted a quiz question asking, “Which of the following is a nonsurgical treatment for stress urinary incontinence?”

 

 

Poll results

A total of 129 readers cast their vote:

50.4% (65 readers) said bladder retraining

36.4% (47 readers) said periurethral bulking

13.2% (17 readers) said antimuscarinic agents

 


The correct answer was periurethral bulking, as bladder retraining and antimuscarinic agents, according to TABLE 1 in Murillo and Zyczynski’s article, are appropriate for managing urge urinary incontinence.

 

Article PDF
Issue
OBG Management - 35(1)
Publications
Page Number
33
Sections
Article PDF
Article PDF

In the article, “Nonsurgical treatments for patients with urinary incontinence” (OBG Manag. September 2022;34:36- 42.), the authors, Ashley J. Murillo, MD, and Halina M. Zyczynski, MD, discuss the successful nonsurgical management of urge urinary incontinence, stress urinary incontinence, and mixed urinary incontinence, presenting the case of a 39-year-old woman with urine leakage during exercise. As a follow-up for readers, OBG Management posted a quiz question asking, “Which of the following is a nonsurgical treatment for stress urinary incontinence?”

 

 

Poll results

A total of 129 readers cast their vote:

50.4% (65 readers) said bladder retraining

36.4% (47 readers) said periurethral bulking

13.2% (17 readers) said antimuscarinic agents

 


The correct answer was periurethral bulking, as bladder retraining and antimuscarinic agents, according to TABLE 1 in Murillo and Zyczynski’s article, are appropriate for managing urge urinary incontinence.

 

In the article, “Nonsurgical treatments for patients with urinary incontinence” (OBG Manag. September 2022;34:36- 42.), the authors, Ashley J. Murillo, MD, and Halina M. Zyczynski, MD, discuss the successful nonsurgical management of urge urinary incontinence, stress urinary incontinence, and mixed urinary incontinence, presenting the case of a 39-year-old woman with urine leakage during exercise. As a follow-up for readers, OBG Management posted a quiz question asking, “Which of the following is a nonsurgical treatment for stress urinary incontinence?”

 

 

Poll results

A total of 129 readers cast their vote:

50.4% (65 readers) said bladder retraining

36.4% (47 readers) said periurethral bulking

13.2% (17 readers) said antimuscarinic agents

 


The correct answer was periurethral bulking, as bladder retraining and antimuscarinic agents, according to TABLE 1 in Murillo and Zyczynski’s article, are appropriate for managing urge urinary incontinence.

 

Issue
OBG Management - 35(1)
Issue
OBG Management - 35(1)
Page Number
33
Page Number
33
Publications
Publications
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Eyebrow Default
Poll results
Gate On Date
Tue, 02/07/2023 - 20:00
Un-Gate On Date
Tue, 02/07/2023 - 20:00
Use ProPublica
CFC Schedule Remove Status
Tue, 02/07/2023 - 20:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Product updates and reviews

Article Type
Changed
Mon, 02/06/2023 - 15:39

HEGENBERGER RETRACTOR:  IS IT HELPFUL FOR PERINEAL REPAIR? 

The Hegenberger Retractor, manufactured by Hegenberger Medical (Abingdon, United Kingdom) is available for purchase in the United States through Rocket Medical. A video that I find particularly useful for explaining its use is available here: https://www.youtube.com /watch?v=p-jilXgXZLY

Background. About 85% of women having a vaginal birth experience some form of perineal trauma, and 60% to 70% receive stitches for those spontaneous tears or intentional incisions. As such, repairing perineal lacerations is a requisite skill for all obstetricians and midwives, and every provider has developed exposure techniques to perform their suturing with the goals of good tissue re-approximation, efficiency, minimized patient discomfort, reduced blood loss, and safety from needle sticks. For several millennia, the most commonly used tissue retractor for these repairs has been one’s own fingers, or those of a colleague. While cost-effective and readily available, fingers do have drawbacks as a vaginal retractor. First, their use as a retractor precludes their use for other tasks. Second, their frequent need to be inserted and replaced (see drawback #1) can be uncomfortable for patients. Third, their limited surface area is often insufficient to appropriately provide adequate tissue retraction for optimal surgical site visualization. Finally, they get tired and typically do not appreciate being stuck with needles. Given all this, it is surprising that so many centuries have passed with so little innovation for this ubiquitous procedure. Fortunately, Danish midwife Malene Hegenberger thought now was a good time to change the status quo.

Design/Functionality. The Hegenberger Retractor is brilliant in its simplicity. Its unique molded plastic design is smooth, ergonomic, nonconductive, and packaged as a single-use sterile device. Amazingly, it has a near-perfect pliability balance, making it simultaneously easy to compress for insertion while providing enough retraction tension for good visualization once it has been reexpanded. The subtle ridges on the compression points are just enough to allow for a good grip, and the notches on the sides are a convenient addition for holding extra suture if needed. The device has been cleared by the US Food and Drug Administration (FDA) as a Class 1 device and is approved for sale in the United States. In my experience with its use, I thought it was easy to place and provided excellent exposure for the repairs I was doing. In fact, I thought it provided as good if not better exposure than what I would expect from a Gelpi retractor without any of the trauma the Gelpi adds with its pointed ends. Smile emoji!

Innovation. In the early 1800s, French midwifery pioneer Marie Boivin introduced a novel pelvimeter and a revolutionary 2-part speculum to the technology of the day. Why it took more than 200 years for the ideas of another cutting-edge midwife to breach the walls of the obstetric technological establishment remains a mystery, but fortunately it has been done. While seemingly obvious, the Hegenberger Retractor is the culmination of years of work and 88 prototypes. It looks simple, but the secret to its functionality is the precision with which each dimension and every curve was designed. The device has been cleared by the FDA as a Class 1 device and is approved for sale in the United States. 

Summary. There are a lot of reasons to like the Hegenberger Retractor. I like it for its simplicity; I like it for its functionality; I like it for its ability to fill a real need. On the downside, I do not like that it is a single-use plastic device, and I am not happy about adding cost to obstetric care. Most of all, I hate that I did not invent it. 

Is the Hegenberger Retractor going to be needed to repair every obstetric laceration? No. Will it provide perfect exposure to repair every obstetric laceration? Of course not. But it is an incredibly clever device that will be very helpful in many situations, and I suspect it will soon become a mainstay on most maternity units as it gains recognition.

FOR MORE INFORMATION, VISIT www.rocketmedical.com

References
  1. McCandlish R, Bowler U, van Asten H, et al. A randomised controlled trial of care of the perineum during second stage of normal labour. Br J Obstet Gynaecol. 1998;105:1262-1272.
  2. Ferry G. Marie Boivin: from midwife to gynaecologist. Lancet. 2019;393:2192-2193. doi: 10.1016/S0140-6736(19)31188-2. 
Article PDF
Author and Disclosure Information

James Greenberg, MD

Chief of Gynecology
Associate Professor, Harvard Medical School
Boston, Massachusetts

The views of the author are personal opinions and do not necessarily represent the views of OBG Management. Dr. Greenberg personally trials all the products he reviews. Dr. Greenberg has no conflicts of interest with this product or the company that produces it.

Issue
OBG Management - 35(1)
Publications
Topics
Page Number
28
Sections
Author and Disclosure Information

James Greenberg, MD

Chief of Gynecology
Associate Professor, Harvard Medical School
Boston, Massachusetts

The views of the author are personal opinions and do not necessarily represent the views of OBG Management. Dr. Greenberg personally trials all the products he reviews. Dr. Greenberg has no conflicts of interest with this product or the company that produces it.

Author and Disclosure Information

James Greenberg, MD

Chief of Gynecology
Associate Professor, Harvard Medical School
Boston, Massachusetts

The views of the author are personal opinions and do not necessarily represent the views of OBG Management. Dr. Greenberg personally trials all the products he reviews. Dr. Greenberg has no conflicts of interest with this product or the company that produces it.

Article PDF
Article PDF

HEGENBERGER RETRACTOR:  IS IT HELPFUL FOR PERINEAL REPAIR? 

The Hegenberger Retractor, manufactured by Hegenberger Medical (Abingdon, United Kingdom) is available for purchase in the United States through Rocket Medical. A video that I find particularly useful for explaining its use is available here: https://www.youtube.com /watch?v=p-jilXgXZLY

Background. About 85% of women having a vaginal birth experience some form of perineal trauma, and 60% to 70% receive stitches for those spontaneous tears or intentional incisions. As such, repairing perineal lacerations is a requisite skill for all obstetricians and midwives, and every provider has developed exposure techniques to perform their suturing with the goals of good tissue re-approximation, efficiency, minimized patient discomfort, reduced blood loss, and safety from needle sticks. For several millennia, the most commonly used tissue retractor for these repairs has been one’s own fingers, or those of a colleague. While cost-effective and readily available, fingers do have drawbacks as a vaginal retractor. First, their use as a retractor precludes their use for other tasks. Second, their frequent need to be inserted and replaced (see drawback #1) can be uncomfortable for patients. Third, their limited surface area is often insufficient to appropriately provide adequate tissue retraction for optimal surgical site visualization. Finally, they get tired and typically do not appreciate being stuck with needles. Given all this, it is surprising that so many centuries have passed with so little innovation for this ubiquitous procedure. Fortunately, Danish midwife Malene Hegenberger thought now was a good time to change the status quo.

Design/Functionality. The Hegenberger Retractor is brilliant in its simplicity. Its unique molded plastic design is smooth, ergonomic, nonconductive, and packaged as a single-use sterile device. Amazingly, it has a near-perfect pliability balance, making it simultaneously easy to compress for insertion while providing enough retraction tension for good visualization once it has been reexpanded. The subtle ridges on the compression points are just enough to allow for a good grip, and the notches on the sides are a convenient addition for holding extra suture if needed. The device has been cleared by the US Food and Drug Administration (FDA) as a Class 1 device and is approved for sale in the United States. In my experience with its use, I thought it was easy to place and provided excellent exposure for the repairs I was doing. In fact, I thought it provided as good if not better exposure than what I would expect from a Gelpi retractor without any of the trauma the Gelpi adds with its pointed ends. Smile emoji!

Innovation. In the early 1800s, French midwifery pioneer Marie Boivin introduced a novel pelvimeter and a revolutionary 2-part speculum to the technology of the day. Why it took more than 200 years for the ideas of another cutting-edge midwife to breach the walls of the obstetric technological establishment remains a mystery, but fortunately it has been done. While seemingly obvious, the Hegenberger Retractor is the culmination of years of work and 88 prototypes. It looks simple, but the secret to its functionality is the precision with which each dimension and every curve was designed. The device has been cleared by the FDA as a Class 1 device and is approved for sale in the United States. 

Summary. There are a lot of reasons to like the Hegenberger Retractor. I like it for its simplicity; I like it for its functionality; I like it for its ability to fill a real need. On the downside, I do not like that it is a single-use plastic device, and I am not happy about adding cost to obstetric care. Most of all, I hate that I did not invent it. 

Is the Hegenberger Retractor going to be needed to repair every obstetric laceration? No. Will it provide perfect exposure to repair every obstetric laceration? Of course not. But it is an incredibly clever device that will be very helpful in many situations, and I suspect it will soon become a mainstay on most maternity units as it gains recognition.

FOR MORE INFORMATION, VISIT www.rocketmedical.com

HEGENBERGER RETRACTOR:  IS IT HELPFUL FOR PERINEAL REPAIR? 

The Hegenberger Retractor, manufactured by Hegenberger Medical (Abingdon, United Kingdom) is available for purchase in the United States through Rocket Medical. A video that I find particularly useful for explaining its use is available here: https://www.youtube.com /watch?v=p-jilXgXZLY

Background. About 85% of women having a vaginal birth experience some form of perineal trauma, and 60% to 70% receive stitches for those spontaneous tears or intentional incisions. As such, repairing perineal lacerations is a requisite skill for all obstetricians and midwives, and every provider has developed exposure techniques to perform their suturing with the goals of good tissue re-approximation, efficiency, minimized patient discomfort, reduced blood loss, and safety from needle sticks. For several millennia, the most commonly used tissue retractor for these repairs has been one’s own fingers, or those of a colleague. While cost-effective and readily available, fingers do have drawbacks as a vaginal retractor. First, their use as a retractor precludes their use for other tasks. Second, their frequent need to be inserted and replaced (see drawback #1) can be uncomfortable for patients. Third, their limited surface area is often insufficient to appropriately provide adequate tissue retraction for optimal surgical site visualization. Finally, they get tired and typically do not appreciate being stuck with needles. Given all this, it is surprising that so many centuries have passed with so little innovation for this ubiquitous procedure. Fortunately, Danish midwife Malene Hegenberger thought now was a good time to change the status quo.

Design/Functionality. The Hegenberger Retractor is brilliant in its simplicity. Its unique molded plastic design is smooth, ergonomic, nonconductive, and packaged as a single-use sterile device. Amazingly, it has a near-perfect pliability balance, making it simultaneously easy to compress for insertion while providing enough retraction tension for good visualization once it has been reexpanded. The subtle ridges on the compression points are just enough to allow for a good grip, and the notches on the sides are a convenient addition for holding extra suture if needed. The device has been cleared by the US Food and Drug Administration (FDA) as a Class 1 device and is approved for sale in the United States. In my experience with its use, I thought it was easy to place and provided excellent exposure for the repairs I was doing. In fact, I thought it provided as good if not better exposure than what I would expect from a Gelpi retractor without any of the trauma the Gelpi adds with its pointed ends. Smile emoji!

Innovation. In the early 1800s, French midwifery pioneer Marie Boivin introduced a novel pelvimeter and a revolutionary 2-part speculum to the technology of the day. Why it took more than 200 years for the ideas of another cutting-edge midwife to breach the walls of the obstetric technological establishment remains a mystery, but fortunately it has been done. While seemingly obvious, the Hegenberger Retractor is the culmination of years of work and 88 prototypes. It looks simple, but the secret to its functionality is the precision with which each dimension and every curve was designed. The device has been cleared by the FDA as a Class 1 device and is approved for sale in the United States. 

Summary. There are a lot of reasons to like the Hegenberger Retractor. I like it for its simplicity; I like it for its functionality; I like it for its ability to fill a real need. On the downside, I do not like that it is a single-use plastic device, and I am not happy about adding cost to obstetric care. Most of all, I hate that I did not invent it. 

Is the Hegenberger Retractor going to be needed to repair every obstetric laceration? No. Will it provide perfect exposure to repair every obstetric laceration? Of course not. But it is an incredibly clever device that will be very helpful in many situations, and I suspect it will soon become a mainstay on most maternity units as it gains recognition.

FOR MORE INFORMATION, VISIT www.rocketmedical.com

References
  1. McCandlish R, Bowler U, van Asten H, et al. A randomised controlled trial of care of the perineum during second stage of normal labour. Br J Obstet Gynaecol. 1998;105:1262-1272.
  2. Ferry G. Marie Boivin: from midwife to gynaecologist. Lancet. 2019;393:2192-2193. doi: 10.1016/S0140-6736(19)31188-2. 
References
  1. McCandlish R, Bowler U, van Asten H, et al. A randomised controlled trial of care of the perineum during second stage of normal labour. Br J Obstet Gynaecol. 1998;105:1262-1272.
  2. Ferry G. Marie Boivin: from midwife to gynaecologist. Lancet. 2019;393:2192-2193. doi: 10.1016/S0140-6736(19)31188-2. 
Issue
OBG Management - 35(1)
Issue
OBG Management - 35(1)
Page Number
28
Page Number
28
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Sun, 02/05/2023 - 10:45
Un-Gate On Date
Sun, 02/05/2023 - 10:45
Use ProPublica
CFC Schedule Remove Status
Sun, 02/05/2023 - 10:45
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

COMMENT & CONTROVERSY

Article Type
Changed
Mon, 02/06/2023 - 15:41

 

Should treatment be initiated for mild chronic hypertension in pregnancy to improve outcomes?

JAIMEY M. PAULI, MD (JUNE 2022)

Consider this, when it comes to treating chronic hypertension

I welcome the article by Dr. Jaimey Pauli, which focuses on initiating treatment for mild chronic hypertension in pregnancy to reach a goal blood pressure (BP) of <140/90 mm Hg to prevent adverse maternal and fetal outcomes.1 I would like to offer 3 additional thoughts for your consideration. First, it is known that there is a physiological decrease in BP during the second trimester, which results in a normotensive presentation. Thus, it would be beneficial to see if pregnant women with high-normal BP levels before the third trimester be administered a lower dose of antihypertensives. However, there is also a concern that decreased maternal BP may compromise uteroplacental perfusion and fetal circulation, which also could be evaluated.2

Second, I would like to see how comorbidities affect the initiation of antihypertensives for mild chronic hypertension in pregnancy. Research incorporating pregnant women with borderline hypertension and comorbidities such as obesity, hyperlipidemia, and diabetes mellitus type 2 (DM) is likely to yield informative results. This is especially beneficial since, for example, chronic hypertension and DM are independent risk factors for adverse maternal and fetal outcomes; therefore, a mother with both these conditions may have additive effects on obstetric outcomes.3

Lastly, I would suggest you include a brief conversation about prepregnancy ways to manage women with chronic hypertension. Because many women who enter pregnancy with chronic hypertension have hypertension of unknown origin, it would be beneficial to optimize antihypertensive regimens before conception.4 Also, it should be further evaluated whether initiation of lifestyle modifications, such as weight reduction and the DASH diet before pregnancy, for women with chronic hypertension improves pregnancy outcomes.

Cassandra Maafoh, MD

Macon, Georgia

References

  1. Pauli JM. Should treatment be initiated for mild chronic hypertension in pregnancy to improve outcomes? OBG Manag. 2022;34:14-15.
  2. Brown CM, Garovic VD. Drug treatment of hypertension in pregnancy. Drugs. 2014;74:283-296. https://doi.org/10.1007/s40265-014-0187-7.
  3. Yanit KE, Snowden JM, Cheng YW, et al. The impact of chronic hypertension and pregestational diabetes on pregnancy outcomes. Am J Obstet Gynecol. 2012;207. https://doi. org/10.1016/j.ajog.2012.06.066.
  4. Seely EW, Ecker J. Chronic hypertension in pregnancy. Circulation. 2014;129:1254-1261. https:// doi.org/10.1161/circulationaha.113.003904. 

2022 UPDATE ON FEMALE SEXUAL HEALTH

BARBARA LEVY, MD (AUGUST 2022)

Are these new and rare syndromes’ pathophysiological mechanisms related?

I read with great interest Dr. Barbara Levy’s UPDATE in the August 2022 issue on testosterone therapy for women with hypoactive sexual desire disorder (HSDD), as well as her comments on persistent genital arousal disorder/genito-pelvic dysesthesia (PGAD/GPD) that was recently so coined by the International Society for the Study of Women’s Sexual Health (ISSWSH) as a 2-component syndrome.1 The new syndrome, explains Dr. Levy, presents with “the perception of genital arousal that is involuntary, unrelated to sexual desire, without any identified cause, not relieved with orgasm, and distressing to the patient (the PGAD component),” combined with “itching, burning, tingling, or pain” (the GPD component).

Although agreeing with ISSWSH that diagnosis and management require a multidisciplinary biopsychosocial approach, in her practical advice, Dr. Levy mentioned: “neuropathic signaling” with “aberrant sensory processing” as the syndrome’s possible main pathophysiology. Interestingly, there are 2 other rare, chronic, and “poorly recognized source(s) of major distress to a small but significant group of patients.” Persistent idiopathic oro-facial pain (PIFP) disorder2 after dental interventions and burning mouth syndrome (BMS),3 defined by the absence of any local or systemic contributing etiology, also present with continuous local burning and pain (as in GPD). Consequently, PGAD/GPD may indeed have the same pathophysiological explanation—as Dr. Levy suggested—of being a (genital) peripheral chronic neuropathic pain condition.

A potentially promising new therapeutic approach for PGAD/GPD would then be to use the same, or similar, antineuropathic medications (Clonazepam, Nortriptyline, Pregabalin, etc.) in the form of topical vaginal swishing solutions similar to the presently recommended antiepileptic and/or antidepressant oral swishing treatment for PIFP and BMS. As the topical approach works well for oral neuropathic pain, vaginal swishing could potentially be the answer for PGAD/GPD peripheral neuropathic pain. Moreover, such a novel topical approach would significantly increase patient motivation for treatment by avoiding the adverse effects of ingested antiepileptic or antidepressant drugs.

This is the first time that anticonvulsant and/or antidepressant vaginal swishing is proposed as topical therapy for GPD peripheral neuropathic pain, still pending scientific/clinical validation. ●

Zwi Hoch, MD

Newton, Massachusetts

  1. Goldstein I, Komisaruk BR, Pukall CF, et al. International Society for the Study of Women’s Sexual Health (ISSWSH) Review of Epidemiology and Pathophysiology, and a Consensus Nomenclature and Process of Care for the Management of Persistent Genital Arousal Disorder/Genito-Pelvic Dysesthesia (PGAD/GPD). J Sex Med. 2021;18:665-697.
  2. Baad-Hansen L, Benoliel R. Neuropathic orofacial pain: facts and fiction. Cephalgia. 2017;37:670-679.
  3. Kuten-Shorer M, Treister NS, Stock S, et al. Safety and tolerability of topical clonazepam solution for management of oral dysesthesia. Oral Surg Oral Med Oral Pathol Oral Radiol. 2017;124: 146-151. 
Article PDF
Issue
OBG Management - 35(1)
Publications
Topics
Page Number
10-11
Sections
Article PDF
Article PDF

 

Should treatment be initiated for mild chronic hypertension in pregnancy to improve outcomes?

JAIMEY M. PAULI, MD (JUNE 2022)

Consider this, when it comes to treating chronic hypertension

I welcome the article by Dr. Jaimey Pauli, which focuses on initiating treatment for mild chronic hypertension in pregnancy to reach a goal blood pressure (BP) of <140/90 mm Hg to prevent adverse maternal and fetal outcomes.1 I would like to offer 3 additional thoughts for your consideration. First, it is known that there is a physiological decrease in BP during the second trimester, which results in a normotensive presentation. Thus, it would be beneficial to see if pregnant women with high-normal BP levels before the third trimester be administered a lower dose of antihypertensives. However, there is also a concern that decreased maternal BP may compromise uteroplacental perfusion and fetal circulation, which also could be evaluated.2

Second, I would like to see how comorbidities affect the initiation of antihypertensives for mild chronic hypertension in pregnancy. Research incorporating pregnant women with borderline hypertension and comorbidities such as obesity, hyperlipidemia, and diabetes mellitus type 2 (DM) is likely to yield informative results. This is especially beneficial since, for example, chronic hypertension and DM are independent risk factors for adverse maternal and fetal outcomes; therefore, a mother with both these conditions may have additive effects on obstetric outcomes.3

Lastly, I would suggest you include a brief conversation about prepregnancy ways to manage women with chronic hypertension. Because many women who enter pregnancy with chronic hypertension have hypertension of unknown origin, it would be beneficial to optimize antihypertensive regimens before conception.4 Also, it should be further evaluated whether initiation of lifestyle modifications, such as weight reduction and the DASH diet before pregnancy, for women with chronic hypertension improves pregnancy outcomes.

Cassandra Maafoh, MD

Macon, Georgia

References

  1. Pauli JM. Should treatment be initiated for mild chronic hypertension in pregnancy to improve outcomes? OBG Manag. 2022;34:14-15.
  2. Brown CM, Garovic VD. Drug treatment of hypertension in pregnancy. Drugs. 2014;74:283-296. https://doi.org/10.1007/s40265-014-0187-7.
  3. Yanit KE, Snowden JM, Cheng YW, et al. The impact of chronic hypertension and pregestational diabetes on pregnancy outcomes. Am J Obstet Gynecol. 2012;207. https://doi. org/10.1016/j.ajog.2012.06.066.
  4. Seely EW, Ecker J. Chronic hypertension in pregnancy. Circulation. 2014;129:1254-1261. https:// doi.org/10.1161/circulationaha.113.003904. 

2022 UPDATE ON FEMALE SEXUAL HEALTH

BARBARA LEVY, MD (AUGUST 2022)

Are these new and rare syndromes’ pathophysiological mechanisms related?

I read with great interest Dr. Barbara Levy’s UPDATE in the August 2022 issue on testosterone therapy for women with hypoactive sexual desire disorder (HSDD), as well as her comments on persistent genital arousal disorder/genito-pelvic dysesthesia (PGAD/GPD) that was recently so coined by the International Society for the Study of Women’s Sexual Health (ISSWSH) as a 2-component syndrome.1 The new syndrome, explains Dr. Levy, presents with “the perception of genital arousal that is involuntary, unrelated to sexual desire, without any identified cause, not relieved with orgasm, and distressing to the patient (the PGAD component),” combined with “itching, burning, tingling, or pain” (the GPD component).

Although agreeing with ISSWSH that diagnosis and management require a multidisciplinary biopsychosocial approach, in her practical advice, Dr. Levy mentioned: “neuropathic signaling” with “aberrant sensory processing” as the syndrome’s possible main pathophysiology. Interestingly, there are 2 other rare, chronic, and “poorly recognized source(s) of major distress to a small but significant group of patients.” Persistent idiopathic oro-facial pain (PIFP) disorder2 after dental interventions and burning mouth syndrome (BMS),3 defined by the absence of any local or systemic contributing etiology, also present with continuous local burning and pain (as in GPD). Consequently, PGAD/GPD may indeed have the same pathophysiological explanation—as Dr. Levy suggested—of being a (genital) peripheral chronic neuropathic pain condition.

A potentially promising new therapeutic approach for PGAD/GPD would then be to use the same, or similar, antineuropathic medications (Clonazepam, Nortriptyline, Pregabalin, etc.) in the form of topical vaginal swishing solutions similar to the presently recommended antiepileptic and/or antidepressant oral swishing treatment for PIFP and BMS. As the topical approach works well for oral neuropathic pain, vaginal swishing could potentially be the answer for PGAD/GPD peripheral neuropathic pain. Moreover, such a novel topical approach would significantly increase patient motivation for treatment by avoiding the adverse effects of ingested antiepileptic or antidepressant drugs.

This is the first time that anticonvulsant and/or antidepressant vaginal swishing is proposed as topical therapy for GPD peripheral neuropathic pain, still pending scientific/clinical validation. ●

Zwi Hoch, MD

Newton, Massachusetts

  1. Goldstein I, Komisaruk BR, Pukall CF, et al. International Society for the Study of Women’s Sexual Health (ISSWSH) Review of Epidemiology and Pathophysiology, and a Consensus Nomenclature and Process of Care for the Management of Persistent Genital Arousal Disorder/Genito-Pelvic Dysesthesia (PGAD/GPD). J Sex Med. 2021;18:665-697.
  2. Baad-Hansen L, Benoliel R. Neuropathic orofacial pain: facts and fiction. Cephalgia. 2017;37:670-679.
  3. Kuten-Shorer M, Treister NS, Stock S, et al. Safety and tolerability of topical clonazepam solution for management of oral dysesthesia. Oral Surg Oral Med Oral Pathol Oral Radiol. 2017;124: 146-151. 

 

Should treatment be initiated for mild chronic hypertension in pregnancy to improve outcomes?

JAIMEY M. PAULI, MD (JUNE 2022)

Consider this, when it comes to treating chronic hypertension

I welcome the article by Dr. Jaimey Pauli, which focuses on initiating treatment for mild chronic hypertension in pregnancy to reach a goal blood pressure (BP) of <140/90 mm Hg to prevent adverse maternal and fetal outcomes.1 I would like to offer 3 additional thoughts for your consideration. First, it is known that there is a physiological decrease in BP during the second trimester, which results in a normotensive presentation. Thus, it would be beneficial to see if pregnant women with high-normal BP levels before the third trimester be administered a lower dose of antihypertensives. However, there is also a concern that decreased maternal BP may compromise uteroplacental perfusion and fetal circulation, which also could be evaluated.2

Second, I would like to see how comorbidities affect the initiation of antihypertensives for mild chronic hypertension in pregnancy. Research incorporating pregnant women with borderline hypertension and comorbidities such as obesity, hyperlipidemia, and diabetes mellitus type 2 (DM) is likely to yield informative results. This is especially beneficial since, for example, chronic hypertension and DM are independent risk factors for adverse maternal and fetal outcomes; therefore, a mother with both these conditions may have additive effects on obstetric outcomes.3

Lastly, I would suggest you include a brief conversation about prepregnancy ways to manage women with chronic hypertension. Because many women who enter pregnancy with chronic hypertension have hypertension of unknown origin, it would be beneficial to optimize antihypertensive regimens before conception.4 Also, it should be further evaluated whether initiation of lifestyle modifications, such as weight reduction and the DASH diet before pregnancy, for women with chronic hypertension improves pregnancy outcomes.

Cassandra Maafoh, MD

Macon, Georgia

References

  1. Pauli JM. Should treatment be initiated for mild chronic hypertension in pregnancy to improve outcomes? OBG Manag. 2022;34:14-15.
  2. Brown CM, Garovic VD. Drug treatment of hypertension in pregnancy. Drugs. 2014;74:283-296. https://doi.org/10.1007/s40265-014-0187-7.
  3. Yanit KE, Snowden JM, Cheng YW, et al. The impact of chronic hypertension and pregestational diabetes on pregnancy outcomes. Am J Obstet Gynecol. 2012;207. https://doi. org/10.1016/j.ajog.2012.06.066.
  4. Seely EW, Ecker J. Chronic hypertension in pregnancy. Circulation. 2014;129:1254-1261. https:// doi.org/10.1161/circulationaha.113.003904. 

2022 UPDATE ON FEMALE SEXUAL HEALTH

BARBARA LEVY, MD (AUGUST 2022)

Are these new and rare syndromes’ pathophysiological mechanisms related?

I read with great interest Dr. Barbara Levy’s UPDATE in the August 2022 issue on testosterone therapy for women with hypoactive sexual desire disorder (HSDD), as well as her comments on persistent genital arousal disorder/genito-pelvic dysesthesia (PGAD/GPD) that was recently so coined by the International Society for the Study of Women’s Sexual Health (ISSWSH) as a 2-component syndrome.1 The new syndrome, explains Dr. Levy, presents with “the perception of genital arousal that is involuntary, unrelated to sexual desire, without any identified cause, not relieved with orgasm, and distressing to the patient (the PGAD component),” combined with “itching, burning, tingling, or pain” (the GPD component).

Although agreeing with ISSWSH that diagnosis and management require a multidisciplinary biopsychosocial approach, in her practical advice, Dr. Levy mentioned: “neuropathic signaling” with “aberrant sensory processing” as the syndrome’s possible main pathophysiology. Interestingly, there are 2 other rare, chronic, and “poorly recognized source(s) of major distress to a small but significant group of patients.” Persistent idiopathic oro-facial pain (PIFP) disorder2 after dental interventions and burning mouth syndrome (BMS),3 defined by the absence of any local or systemic contributing etiology, also present with continuous local burning and pain (as in GPD). Consequently, PGAD/GPD may indeed have the same pathophysiological explanation—as Dr. Levy suggested—of being a (genital) peripheral chronic neuropathic pain condition.

A potentially promising new therapeutic approach for PGAD/GPD would then be to use the same, or similar, antineuropathic medications (Clonazepam, Nortriptyline, Pregabalin, etc.) in the form of topical vaginal swishing solutions similar to the presently recommended antiepileptic and/or antidepressant oral swishing treatment for PIFP and BMS. As the topical approach works well for oral neuropathic pain, vaginal swishing could potentially be the answer for PGAD/GPD peripheral neuropathic pain. Moreover, such a novel topical approach would significantly increase patient motivation for treatment by avoiding the adverse effects of ingested antiepileptic or antidepressant drugs.

This is the first time that anticonvulsant and/or antidepressant vaginal swishing is proposed as topical therapy for GPD peripheral neuropathic pain, still pending scientific/clinical validation. ●

Zwi Hoch, MD

Newton, Massachusetts

  1. Goldstein I, Komisaruk BR, Pukall CF, et al. International Society for the Study of Women’s Sexual Health (ISSWSH) Review of Epidemiology and Pathophysiology, and a Consensus Nomenclature and Process of Care for the Management of Persistent Genital Arousal Disorder/Genito-Pelvic Dysesthesia (PGAD/GPD). J Sex Med. 2021;18:665-697.
  2. Baad-Hansen L, Benoliel R. Neuropathic orofacial pain: facts and fiction. Cephalgia. 2017;37:670-679.
  3. Kuten-Shorer M, Treister NS, Stock S, et al. Safety and tolerability of topical clonazepam solution for management of oral dysesthesia. Oral Surg Oral Med Oral Pathol Oral Radiol. 2017;124: 146-151. 
Issue
OBG Management - 35(1)
Issue
OBG Management - 35(1)
Page Number
10-11
Page Number
10-11
Publications
Publications
Topics
Article Type
Sections
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.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Perineal leiomyoma after 4th degree laceration

Article Type
Changed
Fri, 02/03/2023 - 14:55
Author and Disclosure Information

Dr. Collins is a Gynecologic Surgeon and Obstetrics Physician, Landstuhl Regional Medical
Center.

Dr. Stone is Chief of Urogynecology, Womack Army Medical Center.

Dr. Shvartsman is at the Uniformed Services University of the Health Sciences.

Dr. Casablanca is at the Levine Cancer Institute, Atrium Health Wake Forest, Charlotte,
North Carolina.

Dr. Savioli is at Walter Reed National Military Medical Center.

Dr. Long is Staff Pathologist at Walter Reed National Military Medical Center.

Dr. Wells is in the Department of Pathology, Walter Reed National Military Medical Center.

Dr. Dengler is Chief, Urogynecology Division, Walter Reed National Military Medical Center.

Dr. Gruber is Director, Female Pelvic Medicine and Reconstructive Surgery, Sibley
Memorial Hospital (Johns Hopkins Medicine).

Dr. Casablanca reports serving as a consultant to AstraZeneca. The other authors report no
financial relationships relevant to this video.

Issue
OBG Management - 35(2)
Publications
Sections
Author and Disclosure Information

Dr. Collins is a Gynecologic Surgeon and Obstetrics Physician, Landstuhl Regional Medical
Center.

Dr. Stone is Chief of Urogynecology, Womack Army Medical Center.

Dr. Shvartsman is at the Uniformed Services University of the Health Sciences.

Dr. Casablanca is at the Levine Cancer Institute, Atrium Health Wake Forest, Charlotte,
North Carolina.

Dr. Savioli is at Walter Reed National Military Medical Center.

Dr. Long is Staff Pathologist at Walter Reed National Military Medical Center.

Dr. Wells is in the Department of Pathology, Walter Reed National Military Medical Center.

Dr. Dengler is Chief, Urogynecology Division, Walter Reed National Military Medical Center.

Dr. Gruber is Director, Female Pelvic Medicine and Reconstructive Surgery, Sibley
Memorial Hospital (Johns Hopkins Medicine).

Dr. Casablanca reports serving as a consultant to AstraZeneca. The other authors report no
financial relationships relevant to this video.

Author and Disclosure Information

Dr. Collins is a Gynecologic Surgeon and Obstetrics Physician, Landstuhl Regional Medical
Center.

Dr. Stone is Chief of Urogynecology, Womack Army Medical Center.

Dr. Shvartsman is at the Uniformed Services University of the Health Sciences.

Dr. Casablanca is at the Levine Cancer Institute, Atrium Health Wake Forest, Charlotte,
North Carolina.

Dr. Savioli is at Walter Reed National Military Medical Center.

Dr. Long is Staff Pathologist at Walter Reed National Military Medical Center.

Dr. Wells is in the Department of Pathology, Walter Reed National Military Medical Center.

Dr. Dengler is Chief, Urogynecology Division, Walter Reed National Military Medical Center.

Dr. Gruber is Director, Female Pelvic Medicine and Reconstructive Surgery, Sibley
Memorial Hospital (Johns Hopkins Medicine).

Dr. Casablanca reports serving as a consultant to AstraZeneca. The other authors report no
financial relationships relevant to this video.

Issue
OBG Management - 35(2)
Issue
OBG Management - 35(2)
Publications
Publications
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Sun, 01/29/2023 - 11:00
Un-Gate On Date
Sun, 01/29/2023 - 11:00
Use ProPublica
CFC Schedule Remove Status
Sun, 01/29/2023 - 11:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

How the Dobbs decision shapes the ObGyn workforce and training landscape

Article Type
Changed
Sun, 02/05/2023 - 10:17
Display Headline
How the Dobbs decision shapes the ObGyn workforce and training landscape

Photo: Shutterstock

Six months after the Supreme Court decision that overturned the constitutional right to abortion, trainees across the United States are asking a critical question in the current resident recruitment season: How will the restrictions on abortion access affect my training as an obstetrician-gynecologist, and will they impact my ability to be the kind of provider I want to be in the future?

Among the myriad of downstream effects to patient care, the Dobbs decision will indisputably impact the scope of residency training for those that provide reproductive health services. Almost half of ObGyn residents train in states that have abortion restrictions in place.1 New educational milestones for abortion training, which are a requirement by the Accreditation Council for Graduate Medical Education (ACGME), were proposed quickly after Dobbs, guiding programs to offer opportunities for training in nonrestricted areas or the “combination of didactic activities, including simulation” to meet the training requirement in abortion care.2

Like many providers, residents already are grappling with precarious and risky circumstances, balancing patient safety and patient-driven care amidst pre-existing and newly enforced abortion restrictions. Whether managing a patient with an undesired pregnancy, severe medical comorbidities, unexpected pregnancy complications such as preterm premature rupture of membranes, or bleeding, or substantial fetal anomalies, ObGyn residents cannot gain the experience of providing the full scope of reproductive health care without the ability to offer all possible management options. While some enacted abortion restrictions have exceptions for the health of or life-saving measures for the mother, there is no standard guidance for timing of interventions, leaving providers confused and in fear of legal retribution. At a time when trainees should be learning to provide patient-centered, evidence-based care, they are instead paralyzed by the legal or professional consequences they may face for offering their best medical judgements.

Furthermore, the lack of exposure to dilation and evacuation procedures for residents in restricted practice areas will undoubtably decrease their confidence in managing acute complications, which is one of the critical facets of residency training. In a surgical field where repetition is crucial for technical competence, highlighted by ACGME minimum case requirements, the decreased volume of abortion procedures is a disadvantage for trainees and a disservice for patients. While anti-choice promoters may argue that involvement in surgical management of early pregnancy loss should suffice for ObGyn training in family planning, this piecemeal approach will leave gaps in technical skills.

The fear of legal ramifications, moral injury, and inadequate surgical training may lead to the siphoning of talented trainees to areas in the country with fewer restrictions.3Dobbs already has demonstrated how limiting abortion access will deepen inequities in reproductive health care service delivery. Approximately 55% of ObGyn trainees and nearly two-thirds of maternal-fetal medicine graduates join the workforce in the state where they received their training.4 Medical students will seek opportunities for high-quality ObGyn training in areas that will help them to be well-prepared, competent physicians—and more often than not, stay in the area or region that they trained in. This will lead to provider shortages in areas where access to reproductive health care and subspecialist providers already is limited, further exacerbating existing health disparities.

During this recruitment season, trainees and residency programs alike will need to reckon with how the ramifications of Dobbs will alter both the immediate and long-term training in comprehensive reproductive health care for the ObGyn workforce. ObGyn trainees have taken a stand in response to the Dobbs decision, and nearly 750 current residents signed onto the statement below as a commitment to high-quality training and patient-centered care. Clinical experience in performing abortions is essential to the provision of comprehensive evidence-based reproductive health care, and access to these procedures is as important for physicians-in-training as it is for patients.

Actions to take to ensure high-quality abortion training in ObGyn residencies include the following:

  • Connect with and stay involved with organizations such as the American College of Obstetricians and Gynecologists (ACOG), Physicians for Reproductive Health (PRH), and Medical Students for Choice (MSFC) for initiatives, toolkits, and resources for training at your institutions.
  • Seek specific abortion training opportunities through the Leadership Training Academy (offered through PRH) or the Abortion Training Institute (offered through MSFC).
  • Ensure that your residency program meets the ACGME criteria of providing opportunities for clinical experiences for abortion care and work with program leadership at a program, state, or regional level to enforce these competencies.
  • Reach out to your local American Civil Liberties Union or other local reproductive legal rights organizations if you want to be involved with advocacy around abortion access and training but have concerns about legal protections.
  • Have a voice at the table for empowering training opportunities by seeking leadership positions through ACOG, ACGME, Council on Resident Education in Obstetrics and Gynecology and the Association of Professors of Gynecology and Obstetrics, American Medical Association, Student National Medical Association, and subspecialty organizations.
  • Vote in every election and promote voting registration and access to your patients, colleagues, and communities. ●

Continue to: The implications of the Dobbs v Jackson Women’s Health Organization decision on the health care and wellbeing of our patients...

 

 

The implications of the Dobbs v Jackson Women’s Health Organization decision on the health care and wellbeing of our patients

On June 24, 2022, the Supreme Court of the United States ruled in a 6-3 majority decision to overturn the constitutional right to abortion protected by Roe v Wade since 1973. As health care providers, we are outraged at the Court’s disregard for an individual’s right to make reproductive decisions for themselves and their families and are deeply concerned about the devastating consequences to reproductive care and outcomes in this country for all people. Reproductive health decisions, including growing a family and whether or not to continue a pregnancy, are complex and incredibly personal. Our role as health care providers is to help guide those decisions with empathy and evidencebased clinical recommendations. This ruling undermines a patient’s right to bodily autonomy, free of impositions from government and political pressures, and it threatens the sanctity of complex medical decision-making between a patient, their family, and their medical team.

As medical professionals, we know that every patient’s situation is unique—banning abortion procedures ties the hands of physicians trying to provide the most medically appropriate options in a compassionate manner. We know that both medical and surgical abortions are safe and can save lives. These procedures can help patients with potentially life-threatening conditions worsened by pregnancy, a poor prognosis for the fetus, or a complication from the pregnancy itself. Physicians use scientific research and individualized approaches to help patients in unique situations, and attempts to legislate personal health decisions compromise the practice of evidence-based medicine.

We also know that this decision will impact some communities more than others. Access to safe abortion care will become dependent on which region of the country a person lives in and whether or not a person has resources to seek this care. Due to continued systemic racism and oppression, patients of color will be disproportionately impacted and likely will suffer worse health outcomes from unsafe abortions. Those that rely on public insurance or who are uninsured will face overwhelming barriers in seeking abortion services. These disparities in reproductive care, which contribute to our nation’s health crises in maternal morbidity and mortality, unintended pregnancy, and neonatal complications, will further entrench health inequities, and patient lives and livelihoods will suffer.

We acknowledge the impact that this decision will have on restricting access to reproductive care. We stand by the fact that abortion care is health care. We vow to uphold the tenets of our profession to place patient autonomy and provision of safe quality medical care at the forefront of our practices.

We, as health care providers and physician trainees, hereby pledge:

  • To continue to provide evidence-based, nonjudgmental counseling for all pregnancy options, including abortion, and support our patients through all reproductive health decisions
  • To promote equity in providing comprehensive reproductive health care, recognizing the impacts of systemic racism and oppression
  • To promote high quality training in providing safe reproductive care in our respective institutions
  • To use our voices in our communities to advocate for all our patients to have the freedom to access the safe and compassionate health care they deserve.

Sincerely,

The undersigned 747 ObGyn resident physicians

Please note that we sign this statement on our own behalf as individuals and not on behalf of our respective institutions.

Orchideh Abar, MD

Laurel S. Aberle, MD

Kathleen E. Ackert, DO

Lauryn Adams, MD

Temiloluwa Adejuyigbe, MD

Oluwatoyosi M. Adeoye, MD

Hufriya Y. Aderianwalla, MD

Fareeza Afzal, MD

Adelaide Agyepong, MD

Erin R. Ahart, MD

Noha T. Ahmed, DO

Faria Ahmed, MD

Tracey O. Akanbi, MD

Eloho E. Akpovi, MD

Austin H. Allen, DO

Amanda M. Allen, MD

Alexis L. Allihien, MD

Jorge L. Alsina, MD

Paulina C. Altshuler, DO

Sivani Aluru, MD

Amal Amir, DO

Jon Anderson, DO

Andreas Antono, MD

Annie N. Apple, MD

Janine Appleton, DO

Aarthi Arab, MD

Sydney R. Archer, MD

Youngeun C. Armbuster, MD

Kara Arnold, MD

Blessing C. Aroh, MD

Savannah Pearson Ayala, MD

Archana K. Ayyar, MD

Ann-Sophie Van Backle, DO

Connor R. Baker, MD

Japjot K. Bal, MD

Abigail E. Barger, MD

Kathryn E. Barron, MD

Silvia Bastea, MD

Samantha V.H. Bayer, MD

Kristen Beierwaltes, MD

Gisel Bello, MD

Michelle A. Benassai, MD

Dana Benyas, MD

Alice F. Berenson, MD

Hanna P. Berlin, MD

Abigail L. Bernard, MD

Eli H. Bernstein, MD

Julia T. Berry, MD

Bryce L. Beyer, MD

Caroline Bilbe, MD

Grace E. Binter, DO

Erin E. Bishop, MD

Sierra G. Bishop, MD

Stephanie S. Bista, MD

Tara E. Bjorklund, DO

Alyssa N. Black, MD

Continue to: Kelsey Boghean, DO...

 

 

Kelsey Boghean, DO

Areta Bojko, MD

Grace E. Bommarito, DO

Aditi R. Bommireddy, MD

Genna C. Bonfiglio, MD

Mary E. Booker, MD

Kayce L. Booth, MD

Samantha T. Boothe, DO

William Borenzweig, MD

Rebecca M. Borneman, MD

Alexander L. Boscia, MD

Gina M. Botsko, MD

Glenn P. Boyles, MD

Avery C. Bramnik, MD

Sophia N. Brancazio, MD

Katarina M. Braun, MD

Anthony Brausch, MD

Emily L. Brekke, MD

Sara E. Brenner, MD

Bailey A. Brown, DO

Kathryn S. Brown, MD

Denese C. Brown, MD

Abena Bruce, MD

Sabrina C. Brunozzi, MD

Madison Buchman, DO

Deirdre G. Buckley, MD

Rachel L. Budker, MD

Leeann M. Bui, MD

Anthony H. Bui, MD

Jessie Bujouves, MD

Kimberley A. Bullard, MD

Sophia G. Bunde, MD

Emily R. Burdette, MD

Iris Burgard, DO

Korbi M. Burkey, MD

Lindsey K. Burleson, MD

Lindsay M. Burton, MD

Brianna N. Byers, MD

Stephanie Cai, MD

Alexandra S. Calderon, MD

Alexandra G. Caldwell, MD

Natalia Calzada, MD

Tamara Cameo, MD

Arielle Caplin, MD

Angela M. Carracino, DO

Anna L. Carroll, MD

Leigha M. Carryl, MD

Ashlie S. Carter, MD

Stephanie Casey, DO

Chase W. Cataline, DO

Carson L. Catasus, MD

Alena R. Cave, MD

Kelly M. Chacon, MD

Avis L. Chan, MD

Shruthi Chandra, MD

Jennifer Chang, MD

Shannon Chang, DO

Gillian Chase, MD

Cindy Chen, MD

Jessie C. Chen, MD

Jessica T. Chen, MD

Wenjin Cheng, MB

Laura J. Cheng, MD

Lucy Cheng, MD

Monica S. Choo, MD

Jody S. Chou, MD

Hannah C. Christopher, DO

Continue to: David J. Chromey, DO...

 

 

David J. Chromey, DO

Grace V. Clark, MD

Celeste Colegrove, MD

Sarah C. Combs, MD

Victoria L. Conniff, MD

Hannah C. Connor, MD

Angela J. Conway, MD

Steffany A. Conyers, MD

Alexandra Cooke, MD

Ashley A. Cooney, MD

Anna Cornelius-Schecter, MD

Alexa M. Corso, DO

Krysten A. Costley, MD

Madeline Coulter, MD

Kelsey Cramer, MD

Anna E. Cronin, MD

Bethany N. Croyle, DO

Carmen A. Cueto, MD

Nicole Cumbo, MD

Mackenzie A. Cummings, MD

Carrie Cummiskey, MD

Hannah M. Cunningham, MD

Sarah D’Souza, DO

Rachael M. D’Auria, MD

Caitlin Dane, MD

Rachel N. Dang, MD

Talin R. Darian, MD

Abigail C. Davies, MD

Berkley Davis, MD

Lois A. Davis, MD

Jennie J. DeBlanc, MD

Ayana G.R. DeGaia, MD, MPH

Katerina N. DeHaan, MD

Rebekka M. Delgado, MD

Brettany C. DeMier, MD

Bonnie W. DePaso, MD

Hemaxi H. Desai, DO

Amberly T. Diep, MD

Abigail K. Dillaha, MD

Sarah K. Dominguez, MD

Abbey P. Donahue, MD

Allan C. Dong, MD

James Doss, MD

Taylor B. Douglas, MD

Abigail G. Downey, MD

Janelle M. Driscoll, MD

Emily Du, MD

Leslie V. Dunmire, MD

Jennifer Duong, DO

Leigh C. Durudogan, MD

Mai N. Dyer, MD, MPH

Rebecca A. Ebbott, MD

Lindsey P. Eck, MD

Molly C. Eckman, MD

Alex Ede, MD, ScM

Claire E. Edelman, MD

Sara E. Edwards, MD

David J. Eggert, DO

Michelle Eide, MD

Etoroabasi Ekpe, MD

Tressa L. Ellett, MD

Laura Peyton Ellis, MD

Kaitlin H. Ellis, MD

Mariah G. Elly, MD

Jennifer Embry, MD

Claire Englert, MD

Brenna Espelien, MD

Kamilah Evans, MD

Joshua A. Ewy, MD

Elana D. Fackler, MD

Lauren E. Falk, MD

Brianna A. Farley, MD

Amanda Stephanie R. Farrell, MD

Sara Fassio, DO

Daniela A. Febres-Cordero, MD

Jasmin E. Feliciano, MD

Alayna H. Feng, MD

Amanda M. Ferraro, MD

Brittany A. Fickau, MD

Brittany H. File, MD

Shannon M. Finner, DO

Mia E. Fischbein, DO

Briah Fischer, MD

Shira Fishbach, MD

Alison C. Fitzgerald, MD

Evan R. Fitzgerald, MD

Margaret R. Flanigan, MD

Kevin C. Flatley, MD

Jordan A. Fletcher, MD

Claudia E. Flores, MD

Lauren A. Forbes, MD

Rana K. Fowlkes, MD

Jennifer M. Franks, MD, MPH

Christina M. Frasik, MD

Haven N. Frazier, DO

Sarah W. Freeman, MD

Emilie O. Fromm, DO

Anna R. Fuchss, MD

Emma K. Gaboury, MD

Madeline H. Ganz, MD

Lex J. Gardner, MD

Keri-Lee Garel, MD

Hailey B. Gaskamp, DO

Brittney A. Gaudet, MD

Gabrielle M. Gear, MD

Eleanor R. Germano, MD

Lauren G. Gernon, MD

Allen Ghareeb, MD

Patricia Giglio Ayers, MD

Jordana L. Gilman, MD

Mianna M. Gilmore, DO

Brian W. Goddard, MD

Julia L. Goldberg, MD

M. Isabel Gonzaga, MD

Fred P. Gonzales, MD

Lillian H. Goodman, MD, MPH

Ashley Goreshnik, MD

Lauren E. Gottshall, MD

Lindsay L. Gould, MD

Kelsea R. Grant, MD

Dorender A. Gray, MD

Sophie Green, MD

Erica A. Green, MD

Danielle C. Greenberg, MD

Kalin J. Gregory-Davis, MD

David M. Greiner, MD

Tyler M. Gresham, MD

Continue to: Nelly Grigorian, MD...

 

 

Nelly Grigorian, MD

Erin L. Grimes, MD

Whitney Grither, MD

Jared M. Grootwassink, MD

Maya E. Gross, MD

Paoula Gueorguieva, MD

Margot M. Gurganus, DO

Rachel L. Gutfreund, MD

Andres Gutierrez, MD

Dorothy L. Hakimian, DO

Ashley N. Hamati, DO

Marie M. Hanna-Wagner, MD

Katie Hansen, MD

Courtney Hargreaves, MD

Stephanie Harlow, MD

Kelsey B. Harper, MD

Devon A. Harris, MD

Lauren E. Harris, MD

Emily S. Hart, DO

Sarah A. Hartley, MD

Becky K. Hartman, MD

Abigail K. Hartmann, MD

Charlotte V. Hastings, MD

Cherise Hatch, DO

Jordan Hauck, DO

Sarena Hayer, MD

Jenna M. Heath, MD

Eric D. Helm, MD

Julie A. Hemphill, MD

Ric A.S. Henderson, MD

Nicola A. Hendricks, MD

Andrea A. Henricks, MD

Jesse M. Herman, DO

Alyssa M. Hernandez, DO

Melissa Hernandez, MD

Alyssa R. Hersh, MD

Alexandra Herweck, MD

Brianna Hickey, MD

Allix M. Hillebrand, MD

Alessandra I. Hirsch, MD

Emily A. Hoffberg, MD

Chloe L. Holmes, DO

Cameron M. Holmes, MD

Helena Y. Hong, MD

Wakako Horiuchi, MD

Shweta Hosakoppal, MD

Jaycee E. Housh, MD

Shannon M. Howard, MD

Meredith C. Huszagh, MD

Yihharn P. Hwang, MD

Emma C. Hyde, MD

Brooke Hyman, MD

Hala Ali Ibrahim, MD

Gnendy Indig, MD

Erin E. Isaacson, MD

Shruti S. Iyer, DO

Audrey J. Jaeger, DO

Shobha Jagannatham, MD

Cyrus M. Jalai, MD

Emma V. James, MD

Isabel Janmey, MD

Phoebe Jen, DO

Corey L. Johnson, MD

Crystal J. Johnson, MD

Andrea M. Johnson, MD

Nat C. Jones, MD

Briana L. Jones, DO

Rebecca J. Josephson, MD

Sarah Natasha Jost-Haynes, MD

 

Continue to: Hannah S. Juhel, MD...

 

 

Hannah S. Juhel, MD

Erin Jun, DO

Katherine B. Kaak, MD

Dhara N. Kadakia, MD

Amanda D. Kadesh, MD

Riana K. Kahlon, MD

Nadi N. Kaonga, MD

Moli Karsalia, MD

Stephanie L. Kass, MD

Amanda M. Katz, MD

Chelsea S. Katz, MD

Virginia Kaufman, MD

Gurpinder Kaur, MD

Jessica A. Keesee, MD

Cassandra N. Kelly, MD

Whitney Kelly, DO

Hannah V. Kennedy, MD

Bethany H. Kette, MD

Iman Khan, MD

Maryam M. Khan, MD

Alisa Jion Kim, MD

Tesia G. Kim, MD

Anne E. Kim, MD

Emily H. King, MD

Tarynne E. Kinghorn, MD

Holly T. Kiper, DO

Thomas Kishkovich, MD

Quinn M. Kistenfeger, MD

Sofia E. Klar, DO

Jessica B. Klugman, MD

Hope E. Knochenhauer, MD

Kathleen J. Koenigs, MD

Olga Kontarovich, DO

Alison Kosmacki, MD

Ana E. Kouri, MD

Olga M. Kovalenko, MD

Leigh T. Kowalski, MD

Kayla A. Krajick, MD

Elizabeth S. Kravitz, MD

Shruti Rani Kumar, MD

Alyssa Kurtz, DO

Lauren H. Kus, MD

Arkadiy Kusayev, DO

Amanda E. Lacue, MD

Nava Lalehzari, MD

Amber Lalla, MD

Allie C. Lamari, DO

Kelly L. Lamiman, MD

Stephen Lammers, MD

Monet Lane, MD

Madeline L. Lang, MD

Liana Langdon-Embry, MD

Carolyn Larkins, MD

Leah E. Larson, MD

Matthew W. Lee, MD

Eunjae Lee, MD

Alice Lee, MD

Jared Z. Lee, MD

Charlotte M. Lee, MD

Nicole R. Legro, MD

Aurora Leibold, MD

Rosiris Leon-Rivera, MD, PhD

Anna M. Leone, MD

Keiko M. Leong, MD

Lindsey M. LePoidevin, MD

Molly E. Levine, MD

Khrystyna Levytska, MD

Dana L. Lewis, DO

Jessica L. Li, MD

Kristina Lilja, MD

Deanna M. Lines, DO

Annalise Littman, MD

Julia F. Liu, MD

Tyler B. Lloyd, MD

Alyssa Lo, MD

K’ara A. Locke, MD

Minica Long, MD

Melissa Lopez, MD

Wilfredo A. Lopez, MD

Connie F. Lu, MD

Tyler J. Lueck, MD

Katherine L. Lukas, MD

Davlyn L. Luke, MD

Shani Ma, MD

Colton Mabis, MD

Lauren T. MacNeill, MD

Rachel Madding, MD

Mona Makhamreh, MD

Francesca R. Mancuso, MD

Kelsey L. Manfredi, MD

Valeria Mantilla, MD

Kaitlin M. Mar, MD

Starcher R. Margaret, MD

Audrey M. Marinelli, MD

Brittany A. Marinelli, MD

Emily S. Markovic, MD

Hannah L. Marshall, MD

Aaron Masjedi, MD

Isabelle M. Mason, MD

Akailah T. Mason-Otey, MD

Nicole Massad, MD

Megan M. Masten, MD

Stephanie M. Masters, MD

Anastasia Matthews, MD

Natalia del Mazo, MD

Sara A. McAllaster, MD

Continue to: Nicole McAndrew, DO...

 

 

Nicole McAndrew, DO

Madeline G. McCosker, MD

Jamie L. McDowell, DO

Christine E. McGough, MD

Mackenzi R. McHugh, MD

Madeline M. McIntire, MD

Cynthia R. McKinney, MD

Kirsten D. McLane, MD

Shian F. McLeish, MD

Megan I. McNitt, MD

Sarah R. McShane, MD

Grace R. Meade, MD

Nikki Ann R. Medina, DO

Tiffany L. Mei, MD

Jenna Meiman, MD

Anna M. Melicher, MD

Rosa M. Mendez, MD

Riley Mickelsen, MD

Sage A. Mikami, MD

Aletheia B. Millien, MD

Hannah C. Milthorpe, MD

Caroline J. Min, MD

Julie A. Mina, MD

Annie G. Minns, MD

Natalie Mironov, DO

Elizabeth L. Mirsky, MD

Astha Mittal, MD

Rachel E. Mnuk, MD

Silki Modi, MD

Sudarshan J. Mohan, MD

Roxana Mohhebali-Solis, MD

Mugdha V. Mokashi, MD

Jessica A. Montgomery, MD

Ellen Moore, MD

Savannah J. Morehouse, MD

Kristen L. Moriarty, MD

Alexa P. Morrison, MD

Bijan Morshedi, MD

Matthew H. Mossayebi, MD

Kathy Mostajeran, DO

Sharan Mullen, DO

Ellen C. Murphy, MD

Emma Chew Murphy, MD

Lauren M. Murphy, MD

Bria Murray, MD

Erin C. Nacev, MD

Preetha Nandi, MD

Blaire E. Nasstrom, DO

Hallie N. Nelson, MD

Katherine A. Nelson, MD

Margaret S. Nemetz, MD

Daniela Ben Neriah, DO

Cosima M. Neumann, MD

Mollie H. Newbern, DO

Gisella M. Newbery, MD

Stephanie Nguyen, MD

Christine G.T. Nguyen, MD

Desiree Nguyen, MD

Jacqueline W. Nichols, MD

Annika M. Nilsen, MD

Margaret A. Nixon, MD

Emily M. Norkett, MD

Allison N. Nostrant, DO

Susan E. Nourse, MD

Aliya S. Nurani, MD

Emily E. Nuss, MD

Jeanne O. Nwagwu, DO

Kelsey E. O’Hagan, MD

Margaret O’Neill, MD

Emily A. O’Brien, MD

Carly M. O’Connor-Terry, MD, MS

Madison O. Odom, MD

Cynthia I. Okot-Kotber, MD

Sarah P. Oliver, MD

Leanne P. Ondreicka, MD

Ngozika G. Onyiuke, MD

Erika Gonzalez Osorio, MD

Marika L. Osterbur Badhey, MD

Linda A. Otieno, MD

Claire H. Packer, MD

Chloe W. Page, DO

Marissa Palmor, MD

Rishitha Panditi, MD

Katherine A. Panushka, MD

Kelsey J. Pape, MD

Rachel R. Paquette, DO

Hillary C. Park, DO

Kendall M. Parrott, MD

Ekta Partani, MD

Karishma Patel, MD

Shivani Patel, MD

Continue to: Priya Patel, MD...

 

 

Priya Patel, MD

Jenna M. Patterson, MD

Ashleigh Pavlovic, MD

Katie M. Peagler, MD

Katherine T. Pellino, MD

Nicholas Per, MD

Elana Perry, MD

Emily J. Peters, MD

Sara E. Peterson, MD

Michelle R. Petrich, MD

Destiny L. Phillips, MD

Chloe Phillips, MD

Megan E. Piacquadio, DO

Sara C. Pierpoint, MD

Celeste M. Pilato, MD

Emma Pindra, MD

Minerva L.R. Pineda, MD

Rebecca Pisan, MD

Alessandra R. Piscina, MD

Rachael Piver, MD

Andrew J. Polio, MD

Hector S. Porragas, MD

Natalie Posever, MD

Allison R. Powell, MD

Mahima V. Prasad, MD

Angelina D. Prat, DO

Rebecca L. Purvis, MD

Teresa L. Qi, MD

Nicholas R. Quam, MD

Candice A. Quarella, MD

Nicholas W. Racchi, DO

Jeannie G. Radoc, MD

Samuel Raine, MD

Anna C. Raines, MD

Stephanie A. Rains, MD

Nicole M. Rainville, DO

Karissa Rajagopal, DO

Kristian R. Ramage, MD

Praveen Ramesh, MD

Tia M. Ramirez, MD

Jania Ramos, MD

Neel K. Rana, MD

Urvi Rana, DO

Indira Ranaweera, MD

Sindhuja Ranganathan, DO

Chloe R. Rasmussen, MD

Laura P. Reguero-Cadilla, MD

Devin M. Reilly, MD

Kimberly E. Reimold, MD

Cory R. Reiter, MD, PhD

Maya E. Reuven, DO

Jessica Reyes-Peterson, MD

Jacqueline Rice, MD

Rebecca L. Richardson, MD

Mikaela J. Rico, DO

Katelyn Rittenhouse, MD

Giuliana A. Rivera Casul, MD

Jill N.T. Roberts, MD

Luke N. Roberts, MD

Esther Robin, MD

Marcella Israel Rocha, MD

Zoe A. Roecker, MD

Hilary E. Rogers, MD

Kelsey A. Roof, MD

Zarah Rosen, MD

Cecilia M. Rossi, MD

Eva S. Rostonics, MD

Felix Rubio, MD

Amela Rugova, MD

Anna J. Rujan, MD

Erika T. Russ, MD

Colin Russell, MD

Ruby L. Russell, MD

Isabella A. Sabatina, MD

Gouri Sadananda, MD

Aashna Saini, MD

Salomeh M. Salari, MD

Ndeye N. Sall, MD

Nicole M. Salvador, MD

Aayushi Sardana, MD

Kendall M. Sarson, MD

Rita Abigail Sartor, MD

Continue to: Haley A. Scarbrough, MD...

 

 

Haley A. Scarbrough, MD

Kimberly Schaefer, MD

Demetra Schermerhorn, MD

Ellen C. Schleckman, MD

Maura A. Schlussel, MD

Ellie Schmidt, MD

Alison M. Schmidt, MD

Evan A. Schrader, MD

Morgan A. Schriever, MD

Brianna L. Schumaker Nguyen, DO

Whitney E. Scott, MD

Claire Scrivani, MD

Catherine E. Seaman, MD

Rachel D. Seaman, MD

Danielle J. Seltzer, MD

Joshua R. Shaffer, MD

Emily A. Shaffer, MD

Delia S. Shash, MD

Ishana P. Shetty, MD

Tushar Shetty, MD

Carol Shi, MD

Sarah P. Shim, MD

Emma C. Siewert, MD

Seth M. Sigler, DO

Rebecca L. SigourneyTennyck, MD

Daniella D. Silvino, DO

Andrea M. Simi, MD

Amelia R. Simmons, MD

Amy E. Skeels, DO

Ashley E.S. Keith, MD

Hannah C. Smerker, DO

Katarina Smigoc, MD

Madeline I. Smith, MD

Jessica D. Smith, MD

Melanie R. Smith, MD

Alicia L. Smith, MD

Chloe Smith, MD

Ayanna Smith, MD

Melanie R. Smith, MD

Megan M. Smith, MD

Haverly J. Snyder, MD

Beatrice R. Soderholm, DO

Brianna C. Sohl, MD

Samantha A. Solaru, MD

Michael Solotke, MD

Dara A.H. Som, MD

Alexandra R. Sotiros-Lowry, MD

Melanie Spall, DO

Alicia C. Speak, DO

Lisa M. Spencer, MD

Prakrithi Srinand, MD

Sierra M. Starr, MD

Kathryne E. Staudinger, MD

Emily K. Steele, MD

Morgan R. Steffen, DO

Tricia R. Stepanek, MD

Taylor P. Stewart, MD

Kelsey A. Stewart, MD

Alyssa M. Stiff, MD

Alexandra B. Stiles, MD

Nairi K. Strauch, MD

Margaret J. Stroup, DO

Sean C. Stuart, DO

Hannah M. Stump, MD

Shalini B. Subbarao, MD

Lakshmi Subramani, MD

Heather E. Sweeney, MD

Kristin I. Swope, MD

Suha Syed, MD

Mireya P. Taboada, MD

Eneti S. Tagaloa, MD

Rachel Tang, DO

Adam R. Taylor, MD

Simone R. Thibault, MD

Kimberly A. Thill, MD

Dhanu Thiyag, MD

Andrew T. Thornton, MD

Wendy Tian, MD

Stephanie Tilberry, MD

Amanda L. Tillett, MD

Amanda M. Tjitro, MD

Logan P. Todhunter, DO

David Toffey, MD

Maris K. Toland, MD

Rachel E. Tomassi, MD

Sarah Tounsi, MD

Antonia K. Traina, MD

Taylor Tran, MD

Diem Samantha Tran, DO

Emily C. Trautner, MD

Emma Trawick, MD

Continue to: Elissa Trieu, MD...

 

 

Elissa Trieu, MD

Ariel Trilling, MD

Samantha Truong, MD

Mary M. Tsaturian, MD

Athena Tudino, MD

Kati A. Turner, MD

Nicole-Marie Tuzinkiewicz, MD

Gayathri D. Vadlamudi, MD

Stylianos Vagios, MD

Pauline V. Van Dijck, DO

Kaylee A. VanDommelen, MD

Isha B. Vasudeva, MD

Shivani J. Vasudeva, DO

Diana Q. Vazquez Parker, MD

Ridhima Vemula, MD

Elena C. Vinopal, MD

Caroline J. Violette, MD

Pascal T. Vo, DO

Michelle H. Vu, MD

Macy M. Walz, MD

Angelia Wang, MD

Eileen Wang, MD

Courtney Y. Wang, MD

Joyce Wang, MD

Meryl G. Warshafsky, MD

Sophie E.N. Weinstein, MD

Sarah H. Weinstein, MD

Annalyn M. Welp, MD

Shannon M. Wentworth, MD

Erika M. Wert, MD

Rachel C. White, MBchB

Morgan N. Wilhoite, DO

Mercedes Williams, MD

Hayley Williams, MD

Jacquelyn D. Williams, MD

Mary H. Williamson, MD

Elise Wilson, MD

Lauren M. Witchey, MD

Emily A. Wolverton, MD

Stephanie Y. Wong, MD

Jenny Wu, MD

Jackie Xiang, MD

Nancy S. Yang, MD

Kevin P. Yeagle, MD

Halina M. Yee, MD

Alyssa M. Yeung, MD

Samuel K. Yost, MD

Megan Yuen, MD

Nayab Zafar, DO

Cindy X. Zhang, DO

Yingao Zhang, MD

Helen Zhao, MD

Chelsea Zhu, MD

Billie E. Zidel, MD

Ryan A. Zoldowski, MD

References

 

  1. Vinekar K, Karlapudi A, Nathan L, et al. Projected implications of overturning Roe v Wade on abortion training in US obstetrics and gynecology residency programs. Obstet Gynecol. 2022;140:146-149.
  2. ACGME program requirements for graduate medical education in obstetrics and gynecology summary and impact of interim requirement revisions. ACGME website. Accessed December 18, 2022. https://www.acgme.org/globalassets/pfassets/reviewandcomment/220_obstetricsandgynecology_2022-06-24_impact.pdf
  3. Crear-Perry J, Hassan A, Daniel S. Advancing birth equity in a post-Dobbs US. JAMA. 2022;328:1689-1690.
  4. Report on residents. AAMC website. Accessed December 18, 2022. https://www.aamc.org/data-reports/students-residents/interactive-data/report-residents/2021/table-c4-physician-reten tion-state-residency-training-last-completed-gme
Article PDF
Author and Disclosure Information

Dr. Nandi is Resident, Tufts University School of Medicine, Boston, Massachusetts.

Dr. Toland is Resident, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire.

Dr. Evans is Associate Director for Residency Program and Assistant Professor, Obstetrics and Gynecology, Tufts University School of Medicine.

The authors report no financial relationships relevant to this article.

Issue
OBG Management - 35(1)
Publications
Topics
Page Number
45-46, e47-e51
Sections
Author and Disclosure Information

Dr. Nandi is Resident, Tufts University School of Medicine, Boston, Massachusetts.

Dr. Toland is Resident, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire.

Dr. Evans is Associate Director for Residency Program and Assistant Professor, Obstetrics and Gynecology, Tufts University School of Medicine.

The authors report no financial relationships relevant to this article.

Author and Disclosure Information

Dr. Nandi is Resident, Tufts University School of Medicine, Boston, Massachusetts.

Dr. Toland is Resident, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire.

Dr. Evans is Associate Director for Residency Program and Assistant Professor, Obstetrics and Gynecology, Tufts University School of Medicine.

The authors report no financial relationships relevant to this article.

Article PDF
Article PDF

Photo: Shutterstock

Six months after the Supreme Court decision that overturned the constitutional right to abortion, trainees across the United States are asking a critical question in the current resident recruitment season: How will the restrictions on abortion access affect my training as an obstetrician-gynecologist, and will they impact my ability to be the kind of provider I want to be in the future?

Among the myriad of downstream effects to patient care, the Dobbs decision will indisputably impact the scope of residency training for those that provide reproductive health services. Almost half of ObGyn residents train in states that have abortion restrictions in place.1 New educational milestones for abortion training, which are a requirement by the Accreditation Council for Graduate Medical Education (ACGME), were proposed quickly after Dobbs, guiding programs to offer opportunities for training in nonrestricted areas or the “combination of didactic activities, including simulation” to meet the training requirement in abortion care.2

Like many providers, residents already are grappling with precarious and risky circumstances, balancing patient safety and patient-driven care amidst pre-existing and newly enforced abortion restrictions. Whether managing a patient with an undesired pregnancy, severe medical comorbidities, unexpected pregnancy complications such as preterm premature rupture of membranes, or bleeding, or substantial fetal anomalies, ObGyn residents cannot gain the experience of providing the full scope of reproductive health care without the ability to offer all possible management options. While some enacted abortion restrictions have exceptions for the health of or life-saving measures for the mother, there is no standard guidance for timing of interventions, leaving providers confused and in fear of legal retribution. At a time when trainees should be learning to provide patient-centered, evidence-based care, they are instead paralyzed by the legal or professional consequences they may face for offering their best medical judgements.

Furthermore, the lack of exposure to dilation and evacuation procedures for residents in restricted practice areas will undoubtably decrease their confidence in managing acute complications, which is one of the critical facets of residency training. In a surgical field where repetition is crucial for technical competence, highlighted by ACGME minimum case requirements, the decreased volume of abortion procedures is a disadvantage for trainees and a disservice for patients. While anti-choice promoters may argue that involvement in surgical management of early pregnancy loss should suffice for ObGyn training in family planning, this piecemeal approach will leave gaps in technical skills.

The fear of legal ramifications, moral injury, and inadequate surgical training may lead to the siphoning of talented trainees to areas in the country with fewer restrictions.3Dobbs already has demonstrated how limiting abortion access will deepen inequities in reproductive health care service delivery. Approximately 55% of ObGyn trainees and nearly two-thirds of maternal-fetal medicine graduates join the workforce in the state where they received their training.4 Medical students will seek opportunities for high-quality ObGyn training in areas that will help them to be well-prepared, competent physicians—and more often than not, stay in the area or region that they trained in. This will lead to provider shortages in areas where access to reproductive health care and subspecialist providers already is limited, further exacerbating existing health disparities.

During this recruitment season, trainees and residency programs alike will need to reckon with how the ramifications of Dobbs will alter both the immediate and long-term training in comprehensive reproductive health care for the ObGyn workforce. ObGyn trainees have taken a stand in response to the Dobbs decision, and nearly 750 current residents signed onto the statement below as a commitment to high-quality training and patient-centered care. Clinical experience in performing abortions is essential to the provision of comprehensive evidence-based reproductive health care, and access to these procedures is as important for physicians-in-training as it is for patients.

Actions to take to ensure high-quality abortion training in ObGyn residencies include the following:

  • Connect with and stay involved with organizations such as the American College of Obstetricians and Gynecologists (ACOG), Physicians for Reproductive Health (PRH), and Medical Students for Choice (MSFC) for initiatives, toolkits, and resources for training at your institutions.
  • Seek specific abortion training opportunities through the Leadership Training Academy (offered through PRH) or the Abortion Training Institute (offered through MSFC).
  • Ensure that your residency program meets the ACGME criteria of providing opportunities for clinical experiences for abortion care and work with program leadership at a program, state, or regional level to enforce these competencies.
  • Reach out to your local American Civil Liberties Union or other local reproductive legal rights organizations if you want to be involved with advocacy around abortion access and training but have concerns about legal protections.
  • Have a voice at the table for empowering training opportunities by seeking leadership positions through ACOG, ACGME, Council on Resident Education in Obstetrics and Gynecology and the Association of Professors of Gynecology and Obstetrics, American Medical Association, Student National Medical Association, and subspecialty organizations.
  • Vote in every election and promote voting registration and access to your patients, colleagues, and communities. ●

Continue to: The implications of the Dobbs v Jackson Women’s Health Organization decision on the health care and wellbeing of our patients...

 

 

The implications of the Dobbs v Jackson Women’s Health Organization decision on the health care and wellbeing of our patients

On June 24, 2022, the Supreme Court of the United States ruled in a 6-3 majority decision to overturn the constitutional right to abortion protected by Roe v Wade since 1973. As health care providers, we are outraged at the Court’s disregard for an individual’s right to make reproductive decisions for themselves and their families and are deeply concerned about the devastating consequences to reproductive care and outcomes in this country for all people. Reproductive health decisions, including growing a family and whether or not to continue a pregnancy, are complex and incredibly personal. Our role as health care providers is to help guide those decisions with empathy and evidencebased clinical recommendations. This ruling undermines a patient’s right to bodily autonomy, free of impositions from government and political pressures, and it threatens the sanctity of complex medical decision-making between a patient, their family, and their medical team.

As medical professionals, we know that every patient’s situation is unique—banning abortion procedures ties the hands of physicians trying to provide the most medically appropriate options in a compassionate manner. We know that both medical and surgical abortions are safe and can save lives. These procedures can help patients with potentially life-threatening conditions worsened by pregnancy, a poor prognosis for the fetus, or a complication from the pregnancy itself. Physicians use scientific research and individualized approaches to help patients in unique situations, and attempts to legislate personal health decisions compromise the practice of evidence-based medicine.

We also know that this decision will impact some communities more than others. Access to safe abortion care will become dependent on which region of the country a person lives in and whether or not a person has resources to seek this care. Due to continued systemic racism and oppression, patients of color will be disproportionately impacted and likely will suffer worse health outcomes from unsafe abortions. Those that rely on public insurance or who are uninsured will face overwhelming barriers in seeking abortion services. These disparities in reproductive care, which contribute to our nation’s health crises in maternal morbidity and mortality, unintended pregnancy, and neonatal complications, will further entrench health inequities, and patient lives and livelihoods will suffer.

We acknowledge the impact that this decision will have on restricting access to reproductive care. We stand by the fact that abortion care is health care. We vow to uphold the tenets of our profession to place patient autonomy and provision of safe quality medical care at the forefront of our practices.

We, as health care providers and physician trainees, hereby pledge:

  • To continue to provide evidence-based, nonjudgmental counseling for all pregnancy options, including abortion, and support our patients through all reproductive health decisions
  • To promote equity in providing comprehensive reproductive health care, recognizing the impacts of systemic racism and oppression
  • To promote high quality training in providing safe reproductive care in our respective institutions
  • To use our voices in our communities to advocate for all our patients to have the freedom to access the safe and compassionate health care they deserve.

Sincerely,

The undersigned 747 ObGyn resident physicians

Please note that we sign this statement on our own behalf as individuals and not on behalf of our respective institutions.

Orchideh Abar, MD

Laurel S. Aberle, MD

Kathleen E. Ackert, DO

Lauryn Adams, MD

Temiloluwa Adejuyigbe, MD

Oluwatoyosi M. Adeoye, MD

Hufriya Y. Aderianwalla, MD

Fareeza Afzal, MD

Adelaide Agyepong, MD

Erin R. Ahart, MD

Noha T. Ahmed, DO

Faria Ahmed, MD

Tracey O. Akanbi, MD

Eloho E. Akpovi, MD

Austin H. Allen, DO

Amanda M. Allen, MD

Alexis L. Allihien, MD

Jorge L. Alsina, MD

Paulina C. Altshuler, DO

Sivani Aluru, MD

Amal Amir, DO

Jon Anderson, DO

Andreas Antono, MD

Annie N. Apple, MD

Janine Appleton, DO

Aarthi Arab, MD

Sydney R. Archer, MD

Youngeun C. Armbuster, MD

Kara Arnold, MD

Blessing C. Aroh, MD

Savannah Pearson Ayala, MD

Archana K. Ayyar, MD

Ann-Sophie Van Backle, DO

Connor R. Baker, MD

Japjot K. Bal, MD

Abigail E. Barger, MD

Kathryn E. Barron, MD

Silvia Bastea, MD

Samantha V.H. Bayer, MD

Kristen Beierwaltes, MD

Gisel Bello, MD

Michelle A. Benassai, MD

Dana Benyas, MD

Alice F. Berenson, MD

Hanna P. Berlin, MD

Abigail L. Bernard, MD

Eli H. Bernstein, MD

Julia T. Berry, MD

Bryce L. Beyer, MD

Caroline Bilbe, MD

Grace E. Binter, DO

Erin E. Bishop, MD

Sierra G. Bishop, MD

Stephanie S. Bista, MD

Tara E. Bjorklund, DO

Alyssa N. Black, MD

Continue to: Kelsey Boghean, DO...

 

 

Kelsey Boghean, DO

Areta Bojko, MD

Grace E. Bommarito, DO

Aditi R. Bommireddy, MD

Genna C. Bonfiglio, MD

Mary E. Booker, MD

Kayce L. Booth, MD

Samantha T. Boothe, DO

William Borenzweig, MD

Rebecca M. Borneman, MD

Alexander L. Boscia, MD

Gina M. Botsko, MD

Glenn P. Boyles, MD

Avery C. Bramnik, MD

Sophia N. Brancazio, MD

Katarina M. Braun, MD

Anthony Brausch, MD

Emily L. Brekke, MD

Sara E. Brenner, MD

Bailey A. Brown, DO

Kathryn S. Brown, MD

Denese C. Brown, MD

Abena Bruce, MD

Sabrina C. Brunozzi, MD

Madison Buchman, DO

Deirdre G. Buckley, MD

Rachel L. Budker, MD

Leeann M. Bui, MD

Anthony H. Bui, MD

Jessie Bujouves, MD

Kimberley A. Bullard, MD

Sophia G. Bunde, MD

Emily R. Burdette, MD

Iris Burgard, DO

Korbi M. Burkey, MD

Lindsey K. Burleson, MD

Lindsay M. Burton, MD

Brianna N. Byers, MD

Stephanie Cai, MD

Alexandra S. Calderon, MD

Alexandra G. Caldwell, MD

Natalia Calzada, MD

Tamara Cameo, MD

Arielle Caplin, MD

Angela M. Carracino, DO

Anna L. Carroll, MD

Leigha M. Carryl, MD

Ashlie S. Carter, MD

Stephanie Casey, DO

Chase W. Cataline, DO

Carson L. Catasus, MD

Alena R. Cave, MD

Kelly M. Chacon, MD

Avis L. Chan, MD

Shruthi Chandra, MD

Jennifer Chang, MD

Shannon Chang, DO

Gillian Chase, MD

Cindy Chen, MD

Jessie C. Chen, MD

Jessica T. Chen, MD

Wenjin Cheng, MB

Laura J. Cheng, MD

Lucy Cheng, MD

Monica S. Choo, MD

Jody S. Chou, MD

Hannah C. Christopher, DO

Continue to: David J. Chromey, DO...

 

 

David J. Chromey, DO

Grace V. Clark, MD

Celeste Colegrove, MD

Sarah C. Combs, MD

Victoria L. Conniff, MD

Hannah C. Connor, MD

Angela J. Conway, MD

Steffany A. Conyers, MD

Alexandra Cooke, MD

Ashley A. Cooney, MD

Anna Cornelius-Schecter, MD

Alexa M. Corso, DO

Krysten A. Costley, MD

Madeline Coulter, MD

Kelsey Cramer, MD

Anna E. Cronin, MD

Bethany N. Croyle, DO

Carmen A. Cueto, MD

Nicole Cumbo, MD

Mackenzie A. Cummings, MD

Carrie Cummiskey, MD

Hannah M. Cunningham, MD

Sarah D’Souza, DO

Rachael M. D’Auria, MD

Caitlin Dane, MD

Rachel N. Dang, MD

Talin R. Darian, MD

Abigail C. Davies, MD

Berkley Davis, MD

Lois A. Davis, MD

Jennie J. DeBlanc, MD

Ayana G.R. DeGaia, MD, MPH

Katerina N. DeHaan, MD

Rebekka M. Delgado, MD

Brettany C. DeMier, MD

Bonnie W. DePaso, MD

Hemaxi H. Desai, DO

Amberly T. Diep, MD

Abigail K. Dillaha, MD

Sarah K. Dominguez, MD

Abbey P. Donahue, MD

Allan C. Dong, MD

James Doss, MD

Taylor B. Douglas, MD

Abigail G. Downey, MD

Janelle M. Driscoll, MD

Emily Du, MD

Leslie V. Dunmire, MD

Jennifer Duong, DO

Leigh C. Durudogan, MD

Mai N. Dyer, MD, MPH

Rebecca A. Ebbott, MD

Lindsey P. Eck, MD

Molly C. Eckman, MD

Alex Ede, MD, ScM

Claire E. Edelman, MD

Sara E. Edwards, MD

David J. Eggert, DO

Michelle Eide, MD

Etoroabasi Ekpe, MD

Tressa L. Ellett, MD

Laura Peyton Ellis, MD

Kaitlin H. Ellis, MD

Mariah G. Elly, MD

Jennifer Embry, MD

Claire Englert, MD

Brenna Espelien, MD

Kamilah Evans, MD

Joshua A. Ewy, MD

Elana D. Fackler, MD

Lauren E. Falk, MD

Brianna A. Farley, MD

Amanda Stephanie R. Farrell, MD

Sara Fassio, DO

Daniela A. Febres-Cordero, MD

Jasmin E. Feliciano, MD

Alayna H. Feng, MD

Amanda M. Ferraro, MD

Brittany A. Fickau, MD

Brittany H. File, MD

Shannon M. Finner, DO

Mia E. Fischbein, DO

Briah Fischer, MD

Shira Fishbach, MD

Alison C. Fitzgerald, MD

Evan R. Fitzgerald, MD

Margaret R. Flanigan, MD

Kevin C. Flatley, MD

Jordan A. Fletcher, MD

Claudia E. Flores, MD

Lauren A. Forbes, MD

Rana K. Fowlkes, MD

Jennifer M. Franks, MD, MPH

Christina M. Frasik, MD

Haven N. Frazier, DO

Sarah W. Freeman, MD

Emilie O. Fromm, DO

Anna R. Fuchss, MD

Emma K. Gaboury, MD

Madeline H. Ganz, MD

Lex J. Gardner, MD

Keri-Lee Garel, MD

Hailey B. Gaskamp, DO

Brittney A. Gaudet, MD

Gabrielle M. Gear, MD

Eleanor R. Germano, MD

Lauren G. Gernon, MD

Allen Ghareeb, MD

Patricia Giglio Ayers, MD

Jordana L. Gilman, MD

Mianna M. Gilmore, DO

Brian W. Goddard, MD

Julia L. Goldberg, MD

M. Isabel Gonzaga, MD

Fred P. Gonzales, MD

Lillian H. Goodman, MD, MPH

Ashley Goreshnik, MD

Lauren E. Gottshall, MD

Lindsay L. Gould, MD

Kelsea R. Grant, MD

Dorender A. Gray, MD

Sophie Green, MD

Erica A. Green, MD

Danielle C. Greenberg, MD

Kalin J. Gregory-Davis, MD

David M. Greiner, MD

Tyler M. Gresham, MD

Continue to: Nelly Grigorian, MD...

 

 

Nelly Grigorian, MD

Erin L. Grimes, MD

Whitney Grither, MD

Jared M. Grootwassink, MD

Maya E. Gross, MD

Paoula Gueorguieva, MD

Margot M. Gurganus, DO

Rachel L. Gutfreund, MD

Andres Gutierrez, MD

Dorothy L. Hakimian, DO

Ashley N. Hamati, DO

Marie M. Hanna-Wagner, MD

Katie Hansen, MD

Courtney Hargreaves, MD

Stephanie Harlow, MD

Kelsey B. Harper, MD

Devon A. Harris, MD

Lauren E. Harris, MD

Emily S. Hart, DO

Sarah A. Hartley, MD

Becky K. Hartman, MD

Abigail K. Hartmann, MD

Charlotte V. Hastings, MD

Cherise Hatch, DO

Jordan Hauck, DO

Sarena Hayer, MD

Jenna M. Heath, MD

Eric D. Helm, MD

Julie A. Hemphill, MD

Ric A.S. Henderson, MD

Nicola A. Hendricks, MD

Andrea A. Henricks, MD

Jesse M. Herman, DO

Alyssa M. Hernandez, DO

Melissa Hernandez, MD

Alyssa R. Hersh, MD

Alexandra Herweck, MD

Brianna Hickey, MD

Allix M. Hillebrand, MD

Alessandra I. Hirsch, MD

Emily A. Hoffberg, MD

Chloe L. Holmes, DO

Cameron M. Holmes, MD

Helena Y. Hong, MD

Wakako Horiuchi, MD

Shweta Hosakoppal, MD

Jaycee E. Housh, MD

Shannon M. Howard, MD

Meredith C. Huszagh, MD

Yihharn P. Hwang, MD

Emma C. Hyde, MD

Brooke Hyman, MD

Hala Ali Ibrahim, MD

Gnendy Indig, MD

Erin E. Isaacson, MD

Shruti S. Iyer, DO

Audrey J. Jaeger, DO

Shobha Jagannatham, MD

Cyrus M. Jalai, MD

Emma V. James, MD

Isabel Janmey, MD

Phoebe Jen, DO

Corey L. Johnson, MD

Crystal J. Johnson, MD

Andrea M. Johnson, MD

Nat C. Jones, MD

Briana L. Jones, DO

Rebecca J. Josephson, MD

Sarah Natasha Jost-Haynes, MD

 

Continue to: Hannah S. Juhel, MD...

 

 

Hannah S. Juhel, MD

Erin Jun, DO

Katherine B. Kaak, MD

Dhara N. Kadakia, MD

Amanda D. Kadesh, MD

Riana K. Kahlon, MD

Nadi N. Kaonga, MD

Moli Karsalia, MD

Stephanie L. Kass, MD

Amanda M. Katz, MD

Chelsea S. Katz, MD

Virginia Kaufman, MD

Gurpinder Kaur, MD

Jessica A. Keesee, MD

Cassandra N. Kelly, MD

Whitney Kelly, DO

Hannah V. Kennedy, MD

Bethany H. Kette, MD

Iman Khan, MD

Maryam M. Khan, MD

Alisa Jion Kim, MD

Tesia G. Kim, MD

Anne E. Kim, MD

Emily H. King, MD

Tarynne E. Kinghorn, MD

Holly T. Kiper, DO

Thomas Kishkovich, MD

Quinn M. Kistenfeger, MD

Sofia E. Klar, DO

Jessica B. Klugman, MD

Hope E. Knochenhauer, MD

Kathleen J. Koenigs, MD

Olga Kontarovich, DO

Alison Kosmacki, MD

Ana E. Kouri, MD

Olga M. Kovalenko, MD

Leigh T. Kowalski, MD

Kayla A. Krajick, MD

Elizabeth S. Kravitz, MD

Shruti Rani Kumar, MD

Alyssa Kurtz, DO

Lauren H. Kus, MD

Arkadiy Kusayev, DO

Amanda E. Lacue, MD

Nava Lalehzari, MD

Amber Lalla, MD

Allie C. Lamari, DO

Kelly L. Lamiman, MD

Stephen Lammers, MD

Monet Lane, MD

Madeline L. Lang, MD

Liana Langdon-Embry, MD

Carolyn Larkins, MD

Leah E. Larson, MD

Matthew W. Lee, MD

Eunjae Lee, MD

Alice Lee, MD

Jared Z. Lee, MD

Charlotte M. Lee, MD

Nicole R. Legro, MD

Aurora Leibold, MD

Rosiris Leon-Rivera, MD, PhD

Anna M. Leone, MD

Keiko M. Leong, MD

Lindsey M. LePoidevin, MD

Molly E. Levine, MD

Khrystyna Levytska, MD

Dana L. Lewis, DO

Jessica L. Li, MD

Kristina Lilja, MD

Deanna M. Lines, DO

Annalise Littman, MD

Julia F. Liu, MD

Tyler B. Lloyd, MD

Alyssa Lo, MD

K’ara A. Locke, MD

Minica Long, MD

Melissa Lopez, MD

Wilfredo A. Lopez, MD

Connie F. Lu, MD

Tyler J. Lueck, MD

Katherine L. Lukas, MD

Davlyn L. Luke, MD

Shani Ma, MD

Colton Mabis, MD

Lauren T. MacNeill, MD

Rachel Madding, MD

Mona Makhamreh, MD

Francesca R. Mancuso, MD

Kelsey L. Manfredi, MD

Valeria Mantilla, MD

Kaitlin M. Mar, MD

Starcher R. Margaret, MD

Audrey M. Marinelli, MD

Brittany A. Marinelli, MD

Emily S. Markovic, MD

Hannah L. Marshall, MD

Aaron Masjedi, MD

Isabelle M. Mason, MD

Akailah T. Mason-Otey, MD

Nicole Massad, MD

Megan M. Masten, MD

Stephanie M. Masters, MD

Anastasia Matthews, MD

Natalia del Mazo, MD

Sara A. McAllaster, MD

Continue to: Nicole McAndrew, DO...

 

 

Nicole McAndrew, DO

Madeline G. McCosker, MD

Jamie L. McDowell, DO

Christine E. McGough, MD

Mackenzi R. McHugh, MD

Madeline M. McIntire, MD

Cynthia R. McKinney, MD

Kirsten D. McLane, MD

Shian F. McLeish, MD

Megan I. McNitt, MD

Sarah R. McShane, MD

Grace R. Meade, MD

Nikki Ann R. Medina, DO

Tiffany L. Mei, MD

Jenna Meiman, MD

Anna M. Melicher, MD

Rosa M. Mendez, MD

Riley Mickelsen, MD

Sage A. Mikami, MD

Aletheia B. Millien, MD

Hannah C. Milthorpe, MD

Caroline J. Min, MD

Julie A. Mina, MD

Annie G. Minns, MD

Natalie Mironov, DO

Elizabeth L. Mirsky, MD

Astha Mittal, MD

Rachel E. Mnuk, MD

Silki Modi, MD

Sudarshan J. Mohan, MD

Roxana Mohhebali-Solis, MD

Mugdha V. Mokashi, MD

Jessica A. Montgomery, MD

Ellen Moore, MD

Savannah J. Morehouse, MD

Kristen L. Moriarty, MD

Alexa P. Morrison, MD

Bijan Morshedi, MD

Matthew H. Mossayebi, MD

Kathy Mostajeran, DO

Sharan Mullen, DO

Ellen C. Murphy, MD

Emma Chew Murphy, MD

Lauren M. Murphy, MD

Bria Murray, MD

Erin C. Nacev, MD

Preetha Nandi, MD

Blaire E. Nasstrom, DO

Hallie N. Nelson, MD

Katherine A. Nelson, MD

Margaret S. Nemetz, MD

Daniela Ben Neriah, DO

Cosima M. Neumann, MD

Mollie H. Newbern, DO

Gisella M. Newbery, MD

Stephanie Nguyen, MD

Christine G.T. Nguyen, MD

Desiree Nguyen, MD

Jacqueline W. Nichols, MD

Annika M. Nilsen, MD

Margaret A. Nixon, MD

Emily M. Norkett, MD

Allison N. Nostrant, DO

Susan E. Nourse, MD

Aliya S. Nurani, MD

Emily E. Nuss, MD

Jeanne O. Nwagwu, DO

Kelsey E. O’Hagan, MD

Margaret O’Neill, MD

Emily A. O’Brien, MD

Carly M. O’Connor-Terry, MD, MS

Madison O. Odom, MD

Cynthia I. Okot-Kotber, MD

Sarah P. Oliver, MD

Leanne P. Ondreicka, MD

Ngozika G. Onyiuke, MD

Erika Gonzalez Osorio, MD

Marika L. Osterbur Badhey, MD

Linda A. Otieno, MD

Claire H. Packer, MD

Chloe W. Page, DO

Marissa Palmor, MD

Rishitha Panditi, MD

Katherine A. Panushka, MD

Kelsey J. Pape, MD

Rachel R. Paquette, DO

Hillary C. Park, DO

Kendall M. Parrott, MD

Ekta Partani, MD

Karishma Patel, MD

Shivani Patel, MD

Continue to: Priya Patel, MD...

 

 

Priya Patel, MD

Jenna M. Patterson, MD

Ashleigh Pavlovic, MD

Katie M. Peagler, MD

Katherine T. Pellino, MD

Nicholas Per, MD

Elana Perry, MD

Emily J. Peters, MD

Sara E. Peterson, MD

Michelle R. Petrich, MD

Destiny L. Phillips, MD

Chloe Phillips, MD

Megan E. Piacquadio, DO

Sara C. Pierpoint, MD

Celeste M. Pilato, MD

Emma Pindra, MD

Minerva L.R. Pineda, MD

Rebecca Pisan, MD

Alessandra R. Piscina, MD

Rachael Piver, MD

Andrew J. Polio, MD

Hector S. Porragas, MD

Natalie Posever, MD

Allison R. Powell, MD

Mahima V. Prasad, MD

Angelina D. Prat, DO

Rebecca L. Purvis, MD

Teresa L. Qi, MD

Nicholas R. Quam, MD

Candice A. Quarella, MD

Nicholas W. Racchi, DO

Jeannie G. Radoc, MD

Samuel Raine, MD

Anna C. Raines, MD

Stephanie A. Rains, MD

Nicole M. Rainville, DO

Karissa Rajagopal, DO

Kristian R. Ramage, MD

Praveen Ramesh, MD

Tia M. Ramirez, MD

Jania Ramos, MD

Neel K. Rana, MD

Urvi Rana, DO

Indira Ranaweera, MD

Sindhuja Ranganathan, DO

Chloe R. Rasmussen, MD

Laura P. Reguero-Cadilla, MD

Devin M. Reilly, MD

Kimberly E. Reimold, MD

Cory R. Reiter, MD, PhD

Maya E. Reuven, DO

Jessica Reyes-Peterson, MD

Jacqueline Rice, MD

Rebecca L. Richardson, MD

Mikaela J. Rico, DO

Katelyn Rittenhouse, MD

Giuliana A. Rivera Casul, MD

Jill N.T. Roberts, MD

Luke N. Roberts, MD

Esther Robin, MD

Marcella Israel Rocha, MD

Zoe A. Roecker, MD

Hilary E. Rogers, MD

Kelsey A. Roof, MD

Zarah Rosen, MD

Cecilia M. Rossi, MD

Eva S. Rostonics, MD

Felix Rubio, MD

Amela Rugova, MD

Anna J. Rujan, MD

Erika T. Russ, MD

Colin Russell, MD

Ruby L. Russell, MD

Isabella A. Sabatina, MD

Gouri Sadananda, MD

Aashna Saini, MD

Salomeh M. Salari, MD

Ndeye N. Sall, MD

Nicole M. Salvador, MD

Aayushi Sardana, MD

Kendall M. Sarson, MD

Rita Abigail Sartor, MD

Continue to: Haley A. Scarbrough, MD...

 

 

Haley A. Scarbrough, MD

Kimberly Schaefer, MD

Demetra Schermerhorn, MD

Ellen C. Schleckman, MD

Maura A. Schlussel, MD

Ellie Schmidt, MD

Alison M. Schmidt, MD

Evan A. Schrader, MD

Morgan A. Schriever, MD

Brianna L. Schumaker Nguyen, DO

Whitney E. Scott, MD

Claire Scrivani, MD

Catherine E. Seaman, MD

Rachel D. Seaman, MD

Danielle J. Seltzer, MD

Joshua R. Shaffer, MD

Emily A. Shaffer, MD

Delia S. Shash, MD

Ishana P. Shetty, MD

Tushar Shetty, MD

Carol Shi, MD

Sarah P. Shim, MD

Emma C. Siewert, MD

Seth M. Sigler, DO

Rebecca L. SigourneyTennyck, MD

Daniella D. Silvino, DO

Andrea M. Simi, MD

Amelia R. Simmons, MD

Amy E. Skeels, DO

Ashley E.S. Keith, MD

Hannah C. Smerker, DO

Katarina Smigoc, MD

Madeline I. Smith, MD

Jessica D. Smith, MD

Melanie R. Smith, MD

Alicia L. Smith, MD

Chloe Smith, MD

Ayanna Smith, MD

Melanie R. Smith, MD

Megan M. Smith, MD

Haverly J. Snyder, MD

Beatrice R. Soderholm, DO

Brianna C. Sohl, MD

Samantha A. Solaru, MD

Michael Solotke, MD

Dara A.H. Som, MD

Alexandra R. Sotiros-Lowry, MD

Melanie Spall, DO

Alicia C. Speak, DO

Lisa M. Spencer, MD

Prakrithi Srinand, MD

Sierra M. Starr, MD

Kathryne E. Staudinger, MD

Emily K. Steele, MD

Morgan R. Steffen, DO

Tricia R. Stepanek, MD

Taylor P. Stewart, MD

Kelsey A. Stewart, MD

Alyssa M. Stiff, MD

Alexandra B. Stiles, MD

Nairi K. Strauch, MD

Margaret J. Stroup, DO

Sean C. Stuart, DO

Hannah M. Stump, MD

Shalini B. Subbarao, MD

Lakshmi Subramani, MD

Heather E. Sweeney, MD

Kristin I. Swope, MD

Suha Syed, MD

Mireya P. Taboada, MD

Eneti S. Tagaloa, MD

Rachel Tang, DO

Adam R. Taylor, MD

Simone R. Thibault, MD

Kimberly A. Thill, MD

Dhanu Thiyag, MD

Andrew T. Thornton, MD

Wendy Tian, MD

Stephanie Tilberry, MD

Amanda L. Tillett, MD

Amanda M. Tjitro, MD

Logan P. Todhunter, DO

David Toffey, MD

Maris K. Toland, MD

Rachel E. Tomassi, MD

Sarah Tounsi, MD

Antonia K. Traina, MD

Taylor Tran, MD

Diem Samantha Tran, DO

Emily C. Trautner, MD

Emma Trawick, MD

Continue to: Elissa Trieu, MD...

 

 

Elissa Trieu, MD

Ariel Trilling, MD

Samantha Truong, MD

Mary M. Tsaturian, MD

Athena Tudino, MD

Kati A. Turner, MD

Nicole-Marie Tuzinkiewicz, MD

Gayathri D. Vadlamudi, MD

Stylianos Vagios, MD

Pauline V. Van Dijck, DO

Kaylee A. VanDommelen, MD

Isha B. Vasudeva, MD

Shivani J. Vasudeva, DO

Diana Q. Vazquez Parker, MD

Ridhima Vemula, MD

Elena C. Vinopal, MD

Caroline J. Violette, MD

Pascal T. Vo, DO

Michelle H. Vu, MD

Macy M. Walz, MD

Angelia Wang, MD

Eileen Wang, MD

Courtney Y. Wang, MD

Joyce Wang, MD

Meryl G. Warshafsky, MD

Sophie E.N. Weinstein, MD

Sarah H. Weinstein, MD

Annalyn M. Welp, MD

Shannon M. Wentworth, MD

Erika M. Wert, MD

Rachel C. White, MBchB

Morgan N. Wilhoite, DO

Mercedes Williams, MD

Hayley Williams, MD

Jacquelyn D. Williams, MD

Mary H. Williamson, MD

Elise Wilson, MD

Lauren M. Witchey, MD

Emily A. Wolverton, MD

Stephanie Y. Wong, MD

Jenny Wu, MD

Jackie Xiang, MD

Nancy S. Yang, MD

Kevin P. Yeagle, MD

Halina M. Yee, MD

Alyssa M. Yeung, MD

Samuel K. Yost, MD

Megan Yuen, MD

Nayab Zafar, DO

Cindy X. Zhang, DO

Yingao Zhang, MD

Helen Zhao, MD

Chelsea Zhu, MD

Billie E. Zidel, MD

Ryan A. Zoldowski, MD

Photo: Shutterstock

Six months after the Supreme Court decision that overturned the constitutional right to abortion, trainees across the United States are asking a critical question in the current resident recruitment season: How will the restrictions on abortion access affect my training as an obstetrician-gynecologist, and will they impact my ability to be the kind of provider I want to be in the future?

Among the myriad of downstream effects to patient care, the Dobbs decision will indisputably impact the scope of residency training for those that provide reproductive health services. Almost half of ObGyn residents train in states that have abortion restrictions in place.1 New educational milestones for abortion training, which are a requirement by the Accreditation Council for Graduate Medical Education (ACGME), were proposed quickly after Dobbs, guiding programs to offer opportunities for training in nonrestricted areas or the “combination of didactic activities, including simulation” to meet the training requirement in abortion care.2

Like many providers, residents already are grappling with precarious and risky circumstances, balancing patient safety and patient-driven care amidst pre-existing and newly enforced abortion restrictions. Whether managing a patient with an undesired pregnancy, severe medical comorbidities, unexpected pregnancy complications such as preterm premature rupture of membranes, or bleeding, or substantial fetal anomalies, ObGyn residents cannot gain the experience of providing the full scope of reproductive health care without the ability to offer all possible management options. While some enacted abortion restrictions have exceptions for the health of or life-saving measures for the mother, there is no standard guidance for timing of interventions, leaving providers confused and in fear of legal retribution. At a time when trainees should be learning to provide patient-centered, evidence-based care, they are instead paralyzed by the legal or professional consequences they may face for offering their best medical judgements.

Furthermore, the lack of exposure to dilation and evacuation procedures for residents in restricted practice areas will undoubtably decrease their confidence in managing acute complications, which is one of the critical facets of residency training. In a surgical field where repetition is crucial for technical competence, highlighted by ACGME minimum case requirements, the decreased volume of abortion procedures is a disadvantage for trainees and a disservice for patients. While anti-choice promoters may argue that involvement in surgical management of early pregnancy loss should suffice for ObGyn training in family planning, this piecemeal approach will leave gaps in technical skills.

The fear of legal ramifications, moral injury, and inadequate surgical training may lead to the siphoning of talented trainees to areas in the country with fewer restrictions.3Dobbs already has demonstrated how limiting abortion access will deepen inequities in reproductive health care service delivery. Approximately 55% of ObGyn trainees and nearly two-thirds of maternal-fetal medicine graduates join the workforce in the state where they received their training.4 Medical students will seek opportunities for high-quality ObGyn training in areas that will help them to be well-prepared, competent physicians—and more often than not, stay in the area or region that they trained in. This will lead to provider shortages in areas where access to reproductive health care and subspecialist providers already is limited, further exacerbating existing health disparities.

During this recruitment season, trainees and residency programs alike will need to reckon with how the ramifications of Dobbs will alter both the immediate and long-term training in comprehensive reproductive health care for the ObGyn workforce. ObGyn trainees have taken a stand in response to the Dobbs decision, and nearly 750 current residents signed onto the statement below as a commitment to high-quality training and patient-centered care. Clinical experience in performing abortions is essential to the provision of comprehensive evidence-based reproductive health care, and access to these procedures is as important for physicians-in-training as it is for patients.

Actions to take to ensure high-quality abortion training in ObGyn residencies include the following:

  • Connect with and stay involved with organizations such as the American College of Obstetricians and Gynecologists (ACOG), Physicians for Reproductive Health (PRH), and Medical Students for Choice (MSFC) for initiatives, toolkits, and resources for training at your institutions.
  • Seek specific abortion training opportunities through the Leadership Training Academy (offered through PRH) or the Abortion Training Institute (offered through MSFC).
  • Ensure that your residency program meets the ACGME criteria of providing opportunities for clinical experiences for abortion care and work with program leadership at a program, state, or regional level to enforce these competencies.
  • Reach out to your local American Civil Liberties Union or other local reproductive legal rights organizations if you want to be involved with advocacy around abortion access and training but have concerns about legal protections.
  • Have a voice at the table for empowering training opportunities by seeking leadership positions through ACOG, ACGME, Council on Resident Education in Obstetrics and Gynecology and the Association of Professors of Gynecology and Obstetrics, American Medical Association, Student National Medical Association, and subspecialty organizations.
  • Vote in every election and promote voting registration and access to your patients, colleagues, and communities. ●

Continue to: The implications of the Dobbs v Jackson Women’s Health Organization decision on the health care and wellbeing of our patients...

 

 

The implications of the Dobbs v Jackson Women’s Health Organization decision on the health care and wellbeing of our patients

On June 24, 2022, the Supreme Court of the United States ruled in a 6-3 majority decision to overturn the constitutional right to abortion protected by Roe v Wade since 1973. As health care providers, we are outraged at the Court’s disregard for an individual’s right to make reproductive decisions for themselves and their families and are deeply concerned about the devastating consequences to reproductive care and outcomes in this country for all people. Reproductive health decisions, including growing a family and whether or not to continue a pregnancy, are complex and incredibly personal. Our role as health care providers is to help guide those decisions with empathy and evidencebased clinical recommendations. This ruling undermines a patient’s right to bodily autonomy, free of impositions from government and political pressures, and it threatens the sanctity of complex medical decision-making between a patient, their family, and their medical team.

As medical professionals, we know that every patient’s situation is unique—banning abortion procedures ties the hands of physicians trying to provide the most medically appropriate options in a compassionate manner. We know that both medical and surgical abortions are safe and can save lives. These procedures can help patients with potentially life-threatening conditions worsened by pregnancy, a poor prognosis for the fetus, or a complication from the pregnancy itself. Physicians use scientific research and individualized approaches to help patients in unique situations, and attempts to legislate personal health decisions compromise the practice of evidence-based medicine.

We also know that this decision will impact some communities more than others. Access to safe abortion care will become dependent on which region of the country a person lives in and whether or not a person has resources to seek this care. Due to continued systemic racism and oppression, patients of color will be disproportionately impacted and likely will suffer worse health outcomes from unsafe abortions. Those that rely on public insurance or who are uninsured will face overwhelming barriers in seeking abortion services. These disparities in reproductive care, which contribute to our nation’s health crises in maternal morbidity and mortality, unintended pregnancy, and neonatal complications, will further entrench health inequities, and patient lives and livelihoods will suffer.

We acknowledge the impact that this decision will have on restricting access to reproductive care. We stand by the fact that abortion care is health care. We vow to uphold the tenets of our profession to place patient autonomy and provision of safe quality medical care at the forefront of our practices.

We, as health care providers and physician trainees, hereby pledge:

  • To continue to provide evidence-based, nonjudgmental counseling for all pregnancy options, including abortion, and support our patients through all reproductive health decisions
  • To promote equity in providing comprehensive reproductive health care, recognizing the impacts of systemic racism and oppression
  • To promote high quality training in providing safe reproductive care in our respective institutions
  • To use our voices in our communities to advocate for all our patients to have the freedom to access the safe and compassionate health care they deserve.

Sincerely,

The undersigned 747 ObGyn resident physicians

Please note that we sign this statement on our own behalf as individuals and not on behalf of our respective institutions.

Orchideh Abar, MD

Laurel S. Aberle, MD

Kathleen E. Ackert, DO

Lauryn Adams, MD

Temiloluwa Adejuyigbe, MD

Oluwatoyosi M. Adeoye, MD

Hufriya Y. Aderianwalla, MD

Fareeza Afzal, MD

Adelaide Agyepong, MD

Erin R. Ahart, MD

Noha T. Ahmed, DO

Faria Ahmed, MD

Tracey O. Akanbi, MD

Eloho E. Akpovi, MD

Austin H. Allen, DO

Amanda M. Allen, MD

Alexis L. Allihien, MD

Jorge L. Alsina, MD

Paulina C. Altshuler, DO

Sivani Aluru, MD

Amal Amir, DO

Jon Anderson, DO

Andreas Antono, MD

Annie N. Apple, MD

Janine Appleton, DO

Aarthi Arab, MD

Sydney R. Archer, MD

Youngeun C. Armbuster, MD

Kara Arnold, MD

Blessing C. Aroh, MD

Savannah Pearson Ayala, MD

Archana K. Ayyar, MD

Ann-Sophie Van Backle, DO

Connor R. Baker, MD

Japjot K. Bal, MD

Abigail E. Barger, MD

Kathryn E. Barron, MD

Silvia Bastea, MD

Samantha V.H. Bayer, MD

Kristen Beierwaltes, MD

Gisel Bello, MD

Michelle A. Benassai, MD

Dana Benyas, MD

Alice F. Berenson, MD

Hanna P. Berlin, MD

Abigail L. Bernard, MD

Eli H. Bernstein, MD

Julia T. Berry, MD

Bryce L. Beyer, MD

Caroline Bilbe, MD

Grace E. Binter, DO

Erin E. Bishop, MD

Sierra G. Bishop, MD

Stephanie S. Bista, MD

Tara E. Bjorklund, DO

Alyssa N. Black, MD

Continue to: Kelsey Boghean, DO...

 

 

Kelsey Boghean, DO

Areta Bojko, MD

Grace E. Bommarito, DO

Aditi R. Bommireddy, MD

Genna C. Bonfiglio, MD

Mary E. Booker, MD

Kayce L. Booth, MD

Samantha T. Boothe, DO

William Borenzweig, MD

Rebecca M. Borneman, MD

Alexander L. Boscia, MD

Gina M. Botsko, MD

Glenn P. Boyles, MD

Avery C. Bramnik, MD

Sophia N. Brancazio, MD

Katarina M. Braun, MD

Anthony Brausch, MD

Emily L. Brekke, MD

Sara E. Brenner, MD

Bailey A. Brown, DO

Kathryn S. Brown, MD

Denese C. Brown, MD

Abena Bruce, MD

Sabrina C. Brunozzi, MD

Madison Buchman, DO

Deirdre G. Buckley, MD

Rachel L. Budker, MD

Leeann M. Bui, MD

Anthony H. Bui, MD

Jessie Bujouves, MD

Kimberley A. Bullard, MD

Sophia G. Bunde, MD

Emily R. Burdette, MD

Iris Burgard, DO

Korbi M. Burkey, MD

Lindsey K. Burleson, MD

Lindsay M. Burton, MD

Brianna N. Byers, MD

Stephanie Cai, MD

Alexandra S. Calderon, MD

Alexandra G. Caldwell, MD

Natalia Calzada, MD

Tamara Cameo, MD

Arielle Caplin, MD

Angela M. Carracino, DO

Anna L. Carroll, MD

Leigha M. Carryl, MD

Ashlie S. Carter, MD

Stephanie Casey, DO

Chase W. Cataline, DO

Carson L. Catasus, MD

Alena R. Cave, MD

Kelly M. Chacon, MD

Avis L. Chan, MD

Shruthi Chandra, MD

Jennifer Chang, MD

Shannon Chang, DO

Gillian Chase, MD

Cindy Chen, MD

Jessie C. Chen, MD

Jessica T. Chen, MD

Wenjin Cheng, MB

Laura J. Cheng, MD

Lucy Cheng, MD

Monica S. Choo, MD

Jody S. Chou, MD

Hannah C. Christopher, DO

Continue to: David J. Chromey, DO...

 

 

David J. Chromey, DO

Grace V. Clark, MD

Celeste Colegrove, MD

Sarah C. Combs, MD

Victoria L. Conniff, MD

Hannah C. Connor, MD

Angela J. Conway, MD

Steffany A. Conyers, MD

Alexandra Cooke, MD

Ashley A. Cooney, MD

Anna Cornelius-Schecter, MD

Alexa M. Corso, DO

Krysten A. Costley, MD

Madeline Coulter, MD

Kelsey Cramer, MD

Anna E. Cronin, MD

Bethany N. Croyle, DO

Carmen A. Cueto, MD

Nicole Cumbo, MD

Mackenzie A. Cummings, MD

Carrie Cummiskey, MD

Hannah M. Cunningham, MD

Sarah D’Souza, DO

Rachael M. D’Auria, MD

Caitlin Dane, MD

Rachel N. Dang, MD

Talin R. Darian, MD

Abigail C. Davies, MD

Berkley Davis, MD

Lois A. Davis, MD

Jennie J. DeBlanc, MD

Ayana G.R. DeGaia, MD, MPH

Katerina N. DeHaan, MD

Rebekka M. Delgado, MD

Brettany C. DeMier, MD

Bonnie W. DePaso, MD

Hemaxi H. Desai, DO

Amberly T. Diep, MD

Abigail K. Dillaha, MD

Sarah K. Dominguez, MD

Abbey P. Donahue, MD

Allan C. Dong, MD

James Doss, MD

Taylor B. Douglas, MD

Abigail G. Downey, MD

Janelle M. Driscoll, MD

Emily Du, MD

Leslie V. Dunmire, MD

Jennifer Duong, DO

Leigh C. Durudogan, MD

Mai N. Dyer, MD, MPH

Rebecca A. Ebbott, MD

Lindsey P. Eck, MD

Molly C. Eckman, MD

Alex Ede, MD, ScM

Claire E. Edelman, MD

Sara E. Edwards, MD

David J. Eggert, DO

Michelle Eide, MD

Etoroabasi Ekpe, MD

Tressa L. Ellett, MD

Laura Peyton Ellis, MD

Kaitlin H. Ellis, MD

Mariah G. Elly, MD

Jennifer Embry, MD

Claire Englert, MD

Brenna Espelien, MD

Kamilah Evans, MD

Joshua A. Ewy, MD

Elana D. Fackler, MD

Lauren E. Falk, MD

Brianna A. Farley, MD

Amanda Stephanie R. Farrell, MD

Sara Fassio, DO

Daniela A. Febres-Cordero, MD

Jasmin E. Feliciano, MD

Alayna H. Feng, MD

Amanda M. Ferraro, MD

Brittany A. Fickau, MD

Brittany H. File, MD

Shannon M. Finner, DO

Mia E. Fischbein, DO

Briah Fischer, MD

Shira Fishbach, MD

Alison C. Fitzgerald, MD

Evan R. Fitzgerald, MD

Margaret R. Flanigan, MD

Kevin C. Flatley, MD

Jordan A. Fletcher, MD

Claudia E. Flores, MD

Lauren A. Forbes, MD

Rana K. Fowlkes, MD

Jennifer M. Franks, MD, MPH

Christina M. Frasik, MD

Haven N. Frazier, DO

Sarah W. Freeman, MD

Emilie O. Fromm, DO

Anna R. Fuchss, MD

Emma K. Gaboury, MD

Madeline H. Ganz, MD

Lex J. Gardner, MD

Keri-Lee Garel, MD

Hailey B. Gaskamp, DO

Brittney A. Gaudet, MD

Gabrielle M. Gear, MD

Eleanor R. Germano, MD

Lauren G. Gernon, MD

Allen Ghareeb, MD

Patricia Giglio Ayers, MD

Jordana L. Gilman, MD

Mianna M. Gilmore, DO

Brian W. Goddard, MD

Julia L. Goldberg, MD

M. Isabel Gonzaga, MD

Fred P. Gonzales, MD

Lillian H. Goodman, MD, MPH

Ashley Goreshnik, MD

Lauren E. Gottshall, MD

Lindsay L. Gould, MD

Kelsea R. Grant, MD

Dorender A. Gray, MD

Sophie Green, MD

Erica A. Green, MD

Danielle C. Greenberg, MD

Kalin J. Gregory-Davis, MD

David M. Greiner, MD

Tyler M. Gresham, MD

Continue to: Nelly Grigorian, MD...

 

 

Nelly Grigorian, MD

Erin L. Grimes, MD

Whitney Grither, MD

Jared M. Grootwassink, MD

Maya E. Gross, MD

Paoula Gueorguieva, MD

Margot M. Gurganus, DO

Rachel L. Gutfreund, MD

Andres Gutierrez, MD

Dorothy L. Hakimian, DO

Ashley N. Hamati, DO

Marie M. Hanna-Wagner, MD

Katie Hansen, MD

Courtney Hargreaves, MD

Stephanie Harlow, MD

Kelsey B. Harper, MD

Devon A. Harris, MD

Lauren E. Harris, MD

Emily S. Hart, DO

Sarah A. Hartley, MD

Becky K. Hartman, MD

Abigail K. Hartmann, MD

Charlotte V. Hastings, MD

Cherise Hatch, DO

Jordan Hauck, DO

Sarena Hayer, MD

Jenna M. Heath, MD

Eric D. Helm, MD

Julie A. Hemphill, MD

Ric A.S. Henderson, MD

Nicola A. Hendricks, MD

Andrea A. Henricks, MD

Jesse M. Herman, DO

Alyssa M. Hernandez, DO

Melissa Hernandez, MD

Alyssa R. Hersh, MD

Alexandra Herweck, MD

Brianna Hickey, MD

Allix M. Hillebrand, MD

Alessandra I. Hirsch, MD

Emily A. Hoffberg, MD

Chloe L. Holmes, DO

Cameron M. Holmes, MD

Helena Y. Hong, MD

Wakako Horiuchi, MD

Shweta Hosakoppal, MD

Jaycee E. Housh, MD

Shannon M. Howard, MD

Meredith C. Huszagh, MD

Yihharn P. Hwang, MD

Emma C. Hyde, MD

Brooke Hyman, MD

Hala Ali Ibrahim, MD

Gnendy Indig, MD

Erin E. Isaacson, MD

Shruti S. Iyer, DO

Audrey J. Jaeger, DO

Shobha Jagannatham, MD

Cyrus M. Jalai, MD

Emma V. James, MD

Isabel Janmey, MD

Phoebe Jen, DO

Corey L. Johnson, MD

Crystal J. Johnson, MD

Andrea M. Johnson, MD

Nat C. Jones, MD

Briana L. Jones, DO

Rebecca J. Josephson, MD

Sarah Natasha Jost-Haynes, MD

 

Continue to: Hannah S. Juhel, MD...

 

 

Hannah S. Juhel, MD

Erin Jun, DO

Katherine B. Kaak, MD

Dhara N. Kadakia, MD

Amanda D. Kadesh, MD

Riana K. Kahlon, MD

Nadi N. Kaonga, MD

Moli Karsalia, MD

Stephanie L. Kass, MD

Amanda M. Katz, MD

Chelsea S. Katz, MD

Virginia Kaufman, MD

Gurpinder Kaur, MD

Jessica A. Keesee, MD

Cassandra N. Kelly, MD

Whitney Kelly, DO

Hannah V. Kennedy, MD

Bethany H. Kette, MD

Iman Khan, MD

Maryam M. Khan, MD

Alisa Jion Kim, MD

Tesia G. Kim, MD

Anne E. Kim, MD

Emily H. King, MD

Tarynne E. Kinghorn, MD

Holly T. Kiper, DO

Thomas Kishkovich, MD

Quinn M. Kistenfeger, MD

Sofia E. Klar, DO

Jessica B. Klugman, MD

Hope E. Knochenhauer, MD

Kathleen J. Koenigs, MD

Olga Kontarovich, DO

Alison Kosmacki, MD

Ana E. Kouri, MD

Olga M. Kovalenko, MD

Leigh T. Kowalski, MD

Kayla A. Krajick, MD

Elizabeth S. Kravitz, MD

Shruti Rani Kumar, MD

Alyssa Kurtz, DO

Lauren H. Kus, MD

Arkadiy Kusayev, DO

Amanda E. Lacue, MD

Nava Lalehzari, MD

Amber Lalla, MD

Allie C. Lamari, DO

Kelly L. Lamiman, MD

Stephen Lammers, MD

Monet Lane, MD

Madeline L. Lang, MD

Liana Langdon-Embry, MD

Carolyn Larkins, MD

Leah E. Larson, MD

Matthew W. Lee, MD

Eunjae Lee, MD

Alice Lee, MD

Jared Z. Lee, MD

Charlotte M. Lee, MD

Nicole R. Legro, MD

Aurora Leibold, MD

Rosiris Leon-Rivera, MD, PhD

Anna M. Leone, MD

Keiko M. Leong, MD

Lindsey M. LePoidevin, MD

Molly E. Levine, MD

Khrystyna Levytska, MD

Dana L. Lewis, DO

Jessica L. Li, MD

Kristina Lilja, MD

Deanna M. Lines, DO

Annalise Littman, MD

Julia F. Liu, MD

Tyler B. Lloyd, MD

Alyssa Lo, MD

K’ara A. Locke, MD

Minica Long, MD

Melissa Lopez, MD

Wilfredo A. Lopez, MD

Connie F. Lu, MD

Tyler J. Lueck, MD

Katherine L. Lukas, MD

Davlyn L. Luke, MD

Shani Ma, MD

Colton Mabis, MD

Lauren T. MacNeill, MD

Rachel Madding, MD

Mona Makhamreh, MD

Francesca R. Mancuso, MD

Kelsey L. Manfredi, MD

Valeria Mantilla, MD

Kaitlin M. Mar, MD

Starcher R. Margaret, MD

Audrey M. Marinelli, MD

Brittany A. Marinelli, MD

Emily S. Markovic, MD

Hannah L. Marshall, MD

Aaron Masjedi, MD

Isabelle M. Mason, MD

Akailah T. Mason-Otey, MD

Nicole Massad, MD

Megan M. Masten, MD

Stephanie M. Masters, MD

Anastasia Matthews, MD

Natalia del Mazo, MD

Sara A. McAllaster, MD

Continue to: Nicole McAndrew, DO...

 

 

Nicole McAndrew, DO

Madeline G. McCosker, MD

Jamie L. McDowell, DO

Christine E. McGough, MD

Mackenzi R. McHugh, MD

Madeline M. McIntire, MD

Cynthia R. McKinney, MD

Kirsten D. McLane, MD

Shian F. McLeish, MD

Megan I. McNitt, MD

Sarah R. McShane, MD

Grace R. Meade, MD

Nikki Ann R. Medina, DO

Tiffany L. Mei, MD

Jenna Meiman, MD

Anna M. Melicher, MD

Rosa M. Mendez, MD

Riley Mickelsen, MD

Sage A. Mikami, MD

Aletheia B. Millien, MD

Hannah C. Milthorpe, MD

Caroline J. Min, MD

Julie A. Mina, MD

Annie G. Minns, MD

Natalie Mironov, DO

Elizabeth L. Mirsky, MD

Astha Mittal, MD

Rachel E. Mnuk, MD

Silki Modi, MD

Sudarshan J. Mohan, MD

Roxana Mohhebali-Solis, MD

Mugdha V. Mokashi, MD

Jessica A. Montgomery, MD

Ellen Moore, MD

Savannah J. Morehouse, MD

Kristen L. Moriarty, MD

Alexa P. Morrison, MD

Bijan Morshedi, MD

Matthew H. Mossayebi, MD

Kathy Mostajeran, DO

Sharan Mullen, DO

Ellen C. Murphy, MD

Emma Chew Murphy, MD

Lauren M. Murphy, MD

Bria Murray, MD

Erin C. Nacev, MD

Preetha Nandi, MD

Blaire E. Nasstrom, DO

Hallie N. Nelson, MD

Katherine A. Nelson, MD

Margaret S. Nemetz, MD

Daniela Ben Neriah, DO

Cosima M. Neumann, MD

Mollie H. Newbern, DO

Gisella M. Newbery, MD

Stephanie Nguyen, MD

Christine G.T. Nguyen, MD

Desiree Nguyen, MD

Jacqueline W. Nichols, MD

Annika M. Nilsen, MD

Margaret A. Nixon, MD

Emily M. Norkett, MD

Allison N. Nostrant, DO

Susan E. Nourse, MD

Aliya S. Nurani, MD

Emily E. Nuss, MD

Jeanne O. Nwagwu, DO

Kelsey E. O’Hagan, MD

Margaret O’Neill, MD

Emily A. O’Brien, MD

Carly M. O’Connor-Terry, MD, MS

Madison O. Odom, MD

Cynthia I. Okot-Kotber, MD

Sarah P. Oliver, MD

Leanne P. Ondreicka, MD

Ngozika G. Onyiuke, MD

Erika Gonzalez Osorio, MD

Marika L. Osterbur Badhey, MD

Linda A. Otieno, MD

Claire H. Packer, MD

Chloe W. Page, DO

Marissa Palmor, MD

Rishitha Panditi, MD

Katherine A. Panushka, MD

Kelsey J. Pape, MD

Rachel R. Paquette, DO

Hillary C. Park, DO

Kendall M. Parrott, MD

Ekta Partani, MD

Karishma Patel, MD

Shivani Patel, MD

Continue to: Priya Patel, MD...

 

 

Priya Patel, MD

Jenna M. Patterson, MD

Ashleigh Pavlovic, MD

Katie M. Peagler, MD

Katherine T. Pellino, MD

Nicholas Per, MD

Elana Perry, MD

Emily J. Peters, MD

Sara E. Peterson, MD

Michelle R. Petrich, MD

Destiny L. Phillips, MD

Chloe Phillips, MD

Megan E. Piacquadio, DO

Sara C. Pierpoint, MD

Celeste M. Pilato, MD

Emma Pindra, MD

Minerva L.R. Pineda, MD

Rebecca Pisan, MD

Alessandra R. Piscina, MD

Rachael Piver, MD

Andrew J. Polio, MD

Hector S. Porragas, MD

Natalie Posever, MD

Allison R. Powell, MD

Mahima V. Prasad, MD

Angelina D. Prat, DO

Rebecca L. Purvis, MD

Teresa L. Qi, MD

Nicholas R. Quam, MD

Candice A. Quarella, MD

Nicholas W. Racchi, DO

Jeannie G. Radoc, MD

Samuel Raine, MD

Anna C. Raines, MD

Stephanie A. Rains, MD

Nicole M. Rainville, DO

Karissa Rajagopal, DO

Kristian R. Ramage, MD

Praveen Ramesh, MD

Tia M. Ramirez, MD

Jania Ramos, MD

Neel K. Rana, MD

Urvi Rana, DO

Indira Ranaweera, MD

Sindhuja Ranganathan, DO

Chloe R. Rasmussen, MD

Laura P. Reguero-Cadilla, MD

Devin M. Reilly, MD

Kimberly E. Reimold, MD

Cory R. Reiter, MD, PhD

Maya E. Reuven, DO

Jessica Reyes-Peterson, MD

Jacqueline Rice, MD

Rebecca L. Richardson, MD

Mikaela J. Rico, DO

Katelyn Rittenhouse, MD

Giuliana A. Rivera Casul, MD

Jill N.T. Roberts, MD

Luke N. Roberts, MD

Esther Robin, MD

Marcella Israel Rocha, MD

Zoe A. Roecker, MD

Hilary E. Rogers, MD

Kelsey A. Roof, MD

Zarah Rosen, MD

Cecilia M. Rossi, MD

Eva S. Rostonics, MD

Felix Rubio, MD

Amela Rugova, MD

Anna J. Rujan, MD

Erika T. Russ, MD

Colin Russell, MD

Ruby L. Russell, MD

Isabella A. Sabatina, MD

Gouri Sadananda, MD

Aashna Saini, MD

Salomeh M. Salari, MD

Ndeye N. Sall, MD

Nicole M. Salvador, MD

Aayushi Sardana, MD

Kendall M. Sarson, MD

Rita Abigail Sartor, MD

Continue to: Haley A. Scarbrough, MD...

 

 

Haley A. Scarbrough, MD

Kimberly Schaefer, MD

Demetra Schermerhorn, MD

Ellen C. Schleckman, MD

Maura A. Schlussel, MD

Ellie Schmidt, MD

Alison M. Schmidt, MD

Evan A. Schrader, MD

Morgan A. Schriever, MD

Brianna L. Schumaker Nguyen, DO

Whitney E. Scott, MD

Claire Scrivani, MD

Catherine E. Seaman, MD

Rachel D. Seaman, MD

Danielle J. Seltzer, MD

Joshua R. Shaffer, MD

Emily A. Shaffer, MD

Delia S. Shash, MD

Ishana P. Shetty, MD

Tushar Shetty, MD

Carol Shi, MD

Sarah P. Shim, MD

Emma C. Siewert, MD

Seth M. Sigler, DO

Rebecca L. SigourneyTennyck, MD

Daniella D. Silvino, DO

Andrea M. Simi, MD

Amelia R. Simmons, MD

Amy E. Skeels, DO

Ashley E.S. Keith, MD

Hannah C. Smerker, DO

Katarina Smigoc, MD

Madeline I. Smith, MD

Jessica D. Smith, MD

Melanie R. Smith, MD

Alicia L. Smith, MD

Chloe Smith, MD

Ayanna Smith, MD

Melanie R. Smith, MD

Megan M. Smith, MD

Haverly J. Snyder, MD

Beatrice R. Soderholm, DO

Brianna C. Sohl, MD

Samantha A. Solaru, MD

Michael Solotke, MD

Dara A.H. Som, MD

Alexandra R. Sotiros-Lowry, MD

Melanie Spall, DO

Alicia C. Speak, DO

Lisa M. Spencer, MD

Prakrithi Srinand, MD

Sierra M. Starr, MD

Kathryne E. Staudinger, MD

Emily K. Steele, MD

Morgan R. Steffen, DO

Tricia R. Stepanek, MD

Taylor P. Stewart, MD

Kelsey A. Stewart, MD

Alyssa M. Stiff, MD

Alexandra B. Stiles, MD

Nairi K. Strauch, MD

Margaret J. Stroup, DO

Sean C. Stuart, DO

Hannah M. Stump, MD

Shalini B. Subbarao, MD

Lakshmi Subramani, MD

Heather E. Sweeney, MD

Kristin I. Swope, MD

Suha Syed, MD

Mireya P. Taboada, MD

Eneti S. Tagaloa, MD

Rachel Tang, DO

Adam R. Taylor, MD

Simone R. Thibault, MD

Kimberly A. Thill, MD

Dhanu Thiyag, MD

Andrew T. Thornton, MD

Wendy Tian, MD

Stephanie Tilberry, MD

Amanda L. Tillett, MD

Amanda M. Tjitro, MD

Logan P. Todhunter, DO

David Toffey, MD

Maris K. Toland, MD

Rachel E. Tomassi, MD

Sarah Tounsi, MD

Antonia K. Traina, MD

Taylor Tran, MD

Diem Samantha Tran, DO

Emily C. Trautner, MD

Emma Trawick, MD

Continue to: Elissa Trieu, MD...

 

 

Elissa Trieu, MD

Ariel Trilling, MD

Samantha Truong, MD

Mary M. Tsaturian, MD

Athena Tudino, MD

Kati A. Turner, MD

Nicole-Marie Tuzinkiewicz, MD

Gayathri D. Vadlamudi, MD

Stylianos Vagios, MD

Pauline V. Van Dijck, DO

Kaylee A. VanDommelen, MD

Isha B. Vasudeva, MD

Shivani J. Vasudeva, DO

Diana Q. Vazquez Parker, MD

Ridhima Vemula, MD

Elena C. Vinopal, MD

Caroline J. Violette, MD

Pascal T. Vo, DO

Michelle H. Vu, MD

Macy M. Walz, MD

Angelia Wang, MD

Eileen Wang, MD

Courtney Y. Wang, MD

Joyce Wang, MD

Meryl G. Warshafsky, MD

Sophie E.N. Weinstein, MD

Sarah H. Weinstein, MD

Annalyn M. Welp, MD

Shannon M. Wentworth, MD

Erika M. Wert, MD

Rachel C. White, MBchB

Morgan N. Wilhoite, DO

Mercedes Williams, MD

Hayley Williams, MD

Jacquelyn D. Williams, MD

Mary H. Williamson, MD

Elise Wilson, MD

Lauren M. Witchey, MD

Emily A. Wolverton, MD

Stephanie Y. Wong, MD

Jenny Wu, MD

Jackie Xiang, MD

Nancy S. Yang, MD

Kevin P. Yeagle, MD

Halina M. Yee, MD

Alyssa M. Yeung, MD

Samuel K. Yost, MD

Megan Yuen, MD

Nayab Zafar, DO

Cindy X. Zhang, DO

Yingao Zhang, MD

Helen Zhao, MD

Chelsea Zhu, MD

Billie E. Zidel, MD

Ryan A. Zoldowski, MD

References

 

  1. Vinekar K, Karlapudi A, Nathan L, et al. Projected implications of overturning Roe v Wade on abortion training in US obstetrics and gynecology residency programs. Obstet Gynecol. 2022;140:146-149.
  2. ACGME program requirements for graduate medical education in obstetrics and gynecology summary and impact of interim requirement revisions. ACGME website. Accessed December 18, 2022. https://www.acgme.org/globalassets/pfassets/reviewandcomment/220_obstetricsandgynecology_2022-06-24_impact.pdf
  3. Crear-Perry J, Hassan A, Daniel S. Advancing birth equity in a post-Dobbs US. JAMA. 2022;328:1689-1690.
  4. Report on residents. AAMC website. Accessed December 18, 2022. https://www.aamc.org/data-reports/students-residents/interactive-data/report-residents/2021/table-c4-physician-reten tion-state-residency-training-last-completed-gme
References

 

  1. Vinekar K, Karlapudi A, Nathan L, et al. Projected implications of overturning Roe v Wade on abortion training in US obstetrics and gynecology residency programs. Obstet Gynecol. 2022;140:146-149.
  2. ACGME program requirements for graduate medical education in obstetrics and gynecology summary and impact of interim requirement revisions. ACGME website. Accessed December 18, 2022. https://www.acgme.org/globalassets/pfassets/reviewandcomment/220_obstetricsandgynecology_2022-06-24_impact.pdf
  3. Crear-Perry J, Hassan A, Daniel S. Advancing birth equity in a post-Dobbs US. JAMA. 2022;328:1689-1690.
  4. Report on residents. AAMC website. Accessed December 18, 2022. https://www.aamc.org/data-reports/students-residents/interactive-data/report-residents/2021/table-c4-physician-reten tion-state-residency-training-last-completed-gme
Issue
OBG Management - 35(1)
Issue
OBG Management - 35(1)
Page Number
45-46, e47-e51
Page Number
45-46, e47-e51
Publications
Publications
Topics
Article Type
Display Headline
How the Dobbs decision shapes the ObGyn workforce and training landscape
Display Headline
How the Dobbs decision shapes the ObGyn workforce and training landscape
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Eyebrow Default
PERSPECTIVES FROM THE FIELD
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Have investigators reached the first steps for redefining a diagnostic definition of preeclampsia that includes morbidity?

Article Type
Changed
Mon, 01/23/2023 - 20:43

 

 

Thadhani R, Lemoine E, Rana S, et al. Circulating angiogenic factor levels in hypertensive disorders of pregnancy. N Engl J Med. 2022;1. DOI: 10.1056/EVIDoa2200161

EXPERT COMMENTARY
 

The standard core lecture on preeclampsia given to all medical students frequently begins with an epic, if not potentially apocryphal, statement regarding how this disease has been noted in the annals of medical history since the time of the Ancients. Although contemporary diagnostic criteria for preeclampsia are not that far out of date, they are close. The increased urinary protein loss and hypertension preceding eclamptic seizures was first noted at the end of the 19th century. The blood pressure and proteinuria criteria used for diagnosis was codified in its contemporary form in the late 1940s. Since then, “tweak” rather than “overhaul” probably best describes the updates of the obstetrical community to the definition of preeclampsia. This has just changed.

 

Details of the study

Thadhani and colleagues prospectively recruited a nationally representative observational cohort of patients hospitalized for hypertension during pregnancy, then followed the patients until either the diagnosis of preeclampsia with severe features or for 2 weeks, whichever came first. At enrollment, circulating levels of the soluble fms-like tyrosine kinase 1 (sFlt-1) and placental growth factor (PlGF) were measured. In a 2-phased design, the first 219 participants were used to define a sFlt-1/PlGF ratio that would predict progression to severe preeclampsia within 2 weeks. The next 556 enrollees served to validate the predictive properties of the ratio. The authors found that a sFlt-1/PlGF ratio of ≥40 predicted progression to preeclampsia with severe features with an area under the curve (AUC) of 0.92.

As products of the trophoblasts, both sFlt-1 and PlGF have been mooted for almost 2 decades as potential predictive, if not diagnostic, aids with respect to preeclampsia. Indeed, both analytes are commercially available in Europe for specifically this purpose and many maternal-fetal medicine practitioners working in the European equivalent American tertiary referral centers use an sFlt-1/PlGF ratio as their primary criteria for a diagnosis of preeclampsia. Within the United States, there was an initial flurry of interest in and an infusion of corporate and federal research support for sFlt-1 and PlGF as diagnostic aids for preeclampsia in the mid-2000s. However, at present, the US Food and Drug Administration (FDA) has not sanctioned these (or any) biomarkers to aid in the diagnosis of preeclampsia. As Thermo-Fisher Scientific (Waltham, Massachusetts) is a supporting partner in this study, it is almost certain that these data will be submitted for review by the FDA as part of an application for a preeclampsia diagnostic. At some point in the near future, American practitioners will potentially be able to join their European colleagues in utilizing these biomarkers in the diagnosis of preeclampsia with severe features. ●

WHAT THIS EVIDENCE MEANS FOR PRACTICE

Thadhani and colleagues observed that the majority of both maternal and neonatal morbidity in their study, including 8 of the 9 neonatal deaths and both cases of eclampsia, occurred among patients with a ratio ≥40 at admission. There was an inverse relation between the sFlt-1/PlGF ratio and the admission to delivery interval. Where only 17% of patients in the highest quartile of ratios remained pregnant at 14 days post-enrollment, more than 79% of the lowest quartile were still pregnant. If not a causal relationship, sFlt-1 and PlGF are clearly associated with not only the occurrence of preeclampsia with severe features but also the degree of morbidity.

The implication for the disposition of patient care resources is clear. Patients at higher risk for preeclampsia could be seen in specialty high-risk clinics with an emphasis on increased monitoring. In situations where tertiary care is more remote, plans could be developed should they need to be transported to centers able to provide the appropriate level of care. Conversely, patients screening at lower ratios may be more appropriately managed as outpatients, or at least in less clinically involved accommodations.

Thadhani et al do note that there were false negative cases in which the sFlt-1/PlGF ratio at admission was <40 but patients nonetheless progressed to preeclampsia with severe features. The majority of these cases had concurrent pre-pregnancy, chronic hypertension. This observation suggests not only the potential for insights into the pathophysiology of the hypertensive diseases in pregnancy but also that the interpretation of the sFlt/PlGF ratio may eventually need to be stratified by preexisting conditions.

The final implications for the observations of this study are perhaps the most tantalizing. If there is a causal relation between the level of the sFlt-1/PlGF ratio and the morbidity of preeclampsia with severe features, then lowering the circulating concentration of sFlt-1 would ameliorate not only the morbidity but also the risk of preeclampsia. Work with plasma phoresies has suggested that this might be possible, albeit via a clinical intervention demanding more intensive resources. The potential for a targeted pharmacologic moderation of sFlt-1 levels would hold great promise in that those identified as at increased risk could be offered an intervention widely available to all.

Article PDF
Author and Disclosure Information

Thomas F. McElrath, MD, PhD, is Attending in Maternal-Fetal Medicine, Brigham & Women’s Hospital; Professor of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School; and Professor of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.

The author reports no financial relationships relevant to this article.

Issue
OBG Management - 35(1)
Publications
Topics
Page Number
12-13
Sections
Author and Disclosure Information

Thomas F. McElrath, MD, PhD, is Attending in Maternal-Fetal Medicine, Brigham & Women’s Hospital; Professor of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School; and Professor of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.

The author reports no financial relationships relevant to this article.

Author and Disclosure Information

Thomas F. McElrath, MD, PhD, is Attending in Maternal-Fetal Medicine, Brigham & Women’s Hospital; Professor of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School; and Professor of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.

The author reports no financial relationships relevant to this article.

Article PDF
Article PDF

 

 

Thadhani R, Lemoine E, Rana S, et al. Circulating angiogenic factor levels in hypertensive disorders of pregnancy. N Engl J Med. 2022;1. DOI: 10.1056/EVIDoa2200161

EXPERT COMMENTARY
 

The standard core lecture on preeclampsia given to all medical students frequently begins with an epic, if not potentially apocryphal, statement regarding how this disease has been noted in the annals of medical history since the time of the Ancients. Although contemporary diagnostic criteria for preeclampsia are not that far out of date, they are close. The increased urinary protein loss and hypertension preceding eclamptic seizures was first noted at the end of the 19th century. The blood pressure and proteinuria criteria used for diagnosis was codified in its contemporary form in the late 1940s. Since then, “tweak” rather than “overhaul” probably best describes the updates of the obstetrical community to the definition of preeclampsia. This has just changed.

 

Details of the study

Thadhani and colleagues prospectively recruited a nationally representative observational cohort of patients hospitalized for hypertension during pregnancy, then followed the patients until either the diagnosis of preeclampsia with severe features or for 2 weeks, whichever came first. At enrollment, circulating levels of the soluble fms-like tyrosine kinase 1 (sFlt-1) and placental growth factor (PlGF) were measured. In a 2-phased design, the first 219 participants were used to define a sFlt-1/PlGF ratio that would predict progression to severe preeclampsia within 2 weeks. The next 556 enrollees served to validate the predictive properties of the ratio. The authors found that a sFlt-1/PlGF ratio of ≥40 predicted progression to preeclampsia with severe features with an area under the curve (AUC) of 0.92.

As products of the trophoblasts, both sFlt-1 and PlGF have been mooted for almost 2 decades as potential predictive, if not diagnostic, aids with respect to preeclampsia. Indeed, both analytes are commercially available in Europe for specifically this purpose and many maternal-fetal medicine practitioners working in the European equivalent American tertiary referral centers use an sFlt-1/PlGF ratio as their primary criteria for a diagnosis of preeclampsia. Within the United States, there was an initial flurry of interest in and an infusion of corporate and federal research support for sFlt-1 and PlGF as diagnostic aids for preeclampsia in the mid-2000s. However, at present, the US Food and Drug Administration (FDA) has not sanctioned these (or any) biomarkers to aid in the diagnosis of preeclampsia. As Thermo-Fisher Scientific (Waltham, Massachusetts) is a supporting partner in this study, it is almost certain that these data will be submitted for review by the FDA as part of an application for a preeclampsia diagnostic. At some point in the near future, American practitioners will potentially be able to join their European colleagues in utilizing these biomarkers in the diagnosis of preeclampsia with severe features. ●

WHAT THIS EVIDENCE MEANS FOR PRACTICE

Thadhani and colleagues observed that the majority of both maternal and neonatal morbidity in their study, including 8 of the 9 neonatal deaths and both cases of eclampsia, occurred among patients with a ratio ≥40 at admission. There was an inverse relation between the sFlt-1/PlGF ratio and the admission to delivery interval. Where only 17% of patients in the highest quartile of ratios remained pregnant at 14 days post-enrollment, more than 79% of the lowest quartile were still pregnant. If not a causal relationship, sFlt-1 and PlGF are clearly associated with not only the occurrence of preeclampsia with severe features but also the degree of morbidity.

The implication for the disposition of patient care resources is clear. Patients at higher risk for preeclampsia could be seen in specialty high-risk clinics with an emphasis on increased monitoring. In situations where tertiary care is more remote, plans could be developed should they need to be transported to centers able to provide the appropriate level of care. Conversely, patients screening at lower ratios may be more appropriately managed as outpatients, or at least in less clinically involved accommodations.

Thadhani et al do note that there were false negative cases in which the sFlt-1/PlGF ratio at admission was <40 but patients nonetheless progressed to preeclampsia with severe features. The majority of these cases had concurrent pre-pregnancy, chronic hypertension. This observation suggests not only the potential for insights into the pathophysiology of the hypertensive diseases in pregnancy but also that the interpretation of the sFlt/PlGF ratio may eventually need to be stratified by preexisting conditions.

The final implications for the observations of this study are perhaps the most tantalizing. If there is a causal relation between the level of the sFlt-1/PlGF ratio and the morbidity of preeclampsia with severe features, then lowering the circulating concentration of sFlt-1 would ameliorate not only the morbidity but also the risk of preeclampsia. Work with plasma phoresies has suggested that this might be possible, albeit via a clinical intervention demanding more intensive resources. The potential for a targeted pharmacologic moderation of sFlt-1 levels would hold great promise in that those identified as at increased risk could be offered an intervention widely available to all.

 

 

Thadhani R, Lemoine E, Rana S, et al. Circulating angiogenic factor levels in hypertensive disorders of pregnancy. N Engl J Med. 2022;1. DOI: 10.1056/EVIDoa2200161

EXPERT COMMENTARY
 

The standard core lecture on preeclampsia given to all medical students frequently begins with an epic, if not potentially apocryphal, statement regarding how this disease has been noted in the annals of medical history since the time of the Ancients. Although contemporary diagnostic criteria for preeclampsia are not that far out of date, they are close. The increased urinary protein loss and hypertension preceding eclamptic seizures was first noted at the end of the 19th century. The blood pressure and proteinuria criteria used for diagnosis was codified in its contemporary form in the late 1940s. Since then, “tweak” rather than “overhaul” probably best describes the updates of the obstetrical community to the definition of preeclampsia. This has just changed.

 

Details of the study

Thadhani and colleagues prospectively recruited a nationally representative observational cohort of patients hospitalized for hypertension during pregnancy, then followed the patients until either the diagnosis of preeclampsia with severe features or for 2 weeks, whichever came first. At enrollment, circulating levels of the soluble fms-like tyrosine kinase 1 (sFlt-1) and placental growth factor (PlGF) were measured. In a 2-phased design, the first 219 participants were used to define a sFlt-1/PlGF ratio that would predict progression to severe preeclampsia within 2 weeks. The next 556 enrollees served to validate the predictive properties of the ratio. The authors found that a sFlt-1/PlGF ratio of ≥40 predicted progression to preeclampsia with severe features with an area under the curve (AUC) of 0.92.

As products of the trophoblasts, both sFlt-1 and PlGF have been mooted for almost 2 decades as potential predictive, if not diagnostic, aids with respect to preeclampsia. Indeed, both analytes are commercially available in Europe for specifically this purpose and many maternal-fetal medicine practitioners working in the European equivalent American tertiary referral centers use an sFlt-1/PlGF ratio as their primary criteria for a diagnosis of preeclampsia. Within the United States, there was an initial flurry of interest in and an infusion of corporate and federal research support for sFlt-1 and PlGF as diagnostic aids for preeclampsia in the mid-2000s. However, at present, the US Food and Drug Administration (FDA) has not sanctioned these (or any) biomarkers to aid in the diagnosis of preeclampsia. As Thermo-Fisher Scientific (Waltham, Massachusetts) is a supporting partner in this study, it is almost certain that these data will be submitted for review by the FDA as part of an application for a preeclampsia diagnostic. At some point in the near future, American practitioners will potentially be able to join their European colleagues in utilizing these biomarkers in the diagnosis of preeclampsia with severe features. ●

WHAT THIS EVIDENCE MEANS FOR PRACTICE

Thadhani and colleagues observed that the majority of both maternal and neonatal morbidity in their study, including 8 of the 9 neonatal deaths and both cases of eclampsia, occurred among patients with a ratio ≥40 at admission. There was an inverse relation between the sFlt-1/PlGF ratio and the admission to delivery interval. Where only 17% of patients in the highest quartile of ratios remained pregnant at 14 days post-enrollment, more than 79% of the lowest quartile were still pregnant. If not a causal relationship, sFlt-1 and PlGF are clearly associated with not only the occurrence of preeclampsia with severe features but also the degree of morbidity.

The implication for the disposition of patient care resources is clear. Patients at higher risk for preeclampsia could be seen in specialty high-risk clinics with an emphasis on increased monitoring. In situations where tertiary care is more remote, plans could be developed should they need to be transported to centers able to provide the appropriate level of care. Conversely, patients screening at lower ratios may be more appropriately managed as outpatients, or at least in less clinically involved accommodations.

Thadhani et al do note that there were false negative cases in which the sFlt-1/PlGF ratio at admission was <40 but patients nonetheless progressed to preeclampsia with severe features. The majority of these cases had concurrent pre-pregnancy, chronic hypertension. This observation suggests not only the potential for insights into the pathophysiology of the hypertensive diseases in pregnancy but also that the interpretation of the sFlt/PlGF ratio may eventually need to be stratified by preexisting conditions.

The final implications for the observations of this study are perhaps the most tantalizing. If there is a causal relation between the level of the sFlt-1/PlGF ratio and the morbidity of preeclampsia with severe features, then lowering the circulating concentration of sFlt-1 would ameliorate not only the morbidity but also the risk of preeclampsia. Work with plasma phoresies has suggested that this might be possible, albeit via a clinical intervention demanding more intensive resources. The potential for a targeted pharmacologic moderation of sFlt-1 levels would hold great promise in that those identified as at increased risk could be offered an intervention widely available to all.

Issue
OBG Management - 35(1)
Issue
OBG Management - 35(1)
Page Number
12-13
Page Number
12-13
Publications
Publications
Topics
Article Type
Sections
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.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media