ArticlePDF Available

The 5-HT2C receptor as a therapeutic target for alcohol and methamphetamine use disorders: A pilot study in treatment-seeking individuals

Wiley
Pharmacology Research & Perspectives
Authors:

Abstract

Alcohol use disorder (AUD) and methamphetamine use disorder (MUD) are prevalent and have high adverse impacts on both the individual and society. Current treatment strategies for these disorders are ineffective at a population level. Lorcaserin, a 5-HT2C receptor agonist, has shown potential at reducing the symptoms of substance use disorder. This pilot study (initiated prior to market withdrawal) examined feasibility and safety of lorcaserin treatment in people undergoing residential detoxification and treatment for AUD and MUD. This was an open label pilot study of lorcaserin where participants (n = 10 AUD; n = 8 MUD) received 10-mg lorcaserin daily for 4 days then twice daily for 1 month. Primary outcome measures included recruitment and retention rate, incidence of treatment-emergent events, incidence of methamphetamine or alcohol withdrawal-related events, heart rate, and blood pressure. Secondary measures included pharmacokinetic data and self-reported alcohol or methamphetamine use, craving, and psychological distress. AUD participants were recruited faster and had a greater retention rate compared with MUD participants. Lorcaserin did not alter vital signs, was well tolerated, and had a similar pharmacokinetic profile to individuals with obesity. Lorcaserin reduced self-reported alcohol and amphetamine-type substance use and craving in AUD and MUD participants, respectively. Self-reported psychological health also improved over the treatment period for all participants. Despite the pilot nature of this study, our data support the notion of 5-HT2C receptors as a therapeutic target for drug and alcohol abuse.
Pharmacol Res Perspect. 2021;9:e00767. 
|
1 of 11
https://doi.org/10.1002/prp2.767
wileyonlinelibrary.com/journal/prp2
Received:5Februa ry2021 
|
Accepted:5March2021
DOI: 10.1002/prp2.767
ORIGINAL ARTICLE
The 5- HT2C receptor as a therapeutic target for alcohol and
methamphetamine use disorders: A pilot study in
treatment- seeking individuals
Erin J. Campbell1| Yvonne Bonomo2| Adam Pastor2| Lisa Collins2|
Amanda Norman2| Peter Galettis3| Janice Johnstone3| Andrew J. Lawrence1
Thisisanop enaccessarticleundertheter msoftheCreativeCommonsAttributionLicense,whichpe rmitsuse,distributionandreproductioninanymedium,
provide d the original wor k is properly cited.
©2021TheAuthors.Pharmacology Researc h & Perspectivespublishe dbyJohnWiley&SonsLtd,BritishPharmacologicalSociet yandAmericanSocietyfor
PharmacologyandExperimentalTherapeutic s.
ErinJ.Ca mpbe llandYvon neBono moareco-firs tautho rs.
Abbreviations:AUD,Alcoho lusediso rder;B MI,bo dymassi ndex;FDA ,Fooda ndDru gAdmini strat ion;MU D,metha mphet amineu sedisor der.
1Florey Institute of Neuroscience and
MentalH ealth ,MelbourneBr ainCentre,
TheUniversityofMelbourne,Parkville,
VIC,Australia
2Depar tmentofAddictionMedicine,
StVincent'sHospitalMelbour ne,The
UniversityofMe lbourne,Parkville,VIC ,
Australia
3SchoolofMedicineandPub licHealth,
TheUniversityofNewcastle,C allaghan,
NSW,Austr alia
Correspondence
AndrewJ.Lawrence,FloreyInstitute
ofNeuroscienceandMentalHealt h,
MelbourneBrainCentre,TheUniversity
ofMelbourne,Pa rkville,VIC3052,
Australia.
Email:andrew.lawrence@florey.edu.au
YvonneBonomo,Departmentof
AddictionMedicine,StV incent'sHospital
Melbourne,TheUniver sityofMelbourne,
Parkville,VIC3010,Australia.
Email:yvonne.bonomo@svha.org.au
Funding information
AJLisaNHMRCPrincipalFellow
(1116930).YBreceivedaSeedingGrant
fromSVHMResearchEndowmentFund
(REF81803).
Abstract
Alcohol use d isorder (AUD) and met hamphetamine us e disorder (MUD) are prev a-
lent and have high adverse impacts on both the individual and society. Current treat-
mentstrategiesforthesedisordersareineffectiveatapopulationlevel.Lorcaserin,a
5-HT2Creceptoragonist,hasshownpotentialatreducingthesymptomsofsubstance
usedisorder.Thispilotstudy(initiatedpriortomarketwithdrawal)examinedfeasibility
andsafetyoflorcaserintreatmentinpeopleundergoingresidentialdetoxificationand
treatmentforAUDandMUD.Thiswasanopenlabelpilotstudyoflorcaserinwhere
participants (n=10AUD;n=8MUD)received10-mglorcaserindailyfor4daysthen
twicedailyfor1month.Primar youtcomemeasuresincludedrecruitmentandreten-
tionrate,incidenceoftreatment-emergentevents,incidenceofmethamphetamineor
alcoholwithdrawal-relatedevents,heartrate,andblood pressure.Secondarymeas-
uresincludedpharmacokinetic dataandself-reportedalcohol or methamphetamine
use,craving, and psychologicaldistress. AUDparticipantswererecruited fasterand
hadagreaterretentionratecomparedwithMUDparticipants.Lorcaserindidnotalter
vitalsigns,waswelltolerated,andhadasimilarpharmacokineticprofiletoindividuals
with obesity.Lorcaserin reduced self-reported alcohol andamphetamine-typesub-
stanceuseandcravinginAUDandMUDparticipants,respectively.Self-reportedpsy-
chological health also improved over the treatment period for all participants. Despite
thepilot nature of thisstudy,ourdata support thenotion of 5-HT2C receptors as a
therapeutic target for drug and alcohol abuse.
KEYWORDS
5-HT2Cagonist,alcohol,craving,lorcaserin,methamphetamine,serotonin
2 of 11 
|
   CAMPBELL Et AL .
1 | INTRODUC TION
Alcoholusedisorder(AUD)posesamajorsocialandeconomicbur-
dentosoci et y,accountingfor~5%ofde athsworl dwidein2016a nd
cost ingupwa rd of $3 6b illio n/ ye arinAus tra li a.1 With regard to illicit
druguse,methamphetamine is the second most regularly used drug
followingcannabisinAustralia,andintheUnitedStates,therewas
athreefoldincreaseinthenumberofmethamphetamine-associated
deaths between 2010 and 2015.2,3Asaresult,therecreationaluse
and abuse of d rugs continues to b e a major global public health is sue.
Importantly, poor retention and frequent relapse remain serious
obstacles for the treatment of subst ance use disorders with con-
tinuous and intense cravings persisting both during and following
treatment.4, 5TherearethreeFoodandDrugAdministration(FDA)-
approved pharmacotherapies for AUD,disulfiram, naltrexone, and
acamprosate.CurrentpharmacotherapeutictreatmentsforAUDre-
main inef fective at a population level due to a combination of limited
effectivenessandunder-prescribing.6,7TherearenoFDA-approved
pharmacotherapies specifically for methamphetamine use disorder
(MUD).Inarecentreview,mostmedicationsevaluatedwerefound
to not have a statistically significant benefit.8Clearly,thereisneed
for improved pharmacotherapeutic treatments aimed at reducing
drug-relatedcravingforbothAUDandMUD.
The neural circuitry surrounding craving and relapse to drug use
involves several brain structures including the ventral tegmental
area, st riatal comp lex, amygdala , hippoca mpus, and th e prefronta l
cortex.9,1 0Serotonin-containingcells (5-hydroxytryptamine[5-HT])
arelocatedpredominantlyintheraphenucleiofthemid/hind-brain,
including the dorsal and median raphe.11 From here , 5-HT fibers
widely innervate the central nervous system including multiple
nodes of reward-related circuitry.12 Indeed, the serotonergic sys-
tem has been implicated in substance use disorder and relapse for
several decades and may represent an avenue for future pharma-
cological interventions.1 3 1 8 Thereare14knownsubtypesof5-HT
receptors, andserotoninsignalingiswellknowntomodulatedopa-
mine activity.19Specifically,the5-HT2C receptorisexpressedinthe
hippocampus,striatum,andamygdalainbothratandhuman.2 0– 2 2
5-HT2C receptor signaling has been implicated in the development
andmaintenanceofAUDandMUD.Forexample,the5-HT2C receptor
agonist,Ro60-0175,decreased alcoholconsumptionin rats whereas
the5-HT2Creceptorantagonist, S B - 2 4 2 0 8 4 ,increased alcohol con-
sumption.23However,Ro60-0175reducedbothalcohol(gelsolution)
andvehicle(plaingelcontainingpolycose)operantself-administration
in rats suggesting possible nonspecific ef fects on caloric intake.24 5-
HT2C receptor signaling is also involved in methamphetamine use; for
example,Ro60-0175reversedmethamphetamineself-administration-
induced decreases in nucleus accumbens shell excitability.25
Additionally, methamphetamine-induced behavioral sensitization is
associatedwith a functional upregulation of5-HT2C receptors in the
ventral pallidum.26Together,thesepreclinicalstudies highlighta role
forthe5-HT2C receptor in both alcohol and methamphetamine use.
The development of therapeutic drugs that selectively target in-
dividual5-HT2receptorsubtypesisdifficult. Indeed, most5-HT2C
receptor a gonists also bi nd to 5-HT2A and/or 5-HT2B receptors.27
Lorcaserinisaselec tiveserotonin2C(5-HT2C) receptor agonist with
a 3-benzazepine scaffold,28 developed as an anti-obesity medica-
tion.Notably,lorcaserinreducestheconsumptionofalcoholinrats
andmethamphetamineuseinrhesusmonkeys,29, 30 implicating the
5-HT2C receptor as a potential treatment target for alcohol and sub-
stance use disorders.15Untilrecently,lorcaserin(Belviq®)wasFDA-
approved asananti-obesitymedication31 but was withdrawn from
the market af ter a safety trial indicated an increased occurrence of
cancer,where7.7%ofparticipantsreceivingdrugdevelopedcancer
relative to7.1%in the placeboarm.31Here, wecarried out a pilot
study, prior to market withdrawal, to evaluate the abilit y of lorca-
serin to suppress alcohol and methamphetamine craving and con-
sumptionintreatment-seekingAUDorMUDparticipants.
2 | MATERIALS AND METHODS
2.1  | Study design and participants
This was an open label pilot study. The protocol and amendments
were approved by the Human Research Ethics Committee of St
Vincent's Hospital Melbourne (HREC031/17).Eligibleparticipants
were males and females over the age of 18 years, diagnosed with
alcohol or methamphetamine substance use disorder (DSM5).
Exclusioncriteriaincludedpregnant(urineβHCGpositive)orbreast-
feeding; highly dependent on medical care for co-existing condi-
tions; other medical treatments for substance dependence including
anti-craving (e.g., acamprosate and naltrexone), aversive (e.g., di-
sulfir am), or substitu tion (e.g., atomoxetine, dexamphetamine, and
methylphenidate) treatments; known allerg y to lorcaserin; already
receivinglorcaserin;severeliverimpairment(ChildPughC);severe
renal impairment (creatinine clearance <30 ml/min); hypertension;
unstable diabetes; history of serotonin syndrome; low body mass
ind ex(BM I<20);andun st ablementals tate(in cludingac tivep sycho-
sisorschizophrenia).Written informedconsent was obtained from
all participants.
Thestudyprotocolspecifiedtherecruitmentof10AUDpartici-
pantsand10MUDparticipants.Recruitmentwasceasedwhenthe
initial FDA al ert was issu ed in Januar y 2020, resu lting in the f inal
recruitmentof10alcohol-and8methamphetamine-dependentpar-
ticipants.NotethattheHRECwasadvisedimmediatelybytheChief
InvestigatorsoftheoriginalFDA alertandthesubsequentproduct
withdrawal. All participants were advisedinwritingof theproduct
withdrawal and the health risks associated with their participation
in the study.
2.2  | Procedures
Participantswere treated with the lowest ef fective dose oflor-
caserin (immediate release) used in the treatment of obesity.
Participantsreceivedlorcaserin10mgoncedailyfor4daysthen
   
|
3 of 11
CAMPB ELL Et AL.
twice da ily for 1 month. All pa rticipant s received symptom atic
treatment using the St Vincent 's Hospital standard protocol.
Withdr awal was acti vely managed as ei ther an outpat ient or, if
required, in a residential setting. Alcohol withdrawal was man-
agedina ccordancewithCl in icalIns tituteWit hd rawalAs sessm en t
of Alcohol (CIWA-Ar).32 Prescribed medications consisted of
5– 20 mg of diazepam2hourlywithamaximumof80mgper
24-hperiod, anti-emetics for nausea, andparacetamol and non-
steroidalanti-inflammatoriesforaches.Allmedicationswereused
onlyasrequiredandceased byDay7followingparticipants’ last
use of alcohol or methamphetamine and the commencement of
lorcaserin.
2.3  | Pharmacokinetic sampling
OnDay7oftreatment,4-mlEDTAbloodsamplesweretakenfor
assessmentofplasmaconcentrationlevelsoftheIP(lorcaserin)at
predose(time0),2,4,and8haf tertreatment.Bloodswererefrig-
erated at ≤4°Candcentrifuged at 2000g for 10 min within 12 h
after blood sampling. Immediately after centrifugation, plasma
were stored in two labeled polypropylene tubes and stored at
≤−20°C forplasma concentration analysis. Allplasma concentra-
tion analyses were performed after all participants had completed
the final visit.
2.4  | Measures
Measures were assessed over five treatment time points (base-
line, Day 7, Day 14, Day 21, and Day 28). A basel ine researcher-
administered questionnaire assessed demographic and clinical
characteristics anddetermined eligibility.Liver function andblood
glucose wereassessed every second week (baseline,Day 14, and
Day28).
The Obsessive Compulsive Drinking Scale was used to assess
alcohol craving.33 A total craving score was calculated by sum-
ming the 14 items of this questionnaire, which were ranked on
a Likert s cale rangin g from 1 to 7.Su mming Items 1–6 calculat ed
the obsessive subscale, and summing Items 7–14 calculated the
Compuls ive Subscale . A brief, 10-item Meth amphetami ne Craving
Quest ionnaire, based o n the Cocaine Craving Qu estionnaire , was
used to assess methamphetamine craving over time. Scores across
each item were averaged.34,35
The KesslerPsychologicalDistressScale(K10)yieldeda global
measureofdistress,whichwasthesumofall10items.
TheAustralianTreatmentOutcomesProfile (ATOP),a validated
Australianversion oftheUKTreatmentOutcomeProfile,was used
toassessself-reporteddruguseandhealthandwell-being.36Higher
scores on t he substan ce use questio ns, measure d using the time-
linefollow-backmethod,reflectedmoredaysofusewhereashigher
scores on h ealth and well-bein g questions indicated gre ater self-
rated health outcomes.
2.5  | Outcomes
Primaryendpoints wereprespecified.Feasibilityendpointswere
recruitment rate and retention in treatment at Days 7, 14, 21,
and28.Safetyendpointswereincidenceoftreatment-emergent
adverseevents (AEs);incidenceof methamphetamine oralcohol
withdrawal-related treatment-emergent events; heart rate; and
blood pressure.
Secondary endpoints weremethamphetamine use (self-report,
salivascreen,andurinedrugscreen)onStudyDays0,7,14,21,and
28;alcoholuse (self-report,breathalcohol,bloodtesting,andurine
drugscreen)onStudyDays0,7,14,21,and28;cravingmeasures—
ObsessiveCompulsiveScale forDrinking(AUDgroup)andCocaine
CravingQuestionnaire(MUDgroup)onStudyDays0,7,14,21,and
28; and K10 psycho logical dist ress and ATOP measures on St udy
Days0,7,14,21,and28.
Afinal outcomewastoassess thepharmacokineticsoflorca-
serin ina clinical population with normalBMI: AUC0–8 h, Cmax,
t½,tmax.
2.6  | Pharmacokinetic analysis
PharmacokineticdatawereanalyzedattheClinicalPharmacology
Laboratory, School of Medicine and Public Health, University
of Newcast le, Australia . Plasma samples we re analyzed using a
validated liquid chromatography with tandem mass spectrom-
etry (LCMSMS) method for lorcaserin. Plasma samples (50 µl)
were prepared by adding twice the volume of acetonitrile, sam-
ples were vortexed and then centrifuged, and the supernatant
was transferred to a vial and injected onto the LCMSMS. The
LCMSMS system consisted of a Shimadzu 8060 LCMS using a
Kinetex C18column anda gradient of 0.1%formic acidandace-
tonitrile. Lorcaserin was linear over the range of 10–50 0 ng/ml.
Usingtheconcentration–timeprofilesdeterminedbytheanalysis
oftheplasmasamples,thepharmacokineticparameters,maximum
plasma concentration (Cmax), time of Cma x (Tmax), area unde r the
plasmaconcentrationtimecur ve(AUC0–12),andplasmahalf-life
(t½),weredeterminedusingPKSolver.37 The data were fitted using
the NCA Extravascular module, with AUC0 − t calculated using
the log-line ar trapezoi dal method . The 12-h t ime point for ea ch
individual was predicted with a one compartment model using
PKSo lve r.
2.7  | Statistical analysis
Unlessotherwisestated,datawereanalyzedseparatelyforAUDand
MUDparticipants.
Demographic data were summarized as numbers and pro-
portions for categorical data and mean (±standard error of mean
[SEM]) for conti nuous data usi ng Microsoft O ffice Exce l 2016.
Clinical characteristics and vital signs were analyzed using a
4 of 11 
|
   CAMPBELL Et AL .
repeatedmeasuresANOVA.Duetothehighdropoutr ateofpar-
ticipa ntswithM UD,eit herthree(baselinetoDay14)orf ivetime
points(baselinetoDay28)wereassessed.Theoralfluidtestand
urinarydrugscreenwereanalyzedusingtheFriedmannonpara-
metrictest.ForparticipantswithAUD,baselineandDay21oral
fluid substance results were not detected for one participant and
baseline blood glucose levels were missing for another partici-
pant.ForparticipantswithMUD,Day7bloodpressuredatawere
missing for one participant, baseline weight data were missing
for two pa rticipants , and Day 7 weight data were mi ssing for
one participant. To allow for the inclusion of these participants
instatistical analyses,a meanimputationofmissing valueswas
conductedinSPSS.ForMUDparticipants, one participant had
all liver function and blood glucose data missing and three par-
ticipants had their urine drug screen data missing and were thus
excluded from analyses. The Obsessive Compulsive Drinking
Scale,MethamphetamineCravingQuestionnaire,andK10Scale
were analy zed using a repe ated measures AN OVA as the dat a
werenormallydistributedandANOVAassumptionswerenotvi-
olated. A re peated measure s ANOVA was used to analy ze the
ATOP data. For par ticipants w ith AUD, the ATOP data for the
numberofdaysparticipantsdrankalcoholandthetypicalquan-
tity of alcohol consumed were not normally distributed and thus
assessed using the Friedman nonparametric test. Two partici-
pantshaddatamissingfromthehealthandwell-beingquestions
andwereexcludedfromthisanalysis.Onlyt wotothreepartici-
pantswithMUD completedtheATOPself-reportquestionnaire
forthedurationofthestudy(baselinetoDay28);thus,datawere
also analyzed from baseline to Day 14, with five participants
completing the questionnaire at these timepoints. All analyses
wereperformedusingSPSSv27(α = .05). Data are presented as
mean±SEM.
2.8  | Nomenclature of targets and ligands
Key protein targets and ligands in this article are hyperlinked
to corresponding entries in http://www.guide topha rmaco logy.
org, the comm on porta l for data from t he IUPHAR/B PS Guide to
PHARMACOLOGY,38 and are permanently archived in the Concise
GuidetoPHARMACOLOGY2019/20.39
3 | RESULTS
3.1  | Demographic information
The participants recruited with AUD were predominantly male
(8/10). The mean age wa s 48.1 years (SEM = 2 .1).Me an typic al
quantity of alcohol consumed on day used was 21.8 standard
drin ks(SEM=4. 4).T hepar tic ipa nt sre cruit edwit hM UDwer ep re-
dom inantlyma le(6/8)withame anage(M=36.6years,SEM=2.9)
of more than 10 yea rs younger tha n the AUD group. Th e mean
typical quantity ofmethamphetamine on aday usedwas 0.66 g
(SEM=0.22).
Table 1 describes further characteristics at baseline for both
participant groups. Age of use characteristics were younger for
AUD participants compared withMUD par ticipants. AUDpartici-
pantshadexperiencedmorewithdrawalsymptomscomparedwith
MUD par ticipants. The majority of AUD participants (90%) had at
leastone previoustreatmentfor dependence comparedwith50%
forMUDparticipants.ThetypesoftreatmentsforAUD were both
pharmacotherapiesandnonpharmacotherapies,but only nonphar-
macotherapiesforMUDparticipants.
3.2  | Primary endpoints
3.2.1  |  Feasibility
Thefirst participantwas recruitedintotheclinical trialonthe24th
ofJuly 2018, and the last participant was recruited on the 2nd of
December2019,equatingtoatotalofjustover16months(496days)
torecruitthe18participants.Forthealcoholgroup(n=10),theme-
dian time between recruiting each participant was just over 1 month
(median=37 days, interquartilerange=16–59days),that is, arate
of0.82AUD participantspermonth. FortheMUDgroup (n=8), it
tooklongerbetweenparticipantstoberecruited(median=73days,
interquartilerange=44–98days),thatis,arateof0.42participants
permonth.FortheAUDparticipants,theretentionintreatmentrate
atDay7was100%,and 80%foreach weekly visit thereafter.For
theMUD participants, the retention rate at Days 7,14, 21,and 28
was87.5%,62.5%, 37.5%,and 50%,respectively.Themain reason
for participant s discontinuing the trial was due to failing to attend
appointments. Note that one patient missed Day 21 appointment
butrepresentedfor Day28,andthisaccountsforthedipin there-
tentionratesovertimeatDay21.Eachwasdeemed“losttofollow-
up” after multiple unsuccessful at tempts by a clinician to contac t the
participant.
3.2.2  |  Safety
Treatment- emergent AEs
Eight AUD participants (80%) reported atleast oneAE, andseven
MUDparticipants(87.5%).Ofthese,sevenoftheAUDpar ticipants
(87.5%),andfiveMUDparticipants(71.4%),reportedatleastoneAE
that was considered by a study doctor to be related to lorcaserin.
Thes ei nc ludedra ti ngsofd efinite ly re late d,proba blyrelated,orpo s-
siblyrelated(Table2).OfallAEs(n=28),19wereconsideredtobe
treatmentemergent(67.9%).
Themostfrequentlyoccurringtreatment-emergentAEswere
“decreasedappetite”(21.1%, 4of19)andheadache(21.1%,4of
19).Theseoccur redthroughoutthe4weeksofstudymedication
administration.Thesecondmostfrequentlyoccurringtreatment-
emergentAEswere“lethargy”(15.8%, 3of19)and“dr ymouth”
   
|
5 of 11
CAMPB ELL Et AL.
(15.8%, 3 of 19), again occurring through the duration of the
trial. None of the treatment-emergent AEs was classified as
TAB LE 1  Participantcharacteristicsatbaseline
Characteristic
Number (%)
Alcohol use
disorder (n = 10)
Methamphetamine
use disorder (n = 8)
Countr y of birth
Australia 8(80.0) 6 (75.0)
Other 2 (20.0)
(Yugoslavia,
United
Kingdom)
2(25.0)(Scotland,
SouthAfrica)
IndigenousAustralian 1 (10.0) 0
Livingstatus
Alone 4(40.0) 2 (25.0)
Partner(withatleast
1 child)
3 (30.0) 2 (25.0)
Partner(nochildren) 2 (20.0) 1 (12.5)
Relationship status
Single 4(40.0) 5 (62.5)
Livingwithspouse
(including
married,defac to,
life partner)
4(40.0) 3 (37.5)
Separated but not
divorced
2 (20.0) 0
Accommodation
Private
accommodation
10 (100.0) 8(100.0)
Employment
Employed 4(40.0) 4(80.0)
Disability support
pension
2 (20.0) 1 (12.5)
Currently
unemployed
1 (10.0) 1 (10.0)
Highestlevelofeducation
Secondary (years
7– 10 )
2 (10.0) 3 (37.5)
Secondary (years
11– 1 2 )
1 (10.0) 3 (37.5)
Tertiary 6 (60.0) 1 (12.5)
Postgraduate 1 (10.0) 1 (12. 5)
Medicalhistor y
Current conditions
Depression 4(40.0%) 3(37.5%)
Anxiety 4(40.0%) 3(37.5%)
Highcholesterol 2(20.0%) 0
Alcohol-relatedconditions
None 7(70.0%) 6(75.0%)
Seizures 2(20.0%) 0
Alcohol or
methamphetamine use
history Alcohol use
Methamphetamine
use
Estimated
amount
(standard
drinks)
Estimatedamount
(g)
(Continues)
Alcohol or
methamphetamine use
history Alcohol use
Methamphetamine
use
Mean(SE) 21.8(4.4) 0.66 (0.22)
Ageatfir st
drink (years)
Ageatfir stuse
(year s)
Mean(SE) 16.3(0.4) 24.0(3.1)
Ageatdaily
drinking
(year s)
Ageatproblematic
use (years)
Mean(SE) 31.6(2.8) 25.7(3.4)
Duration since
daily drinking
(year s)
Duration since
problematic use
(year s)
Mean(SE) 15.8(2.5) 10.4(2.5)
Previousdetoxifications
Atleast1
detoxification
9(90.0%) 4(50.0%)
Previouswithdrawalsymptoms
Sweating 9(15.0%) 1(1.7%)
Anxiety 8(13.3%) 3(5.0%)
Tre mor 7(11.7%) 1(1.7%)
Nausea 5(8.3%) 0
Agitation 5(8.3%) 3(5.0%)
Seizures 2(3.3%) 0
Vomiting 3(5.0%) 0
Diarrhea 2(3.3%) 0
Delirium tremens 1(1.7%) 0
Previoustreatmentfordependence
Atleast1treatment 9(90.0%) 4(50.0%)
Previoustypesoftreatment(oftenmultiple)
Counseling 9(100.0%) 4(100.0%)
Acamprosate
maintenance
9(100.0%) 0
Naltrexone
maintenance
8(88.9%) 0
Alcoholics
anonymous(AA)
7(77.8%) 0
Residential
rehabilitation
activities
4(44.4%) 1(25.0%)
Disulfiram
maintenance
2(22.2%) 0
Other 3(33.3%)
(recover y
and support
program,2×
baclofen)
1(25.0%)( The
Rewired
Program—self-
administered)
TAB LE 1  (Continued)
6 of 11 
|
   CAMPBELL Et AL .
“severe.” One was classified as “moderate” severity.This event
of headache repor ted at Day 21 lasted 2 days and resolved spon-
taneously.Theremaining18treatment-emergentAEswereclas-
sified as “mild”severity. None of thetreatment-emergent AEs
wasaseriousAE.
Withdrawal- related treatment- emergent AEs (n = 1)
Oneeventwas“probablyrelated”tolorcaserin.AnAUDparticipant
exp er iencedongoingfatigue.Theydrop pe doutatDay9,an dnofur-
therfollow-upbythestudystaffwasachieved.
Vital signs
Forparticipantswith AUD, temperature,pulse, systolic anddias-
tolicblood pressure,andweightdidnotsignificantly changeover
time (Table S1). Respiratory rate (breaths/min) significantly in-
creased over the treatment period (Table S1). For par ticipants with
MUD,temperature, pulse, respiratory rate, systolicand diastolic
bloodpressure,andweightdidnot significantlychangeovertime
(Table S2).
3.3  | Secondary endpoints
3.3.1  |  Substanceuse
Alcohol use
Self-repor t. FortheAUDgroup,thenumberofdaysdrinkingalcohol
in the past week did not change between baseline and Day 28
(Table3);however,thetypicalquantit yofalcoholconsumedonany
given day did decrease over time (Table 3).
Clinicalcharacteristics. FortheAUDgroup,breathalcoholcontent,
live rf unc t io n,b loo dg luc os eleve ls,an dur in arydrugsc ree nr esu lt s
did not change over the treatment period (Table S3).
Methamphetamine use
Self-report. FortheMUDgroup,frombaselinetoDay14,thenumber
of days using an amphetamine-typesubstanceinthepast weekdid
notchange(TableS4). FrombaselinetoDay28,the numberof days
usinganamphetamine-type substancein the past week decreased
(Table 3).
Clinicalcharacteristics. Forthe MUD group, liver function,blood
glucose levels, urinary drug screen results, and oral fluid test
results did not change over time (Table S5).
3.3.2  |  Craving
Total craving scores for participants with AUD decreased over
time (F4, 28=7.299,p<.001;Figure1A).Scoresontheobsessive
subscale (F4,28=5.200, p = .0 03) and on the compulsive subscale
(F4, 28 = 7.689, p < .001) also significantly decreased over time
(Figure1A).
Methamphetamine craving for participants with MUD signifi-
cantly decreased frombaseline to Day 14 (F2 ,8 = 5.20 0, p = .036)
andapproachedsignificancefrombaselinetoDay28(F4,8=2.880,
p=.095;Figure1B).
TAB LE 2  Numberoftreatment-emergentadverseevents
(n=19)(definitelyrelated,probablyrelated,orpossiblyrelatedto
lorcaserin)groupedbySystemOrganClassandpreferredterm,for
both groups of participants
Event (System Organ Class/
preferred term)
Alcohol use
disorder
(n = 10)
Methamphetamine
use disorder (n = 8)
Number of participants with
≥1medication-related
adverse event
7 5
Gastrointestinaldisorders
Diarrhea 0 1
Dry mouth 2 1
Lipdry 1 0
Nausea 0 1
Generaldisordersandadministrationsiteconditions
Lethargy 3 0
Metabolismand
nutrition disorders
Decreased appetite 3 1
Nervous system disorders
Dizziness 2 0
Headache 2 2
TAB LE 3 ATOP self- report data from baseline to Day 28 for
participants with alcohol use disorder and methamphetamine use
disorder
ATOP self- report F statistic χ2p value n
Alcoholusedisorderparticipants
Number of days drank
alcohol in the last 7 days
5.756 .218 8
Typicalquantity(standard
drinks) of alcohol
consumed on day used
12.860 .012 8
Psychologicalhealthstatus 16.139 <. 001 6
Physicalhealthstatus 2.828 .052 6
Quality of life 5.689 .003 6
Methamphetamineusedisorderparticipants
Number of days used
amphetamine-type
substance in the last
7 days
4.214 .040 3
Psychologicalhealthstatus 51.000 .001 2
Physicalhealthstatus 1.370 .384 2
Quality of life 1.190 .435 2
Abbreviation:ATOP,AustralianTreatmentOutcomesProfile.
   
|
7 of 11
CAMPB ELL Et AL.
3.3.3  |  K10psychologicaldistress
K10 psychological distress scores significantly decreased over time
forparticipant swithAUD(F4,28=20.629,p < .001; Figure 1C).
K10 psychological distress scores did not change between
baselineandDay14(F2, 8=4.193,p = .057) or bet ween baseline
and Day 28 for pa rticipa nts with MUD (F4 , 8 = 1.537, p = .280;
Figure 1D).
3.3.4  |  ATOPself-reporthealthandwell-beingdata
FortheAUDparticipants,psychological healthstatus and qual-
ityoflifesignificantlyincreasedacrosstheexperimentalperiod
between baseline and Day 28, and physical health status ap-
proachedasignificantimprovement(Table3;Figure1E).
FortheMUDparticipants,frombaselinetoDay14,theperceived
health a nd well-being i n the past wee k did not change ( TableS 4).
FIGURE 1 Self-reportedcraving,psychologicaldistress,andhealthandwell-beingafterlorcaserintreatmentinpar ticipantswithalcohol
usedisorder(AUD)andmethamphetamineusedisorder(MUD).Totalobsessivecompulsivedrinkingscoreoverthe28-daytreatmentperiod.
Tot a l n=8.**p<.01foreachcravingmeasure(A).Averagecravingscoreoverthe28-daytreatmentperiod.n=3–5.*p < .05 baseline to Day
14(B).KesslerPsychologicalDistressself-reportdataoverthe28-daytreatmentperiod.Totaln=8forAUDparticipants,n=3–5forMUD
participants.***p<.001(CandD).Psychologicalhealthstatusandqualityoflifeincreasedoverthetreatmentperiod;however,therewas
nosignificantchangeinphysicalhealthstatusforAUDparticipants.Totaln=8.**p<.01forpsychologicalhealthstatusandqualityoflife
(E).Psychologicalhealthstatusincreasedoverthetreatmentperiod;however,therewasnosignificantchangeinphysicalhealthstatusor
qualityoflife.n=2–5.***p=0.001forpsychologicalhealthstatus(F).Alldataarepresentedasmean±SEM
8 of 11 
|
   CAMPBELL Et AL .
FrombaselinetoDay28,thepsychologicalhealthstatusinthepast
week increased; however,physical health status and quality of life
did not change (Table 3; Figure 1F).
3.4  | Pharmacokinetics of lorcaserin
Pla sm aconcen tr at io ntimeprof il es(0,2,4, 8,and12h)forsixpar tic-
ipants are shown in Figure 2. The pharmacokinetic profile followed
an oral absorption profile (Figure S1) with a Tmax at 2 h and a Cmax
of102± 25ng/mlbeforefollowingaonephaseexponentialdecay,
resulting in a t1/2of7.9±2h.Pharmacokineticparametersaresum-
marizedinTable4.
4 | DISCUSSION
The aim of our pilot studywas to examine the safety and efficacy
of the 5-HT2C receptor agonist lorc aserin in people undergoing
resident ial withdrawa l and seeking tr eatment for AUD an d MUD.
There are l imited data on t he pharmaco kinetics of l orcaseri n, and
in genera l, this has been ob served in an ob ese populatio n with a
Tmaxof1.5h,Cmaxof80ng/mlwithavariabilityof26%,andat½ of
11.9 ± 1.5 h.40InAUDandMUDparticipants,weobservedaslightl y
longer Tmaxof2 .4h ,a highe rCmax of 102 ng/ml with similar variability
of26%,andashortert½of7.9±2h.Withthesmallsam plesizesand
similarvariabilit yofapproximately30%inbothstudies,itisunlikely
that there is a different pharmacokinetic profile between an obese
andAUD/MUDpopulations.
In the present study, AUD participants were recruited fas ter
andhadagreaterretentionratecomparedwithMUDparticipants.
There were no significant changes in vital signs over the treatment
period except respiratory rate did increase for AUD participants.
Importantly,lorcaserin was well tolerated, having few,if any,side
effects. Lorcaserin was not anorectic and had no cardiovascular
side effects. We also tested the effect of lorcaserin on alcohol and
Methamphetaminecraving andconsumption.Althoughnumberof
drinking days did not change, self-reported alcohol consumption
per session decreased for AUD participants and amphetamine-
type substance use decreased for those MUD participants who
weretained.However,therewasnochangeinclinicallymonitored
alcoholor amphetaminecontent,respectively.Notably,lorcaserin
treatme nt resulted in dec reased craving ove r time for both AUD
andMUDparticipants.MeanbaselinecravingscoresforAUDpar-
ticipant s in this study were high compared with those reported for
other similar populations 41andde cr ea se dbymorethan 50 %atDay
28. MUD participants’ craving scorestrended toward significance
butshowed considerable interparticipant variability.Self-reported
psychologicaldistress scores decreased for AUD participants,but
therewasnochangeinpsychologicaldistressforMUDparticipants.
Nevertheless,self-reportedpsychologicalhealthstatussignificantly
improvedoverthetreatmentperiodforbothAUDandMUDpartic-
ipants.K10scoresforAUDparticipantsdroppedtotheAustralian
national average by Day 7.42Overall,anddespitethepilotnatureof
thisstudy,ourdatado indicateclinicallysignificantimprovements
onkey outcomes and support thenotionof5-HT2C receptors as a
therapeutictargetfordrugandalcoholabuse.Clinically,28daysisa
shortdurationoftreatmentforbothalcoholandMethamphetamine
dependence.Longer treatment duration may haveseenimproved
outcomes on a number of physiological variables that were trending
positively(e.g.,GGTlevels).
Lorcaserin has shown promise at reducing symptoms of sub-
stance usedisorder in other,recentclinical trials. For example, lor-
caserin reduced cannabis intake compared with placebo controls.43
Lorcaserin, in combination with counseling, increased abstinence
rates in individuals with nicotine use disorder compared with those
treated with smoking cessation counseling alone.44However,lorca-
serin did not reduce cocaine or oxycodone use compared with pla-
cebo controls.45,46Lorcaserinalsodidnotimproveextended-release
naltrexone induc tion rates in an outp atient sample of individuals
with opioid use disorder compared with placebo.47 There are some
discrepancies regarding the effectiveness of lorcaserin at improving
treatmentoutcomesforthosewithsubstanceusedisorder,but the
reasons for these discrepancies are currently unknown and may be
due to the mechanistic impac t of lorcaserin and the interaction with
the drug of choice based on the neural circuits driving the behavior.
Nevertheless, at least foralcohol, nicotine, an d cannabis, t here is
supporting evidence.
FIGURE 2 Plasmaconcentrationtimeprofiles(0,2,4,8,and
12 h) after lorcaserin was administered. The pharmacokinetic
profile of lorcaserin followed a standard oral absorption profile
forthesixparticipantsexamined.The12-htimepointforeach
individual was predicted with a one compartment model using
PKSo lve r.37AUD,alcoholusedisorder;MUD,methamphetamine
use disorder
TAB LE 4  Pharmacokineticparametersummarydata
Parameter Unit Mean SD CV (%)
t1/2 h7.9 2.0 26
Tmax h2.4 0.8 34
Cmax ng/ml 102 26 26
AUC0–12 ng/ml·h 824 311 38
Css ng/ml 69 26 38
   
|
9 of 11
CAMPB ELL Et AL.
Our findings are also somewhat consistent with preclinical liter-
ature showing reductions in alcohol consumption in rats following
chronic lorcaserin treatment.30 Lorcaserin can also decrease self-
administration of methamphetamine and cocaine in rhesus mon-
keys.29,48,49Similarly,lorcaserindecreasedheroinself-administration
andcue-inducedreinstatementinrhesusmonkeys.50, 51Finally,both
acute and chronic administration of lorcaserin plus pimavanserin,a
selective 5-HT2A receptor antagonist, decreased cocain e relapse-
like behavior in rats, suggesting that perhaps the actions of the
5-HT2C receptor alone are not straightforward.52
Althou gh lorcaser in was recently wi thdrawn from t he market
followingasafetyconcern,wecontendtheinvolvementof5-HT2C
receptors in substance use disorder are relevant and targeting
these receptors is still a viable avenue for treatment, particularly
givenourcurrent findingsandthose discussedabove.The5-HT2C
receptorhasbeenimplicatedinbothcompulsive-andimpulsive-like
behaviorsinrodents,cardinalfeaturesofsubstanceusedisorder.53
Additionally, 5-HT2C receptor activity can modulate dopamine
function.54, 55Ofnotehoweveristhecomplexityofthe5-HTsys-
tem, particularly regarding the interaction between 5-HT2C and
5-HT2Areceptorsandtheirrelationshiptoimpulsive-likebehavior.56
4.1  | Methodological considerations and
future research
Amethodological limitation ofthecurrentstudyisthatallpartici-
pants receivedlorcaserin treatment; thatis,therewasnoplacebo
control. Additionally,this study was open label, limiting the inter-
pretation of our findings. Although overall sample sizewas small,
particularlyforMUDparticipants,thosethatcompletedthestudy
seemingly benefitted from lorcaserin. The differential outcomes be-
tweenAUDandMUDparticipant sgosomewaytomitigateagainst
theseshortfalls;however,furtherstudiesareclearlywarranted.
MUDparticipants weremore difficult to retain and had lower
retention in treatment. The observed difficulty in recruiting and
maintainingMUDparticipantsinthetrialcouldpossiblybebecause
methamphetamine users seek treatment reluctantly due to a lack
of perceived effective treatments. It could also simply reflect that
there aremore people withAUD than MUD. Therewas no differ-
enceintheratesofAEsbetweenthetwogroups,sothisisunlikely
to be a factor. It is difficult to draw firm conclusions due to the small
numberofparticipantsinthestudy.Anyfuturestudyshouldinclude
design elements to improve recruitment rates and mitigate against
attrit ion. For exam ple, continge ncy manage ment approa ches have
been shown to be effective in the treatment of many substance use
disordersandholdpromiseforMUD.5 7, 58
5 | CONCLUSION
Insummary,ourdata suggestthatthe 5-HT2C system is still a vi-
able treatment option forsubst anceuse disorders, particularly
AUD.However,giventhecurrentFDAwithdrawaloflorcaserin,
other 5-HT2C agonists with high selectivity may be beneficial.
Alternatively, due to the conserved nature of the orthosteric
bindingsiteforserotoninat5-HT2f ami lyr ece pto rs, sel ecti vepos -
itiveallosteric modulators, analogoustothoseforacetylcholine
muscarinicreceptors,59mayprovideafruitfullineofenquiryand
enablemoreselectivetargeting.Another promisingtherapeutic
target may b e the use of psyched elics, target ing the serotonin
system,forsubstanceusedisordertreatment.6 0, 61However,ani-
malstudieshaverecentlyquestionedtheefficacyofpsychedelics
inmodelsof AUD.62Nevertheless,ourfindingsaddtothelitera-
ture supporting the 5-HT2C receptor as a therapeutic target for
AUDandMUD.
ACKNOWLEDGMENT
We acknowledge the Victorian State Government Operational
Infrastructure Scheme.
DISCLOSURE
The authors report no conflict of interest.
AUTHOR CONTRIBUTIONS
AJL andYB conceivedand designed research; YB,AP,LC,and AN
col le ctedth edat a;EJ Canaly zeddata;P GandJJcondu cted an dana-
lyzedpharmacokineticdata;EJCandAJLinterpreteddataandwrote
thefirst draftofthemanuscript;allauthorscontributedto,edited,
and approved the final manuscript.
DATA AVA ILAB ILITY STATE MEN T
The data that support the findings of this study are available
from the correspondingauthor upon reasonable request. Some
data may not be made available bec ause of privacy or ethical
restrictions.
ORCID
Erin J. Campbell https://orcid.org/0000-0002-4722-6897
Andrew J. Lawrence https://orcid.org/0000-0001-6836-727X
REFERENCES
1. World HealthO rganization.Global status report on alcohol and
health.2018.
2. AustralianInstituteof HealthandWelfare. Nationaldrug strategy
household sur vey 2016: Detailed findings. 2017.
3. National center for health statistics/centers for disease control;
U.S. Department of Justice; Drug Enforcement Administration.
National drug threat assessment . 2017.
4. C ampbellEJ,LawrenceAJ,PerryCJ.Newstepsfortreatingalcohol
use disorder. Psychopharmacology.2018;235 (6):1759-1773 .
5. Sinha R, Shaham Y, Heilig M . Translational and reverse transla-
tional research on the role of stress in drug cr aving and relapse.
Psychopharmacology.2011;218(1):69-82.
6. KranzlerHR,SoykaM.Diagnosis and pharmacotherapy ofalcohol
use disorder: a review. JAMA.2018;320(8):815-824.
7. LeggioL,FalkDE,RyanML,FertigJ,LittenRZ.Medicationdevelop-
ment for alcohol use disorder: a focus on clinic al studies. Handbook
Exp Pharmacol.2020;258:443-462.
10 of 11 
|
   CAMPBELL Et AL .
8 . Ch an B, Freema n M, Kondo K , et al. Phar macother apy for meth -
amphetamine/amphetamineusedisorder-asystematicreviewand
meta-analysis.Addiction.2019;114(12):2122-2136.
9. EverittBJ, RobbinsTW.Neuralsystems ofreinforcementfordrug
addiction: from actions to habit s to compulsion. Nat Neurosci.
2005; 8(11):14 81-1489.
10. Koob GF, Volkow ND. Neurocircuitr y of addiction.
Neuropsychopharmacology.2010;35(1):217-238 .
11. Steinbusch HW. Distribution of serotonin-immunoreactivity in
the centr al nervous s ystem of the rat-cell b odies and term inals.
Neuroscience.1981;6(4):557-618.
12. Jacobs BL , Azmiti a EC. Struc ture and f unction o f the brain s ero-
tonin sys tem. Physiol Rev.1992;72(1):165-229.
13. Cloninger CR, Bohman M, Sigvardsson S. Inheritance of alco-
hol abuse. Cross-fostering analysis of adopted men. Arch Gen
Psychiatry.1981;38(8):861-868.
14. Cloninge r CR, Sigvards son S, von Knorr ing AL, Bohma n M. The
Swedish studies of the adopted children of alcoholics: a reply to
Littrell.J Stud Alcohol.1988;49:500-509.
15. CollinsGT,GerakLR,FranceCP.Thebehavioralpharmacologyand
therapeutic potential of lorcaserin for substance use disorders.
Neuropharmacology.2018;142:63-71.
16. EzakiN,NakamuraK ,SekineY,etal.Shor talleleof5-HTTLPRasa
risk factor for the development of psychosis in Japanese metham-
phetamine abusers. Ann N Y Acad Sci.2008;1139:49-56.
17. GilliganSB,ReichT,CloningerCR.Etiologicalheterogeneit yinalco-
holism. Genet Epidemiol.1987;4:395-414.
18. JohnsonBA,ElkashefAM,SeneviratneC,etal.;Methamphetamine
Study, G. Association between genotype of the serotonin
transporter-linkedpolymorphicregionoftheserotonintransporter
gene and age of onset of methamphetamine use: a preliminary anal-
ysis. Front Psychiatry.2010;1:145.
19. Costall B, NaylorRJ, MarsdenCD,Pycock C J. Serotonergicmod-
ulation of the dopamine response from the nucleus accumbens. J
Pharm Pharmacol.1976;28(6):523-526.
20. Abramowski D, Rigo M, Duc D, Hoyer D, St aufenbiel M.
Localization of the 5-hydroxytryptamine2C receptor protein in
human and rat brain using specific antisera. Neuropharmacology.
1995;34 (12):1635-1645.
21. Clemett DA, Punhani T, Duxon MS, Blackburn TP, Fone KC.
Immunohistochemicallocalisationofthe5-HT2Creceptorprotein
in the rat CNS. Neuropharmacology.2000;39(1):123-132.
22. Pasqualetti M, Ori M, Castagna M, Marazziti D, C assano GB,
Nardi I. Distribution and cellular localization of the serotonin
type 2C r eceptor messe nger RNA in human b rain. Neuroscience.
1999;92(2):601-611.
23. TomkinsDM ,Joharchi N, Tampakeras M, Martin JR, Wichmann
J,HigginsGA.An investigationof the role of 5-HT(2C) receptors
in modifying ethanol self-administration behaviour. Pharmacol
Biochem Behav.2002;71(4):735-744.
24. KasperJ,TikamdasR,KimMS,etal.Theserotonin-2receptormod-
ulator, (-)-trans-PAT,decreases voluntary ethanol consumption in
rats. Eur J Pharmacol. 2 0 1 3 ; 7 1 8 ( 1 3 ) : 9 8 - 1 0 4 .
25. Graves SM, Clark MJ, TraynorJR, HuXT,Napier TC . Nucleus ac-
cumbensshellexcitabilityisdecreasedbymethamphetamineself-
administrationandincreasedby5-HT2Creceptorinverseagonism
and agonism. Neuropharmacology.2015;89:113-121.
26. Napier TC, I stre ED. Meth amphetam ine-indu ced sensitiz ation in-
cludes afunctionalupregulationofventralpallidal 5-HT2A/2C re-
ceptors. Synapse.2008;62(1):14-21.
27. Wold EA, Wild CT, Cunningham K A, Zhou J. Targeting the 5-
HT2C receptor in biological context and the current state of
5-HT2C receptor ligand development. Curr Top Med Chem.
2019;19(16) :1381-1398.
28. Smith BM, Smith JM,TsaiJH, et al. Discovery and structure-ac tivity
relationship of (1R)-8-chloro-2,3,4,5-tetrahydro-1-methyl-1H-3-
benzazepine(Lorcaserin),a selectiveserotonin 5-HT2C receptor ago-
nist for the treatment of obesity. J Med Ch em.2008;51(2):305-313.
29. GerakLR, Collins GT,France CP.Effects of lorcaserinon cocaine
and methamphetamine self-administration and reinstatement of
responding previously maintained by cocaine in rhesus monkeys.
J Pharmacol Exp Ther.2016;359(3):383-391.
30. RezvaniAH,CauleyMC ,LevinED.Lorc aserin,aselective5-HT(2C)
receptoragonist,decreasesalcoho lint akeinfema leal coholprefer-
ring rat s. Pharmacol Biochem Behav.2014 ;125:8 -14.
31. Food and drug administration. https://www.fda.gov/drugs/drug-
s a f e t y - a n d - a v a i l a b i l i t y / f d a - r e q u e s t s - w i t h d r a w a l - w e i g h t - l o s s - d r u g -
b e l v i q - b e l v i q - x r - l o r c a s e r i n - m a r k e t . A c c e s s e d J u l y 1 0 , 2 0 2 0 .
32. Sullivan J T, Sykora K, Schneiderman J, Naranjo C A, Sellers EM .
Assessment of alcohol withdrawal: the revised clinical institute
withdrawal assessment for alcohol scale (CIWA-Ar). Br J Addict.
1989;84 (11):1353-1357.
33. AntonRF,MoakDH,LathamPK.Theobsessivecompulsivedrink-
ing scale: a new method of asse ssing outcome in alcoholism treat-
ment studies. Arch Gen Psych.1996;53(3):225-231.
34. Sussner BD, Smelson DA, Rodrigues S, Kline A , Losonczy M,
ZiedonisD.Thevalidityandreliabilityofabriefmeasureofcocaine
craving. Drug Alcohol Depend.2006;83(3):233-237.
35. Tiffany ST,Singleton E, Haer tzen CA ,Henning fieldJE.Thedevel-
opment of acocainecravingquestionnaire. Drugand alcohol de-
pendence. Drug Alc Dep.1993;3 4(1):19-28.
36. RyanA ,Holmes J,HuntV,etal.Validationandimplementationof
the AustralianTreatment OutcomesProfilein specialist drug and
alcohol settings. Drug Alc Rev.2014;33(1):33-42.
37. Zh ang Y,H uo M, Zhou J, X ie S. PKSo lver: an ad d-in progr am for
pharmacokineticandpharmacodynamicdataanalysisinMicrosoft
Excel.Comput Methods Programs Biomed.2010;99(3):306-314.
38. HardingSD,SharmanJL,FaccendaE,etal.TheIUPHAR/BPSGuide
to PHARMACOLOGY in 2019:updates and expansion to encom-
passthe newguide toIMMUNOPHARMACOLOGY.Nucleic Acids
Res.2018;46:D1091-D1106.https://doi.org/10.1093/nar/gkx1121
39. Alexander SPH, Christopoulos A , Davenport AP, et al. THE
CONCISE GUIDE TO PHARMACOLOGY 2019/20: G protein-
coupled receptor s. Br J Pharmacol. 2019;176 (S1) :S21-S141 .
40. ChristopherR,Morgan M,FerryJ, etal.Single- and multiple-dose
pharmacokineticsofalorcaserinextended-releasetablet.Clin Ther.
2016;38(10):2227-2238.
41. AntonRF,MoakDH,LathamP.The ObsessiveCompulsiveDrinking
Sc a le: as e lf-rated ins t r ume ntf ort h eq u ant ific atio no fth o ugh t sab o ut
alcohol and drinking behavior. Alc Clin Exp Res.19 95;19(1):92-99.
42. AndrewsG,SladeT.InterpretingscoresontheKesslerpsychologi-
cal dist ress sc ale (K10). Aust N Z J Pub Health.20 01;25(6):494-497.
43. Arout CA , Coope r ZD, Reed SC, e t al. 5HT-2C ago nist lorc aserin
decreasescannabis self-administration in dailycannabissmokers.
Addict Biol. 2021:e12993. https://doi.org/10.1111/adb.12993
44. Shanahan W R, Rose JE, Gli cklich A, Stubb e S, Sanchez-K am M.
Lorcas erinforsmokingcessat ionandass ociatedwe ightgain:aran-
domized12-weekclinicaltrial.Nicotine Tob Res.2017;19(8):944-951.
45. BrandtL,JonesJD,MartinezS,etal.Effectsoflorcaserinonoxyco-
doneself-administrationand subjective responses in participants
with opioid use disorder. Drug Alcohol Depend.2020;208:107859.
46. Pirtle JL, Hickman MD, Boinpelly VC, Surineni K, Thakur HK,
Grasing KW. The serotonin-2C agonist Lorcaserin delays intrave-
nous choice and modifies the subjective and cardiovascular ef-
fectsofcocaine:arandomized,controlledhumanlaborator ystudy.
Pharmacol Biochem Behav.2019;180:52-59.
47. Levin FR , Mariani JJ, Pavlicova M, et al. Lorcaserin treatment for
extended-releasenaltrexoneinductionandretentionforopioiduse
disorder individuals:a pilot, placebo-controlled randomizedtrial.
Drug Alc Dep.2021;219:108482.
48. Collins GT, France CP. Effects of lorcaserin and buspirone, ad-
ministered aloneand as a mixture, on cocaine self-administration
   
|
11 of 11
CAMPB ELL Et AL.
in male and female rhesus monkeys. Exp Clin Psychopharmacol.
2018;26(5):488-496.
49. CollinsGT,GerakLR,JavorsMA,FranceCP.Lorcaserinreducesthe
discriminative stimulus and reinforcing effects of cocaine in rhesus
monkeys. J Pharmacol Exp Ther.2016;356(1):85-95.
50. Gerak LR, CollinsGT,MaguireDR, France CP.Ef fects of lorcase-
rin on reinstatement of responding previously maintained by co-
caine or remifent anil in rhesus monkeys. Exp Clin Psychopharmacol.
2019;27(1):78-86.
51. Kohut SJ, Bergman J. Lorcaserin decreases the reinforcing ef-
fectsof heroin, but not food,inrhesusmonkeys. Eur J Pharmacol.
2018;840:28-32.
52. Anasta sio NC, Sholl er DJ, Fox RG, et al. Su ppression of coc aine
relapse-like behaviors upon pimavanserin and lorcaserin co-
administration. Neuropharmacology.2020;168:108009.
53. Higgins GA, Brown M, St John J, MacMillan C, Silenieks LB,
Thevar kunnel S. Ef fects of 5 -HT2C rece ptor modul ation and th e
NAreuptakeinhibitor atomoxetine in test sofcompulsiveandim-
pulsive behaviour. Neuropharmacology.2020;170:108064.
54. Di GiovanniG,Di MatteoV,Di MascioM, EspositoE.Preferential
modulation of mesolimbic vs. nigrostriatal dopaminergic function
byserotonin(2C/2B)receptoragonists:acombinedinvivoelectro-
physiological and microdialysis study. Synapse.2000;35(1):53-61.
55. Millan MJ, D ekeyne A, Gob ert A. Serot onin (5-HT)2C r eceptors
tonically inhib it dopamine (DA) an d noradrenalin e (NA), but not
5-HT, release in the frontal cortex in vivo. Neuropharmacology.
1998;37( 7):953-955.
56. WinstanleyCA,TheobaldDE,DalleyJW,GlennonJC,RobbinsTW.
5-HT2A and 5-HT2C receptor antagonists haveopposing effects
onameasureofimpulsivity: interactionswith global5-HTdeple-
tion. Psychopharmacology.2004;176(3–4):376-385.
57. BrownHD,DeFulioA.Contingencymanagementforthetreatment
of methamphetamine use disorder: a systematic review. Drug Alc
Dep.2020;216:108307.
58. Roll JM. Contingency management: an evidence-based compo-
nent of methamphetamine use disorder treatments. Addiction.
2007;102:114-120.
59. Wal ker LC, Berizz i AE, Chen NA , et al. Acety lcholine mus carinic
M4 receptors as a therapeutic target for alcohol use disorder:
converging evidence from humans and rodents. Biol Psychiatry.
2020;88(12):898-909.
60. AndersenKAA,Carhart-HarrisR ,Nut tDJ,ErritzoeD.Ther apeutic
effects of classic serotonergic psychedelics: a systematic re-
view of modern-era clinical studies. Acta Psychiatr Scand.
2021;143(2):101-118.
61. Nutt D. Psyc hedelic dr ugs-a new e ra inpsychi atry? Dialogues Clin
Neurosci.2019;21(2):139-147.
62. MeinhardtMW,GungorC,SkorodumovI,MertensLJ,SpanagelR.
PsilocybinandLSDhavenolong-lastingeffectsinananimalmodelof
alcohol relapse. Neuropsychopharmacology.2020;45(8):1316-1322.
SUPPORTING INFORMATION
Additional supporting information may be found online in the
Supporting Information section.
How to cite this article: CampbellEJ,BonomoY,PastorA,et
al.The5-HT2C receptor as a therapeutic target for alcohol
andmethamphetamineusedisorders:Apilotstudyin
treatment-seekingindividuals.Pharmacol Res Perspect.
2021;9:e00767. https://doi.org/10.1002/prp2.767
... Multiple 5-HT receptor ligands elevate GABA levels (Abi-Saab et al., 1999) in the mPFC and alleviate various addictive behaviors, such as drug seeking (Pentkowski et al., 2010) or alcohol withdrawal-induced anxiety (Knapp et al., 2004;Overstreet et al., 2006). In humans, 5-HT-R antagonists (together with 5-HT reuptake inhibitors) are effective in AUD management (Marcinkiewcz et al., 2016) and other addictive disorders (Campbell et al., 2021;Higgins and Fletcher, 2015). ...
... In the present study, we used chronic intermittent ethanol (CIE) vapor inhalation exposure in Sprague-Dawley male rats as an established model of alcohol dependence and investigated the effects of 5-HT on GABA A receptor-mediated spontaneous and miniature inhibitory postsynaptic currents (sIPSCs and mIPSCs, respectively) in layer V pyramidal neurons in ex vivo IL slices from naive, alcohol-dependent (Dep), and alcohol-withdrawn (WD) rats. Given the role of 5-HT 2A and 5-HT 2C receptors in AUD (Abernathy et al., 2010;Campbell et al., 2021) and other addictive behaviors (Cunningham et al., 2013;Fu et al., 2020;Marcinkiewcz et al., 2015) and their high expression in the IL, we tested their role in regulating GABAergic inhibition. We found that WD increased basal presynaptic GABA release. ...
... In the IL, 5-HT exerts its actions mainly via stimulatory 5-HT 2A /5-HT 2C receptors and via inhibitory 5-HT 1A receptors (not tested here) on both pyramidal neurons and inhibitory interneurons (Puig and Gulledge, 2011). The activation of either 5-HT 2A or 5-HT 2C receptors triggers GABA release from local interneurons, thereby enhancing inhibitory tone in the IL (Abi-Saab et al., 1999;Yan, 2002), and their dysregulation is deeply implicated in addictive behaviors (Burstein, 2021;Campbell et al., 2021). We found that 5-HT enhanced spontaneous GABA transmission in naive and Dep rats to a similar extent, but this enhancing effect was impaired in WD rats. ...
Article
The infralimbic cortex (IL) is part of the medial prefrontal cortex (mPFC), exerting top-down control over structures that are critically involved in the development of alcohol use disorder (AUD). Activity of the IL is tightly controlled by γ-aminobutyric acid (GABA) transmission, which is susceptible to chronic alcohol exposure and withdrawal. This inhibitory control is regulated by various neuromodulators, including 5-hydroxytrypta�mine (5-HT; serotonin). We used chronic intermittent ethanol vapor inhalation exposure, a model of AUD, in male Sprague-Dawley rats to induce alcohol dependence (Dep) followed by protracted withdrawal (WD; 2 weeks) and performed ex vivo electrophysiology using whole-cell patch clamp to study GABAergic transmission in layer V of IL pyramidal neurons. We found that WD increased frequencies of spontaneous inhibitory postsynaptic currents (sIPSCs), whereas miniature IPSCs (mIPSCs; recorded in the presence of tetrodotoxin) were unaffected by either Dep or WD. The application of 5-HT (50 μM) increased sIPSC frequencies and amplitudes in naive and Dep rats but reduced sIPSC frequencies in WD rats. Additionally, 5-HT2A receptor antagonist M100907 and 5- HT2C receptor antagonist SB242084 reduced basal GABA release in all groups to a similar extent. The blockage of either 5-HT2A or 5-HT2C receptors in WD rats restored the impaired response to 5-HT, which then resembled responses in naive rats. Our findings expand our understanding of synaptic inhibition in the IL in AUD, indicating that antagonism of 5-HT2A and 5-HT2C receptors may restore GABAergic control over IL pyramidal neurons. Significance statement: Impairment in the serotonergic modulation of GABAergic inhibition in the medial pre�frontal cortex contributes to alcohol use disorder (AUD). We used a well-established rat model of AUD and ex vivo whole-cell patch-clamp electrophysiology to characterize the serotonin modulation of GABAergic transmission in layer V infralimbic (IL) pyramidal neurons in ethanol-naive, ethanol-dependent (Dep), and ethanol-withdrawn (WD) male rats. We found increased basal inhibition following WD from chronic alcohol and altered serotonin modulation. Exogenous serotonin enhanced GABAergic transmission in naive and Dep rats but reduced it in WD rats. 5-HT2A and 5-HT2C receptor blockage in WD rats restored the typical serotonin-mediated enhancement of GABAergic inhibition. Our findings expand our understanding of synaptic inhibition in the infralimbic neurons in AUD.
... The striatum therefore appears to be one of the key loci where Another GPCR touted as a potential therapeutic target for drug and alcohol abuse is the 5-HT 2C receptor; evidence in this regard is reviewed by Ubhayarathna et al. (2024). Indeed, lorcaserin (a 5-HT 2C agonist), which was withdrawn by the FDA, has shown promise in a proof-of concept study in humans with AUD (Campbell et al., 2021). ...
... Despite the abundant literature examining sex differences in punishment sensitivity in humans, this topic is quite understudied in relation to substance use and abuse. This may be partly due to recruitment difficulties and high drop-out rates observed in these populations [90,91]. Additionally, whilst several studies have examined punishment and substance use, they have not examined sex differences [92]. ...
... Furthermore, METH has been reported to interact with the re-uptake of 5-HT, resulting in an acute increase in extracellular level of 5-HT, which, in addition to dopamine, is a key mechanism underlying the drug addiction (37). Moreover, a recent study repor ted that the withdrawal syndrome following the termination of drug addiction is also associated with profound changes in 5-HT activity (38). Leucyl-isoleucine is involved in leucine biosynthesis, and mutations in leucine-rich kinases are the common causes of Parkinson's disease, leading to dopamine neuron loss and motor dysfunction through abnormal increases in neuronal protein synthesis (39). ...
Article
Full-text available
Drug addiction can seriously damage human physical and mental health, while detoxification is a long and difficult process. Although studies have reported changes in the oral microbiome of methamphetamine (METH) users, the role that the microbiome plays in the process of drug addiction is still unknown. This study aims to explore the function of the microbiome based on analysis of the variations in the oral microbiome and metabolome of METH users. We performed the 16S rRNA sequencing analysis based on the oral saliva samples collected from 278 METH users and 105 healthy controls (CTL). In addition, the untargeted metabolomic profiling was conducted based on 220 samples. Compared to the CTL group, alpha diversity was reduced in the group of METH users and the relative abundances of Peptostreptococcus and Gemella were significantly increased, while the relative abundances of Campylobacter and Aggregatibacter were significantly decreased. Variations were also detected in oral metabolic pathways, including enhanced tryptophan metabolism, lysine biosynthesis, purine metabolism, and steroid biosynthesis. Conversely, the metabolic pathways of porphyrin metabolism, glutathione metabolism, and pentose phosphate were significantly reduced. It was speculated that four key microbial taxa, i.e., Peptostreptococcus , Gemella , Campylobacter , and Aggregatibacter , could be involved in the toxicity and addiction mechanisms of METH by affecting the above metabolic pathways. It was found that with the increase of drug use years, the content of tryptamine associated with neuropsychiatric disorders was gradually increased. Our study provides novel insights into exploring the toxic damage and addiction mechanisms underlying the METH addiction. IMPORTANCE It was found that with the increase of drug use years, the content of tryptamine associated with neuropsychiatric disorders gradually increased. The prediction models based on oral microbiome and metabolome could effectively predict the methamphetamine (METH) smoking. Our study provides novel insights into the exploration of the molecular mechanisms regulating the toxic damage and addiction of METH as well as new ideas for early prevention and treatment strategies of METH addiction.
... However, there has been progress in developing several selective 5-HT 2C agonists, which have been investigated as potential therapeutics for the treatment of multiple psychiatric disorders, particularly impulse-related disorders, such as SUDs (Higgins et al., 2013;Campbell et al., 2021). A growing body of evidence supports the influence of 5-HT 2C on impulse regulation, partly through the modulation of dopaminergic neurotransmission in the VTA (Bubar and Cunningham, 2007;Howell and Cunningham, 2015). ...
Article
Serotonergic psychedelics, such as psilocybin and LSD, have garnered significant attention in recent years for their potential therapeutic effects and unique mechanisms of action. These compounds exert their primary effects through activating serotonin 5-HT2A receptors, found predominantly in cortical regions. By interacting with these receptors, serotonergic psychedelics induce alterations in perception, cognition, and emotions, leading to the characteristic psychedelic experience. One of the most crucial aspects of serotonergic psychedelics is their ability to promote neuroplasticity, the formation of new neural connections, and rewire neuronal networks. This neuroplasticity is believed to underlie their therapeutic potential for various mental health conditions, including depression, anxiety, and substance use disorders. In this mini-review, we will discuss how the 5-HT2A receptor activation is just one facet of the complex mechanisms of action of serotonergic psychedelics. They also interact with other serotonin receptor subtypes, such as 5-HT1A and 5-HT2C receptors, and with neurotrophin receptors (e.g., tropomyosin receptor kinase B). These interactions contribute to the complexity of their effects on perception, mood, and cognition. Moreover, as psychedelic research advances, there is an increasing interest in developing nonhallucinogenic derivatives of these drugs to create safer and more targeted medications for psychiatric disorders by removing the hallucinogenic properties while retaining the potential therapeutic benefits. These nonhallucinogenic derivatives would offer patients therapeutic advantages without the intense psychedelic experience, potentially reducing the risks of adverse reactions. Finally, we discuss the potential of psychedelics as substrates for post-translational modification of proteins as part of their mechanism of action.
Article
Full-text available
The illicit use of the psychostimulant methamphetamine (METH) is a major concern, with overdose deaths increasing substantially since the mid-2010s. One challenge to treating METH use disorder (MUD), as with other psychostimulant use disorders, is that there are no available pharmacotherapies that can reduce cravings and help individuals achieve abstinence. The purpose of the current review is to discuss the molecular targets that have been tested in assays measuring the physiological, the cognitive, and the reinforcing effects of METH in both animals and humans. Several drugs show promise as potential pharmacotherapies for MUD when tested in animals, but fail to produce long-term changes in METH use in dependent individuals (eg, modafinil, antipsychotic medications, baclofen). However, these drugs, plus medications like atomoxetine and varenicline, may be better served as treatments to ameliorate the psychotomimetic effects of METH or to reverse METH-induced cognitive deficits. Preclinical studies show that vesicular monoamine transporter 2 inhibitors, metabotropic glutamate receptor ligands, and trace amine-associated receptor agonists are efficacious in attenuating the reinforcing effects of METH; however, clinical studies are needed to determine if these drugs effectively treat MUD. In addition to screening these compounds in individuals with MUD, potential future directions include increased emphasis on sex differences in preclinical studies and utilization of pharmacogenetic approaches to determine if genetic variances are predictive of treatment outcomes. These future directions can help lead to better interventions for treating MUD.
Article
Full-text available
Alcohol Use Disorder (AUD) is a leading cause of death and disability worldwide, but current treatments are insufficient in fully addressing the symptoms that often lead to relapses in alcohol consumption. The brain's serotonin system has been implicated in AUD for decades and is a major regulator of stress-related behaviors associated with increased alcohol consumption. This review will discuss the current literature on the association between neurobiological adaptations in serotonin systems and AUD in humans as well as the effectiveness of serotonin receptor manipulations on alcohol-related behaviors like consumption and withdrawal. We will further discuss how these findings in humans relate to findings in animal models, including a comparison of systemic pharmacological manipulations modulating alcohol consumption. We next provide a detailed overview of brain region-specific roles for serotonin and serotonin receptor signaling in alcohol-related behaviors in preclinical animal models, highlighting the complexity of forming a cohesive model of serotonin function in AUD and providing possible avenues for more effective therapeutic intervention. Throughout the review, we discuss what is known about sex differences in the sequelae of AUD and the role of serotonin in these sequelae. We stress a critical need for additional studies in women and female animals so that we may build a clearer path to elucidating sex-specific serotonergic mechanisms and develop better treatments.
Article
Full-text available
For most psychiatric disorders, including alcohol use disorder (AUD), approved pharmacological treatments are limited in their effectiveness, and new drugs that can easily be translated into the clinic are needed. Currently, great hope lies in the potential of psychedelics to effectively treat AUD. The primary hypothesis is that a single session of psychedelic-guided psychotherapy can restore normal brain function in AUD individuals and thereby reduce the risk of relapse in the long run. Here we applied three different treatment schedules with psilocybin/LSD in order to investigate relapse-like drinking in the alcohol deprivation effect (ADE) model. In contrast to the primary hypothesis, psychedelics had no long-lasting effects on the ADE in male and female rats, neither when administered in a high dosage regime that is comparable to the one used in clinical studies, nor in a chronic microdosing scheme. Only sub-chronic treatment with psilocybin produced a short-lasting anti-relapse effect. However, it is not a translatable treatment option to give psychedelics sub-chronically for relapse prevention. In conclusion, our results in the ADE model do not support the hypothesis that microdosing or high doses of psychedelic reduce relapse behavior. This conclusion has to be confirmed by applying other animal models of AUD. It could also well be that animal models of AUD might be unable to fully capture the therapeutic potential of psychedelic drugs and that only future large-scale clinical trials will be able to demonstrate the efficacy of psychedelics as a new treatment option for AUD.
Article
Background Opioid Use Disorder (OUD) is a significant public health problem associated with severe morbidity and mortality. While effective pharmacotherapies are available, limitations exist with each. Induction onto extended-release naltrexone (XR-NTX) is more difficult than initiation of buprenorphine or methadone, even in inpatient settings, as it is recommended that patients remain abstinent for at least 7 days prior to initiating XR-NTX. The purpose of this trial was to determine if lorcaserin, a 5HT2c agonist, improves outpatient XR-NTX induction rates. Methods An 8-week trial beginning with a brief detoxification period and induction onto XR-NTX. Sixty participants with OUD were enrolled in the trial, with 49 participants at the initiation of detoxification randomized to lorcaserin or placebo for 39 days. Additionally, ancillary medications were provided. The primary outcome was the proportion of participants inducted onto the first XR-NTX injection. Secondary outcomes were withdrawal severity (measured by COWS and SOWS) prior to the first injection and the proportion of participants receiving the second XR-NTX injection. Results The proportion of participants inducted onto the first (lorcaserin: 36 %; placebo: 44 %; p = .67) and the second XR-NTX injection (lorcaserin: 27 %; placebo: 31 %; p = .77) was not significantly different between treatment arms. Prior to the first injection, withdrawal scores did not significantly differ between treatment arms over time (treatment*time interaction COWS: p = .11; SOWS: p = .39). Conclusions Lorcaserin failed to improve outpatient XR-NTX induction rates. Although this study is small, the findings do not support the use of lorcaserin in promoting induction onto XR-NTX or in mitigating withdrawal symptoms.
Article
There are no FDA‐approved treatments for cannabis use disorder (CUD). Preclinical research has shown that the 5HT‐2C agonist lorcaserin attenuates cue‐induced reinstatement of THC seeking and self‐administration. The goal of this placebo‐controlled, counterbalanced, within‐subject human laboratory study was to examine lorcaserin's effects on cannabis intoxication and self‐administration. Lorcaserin (10 mg BID) was administered during one of two 13‐day inpatient phases and placebo during the other; each phase was separated by ≥7 days of washout. Inpatient phases comprised (1) standardized cannabis administration (7.0% THC) at no financial cost (intoxication), counterbalanced with (2) the option to self‐administer cannabis following either 0 or 3 days of abstinence. Cognitive task performance, food intake, subjective ratings of drug effects, objective/subjective sleep measures, and tobacco cigarette use were also assessed. Fifteen normal‐weight, daily cannabis users (4F, 11M) not seeking treatment for CUD completed the study. Lorcaserin significantly reduced cannabis self‐administration following 0 and 3 days of cannabis abstinence and also reduced craving for cannabis during abstinence. Lorcaserin produced small but significant increases in positive cannabis ratings and body weight relative to placebo. Lorcaserin also reduced tobacco cigarette smoking on days of cannabis administration relative to placebo. During abstinence, subjective but not objective measures of sleep quality worsened during lorcaserin maintenance. Overall, lorcaserin's ability to decrease drug taking and cannabis craving in nontreatment‐seeking cannabis users supports further investigation of 5HT‐2C agonists as potential pharmacotherapies for CUD.
Article
Methamphetamine use continues to be an important public health problem. Contingency management is among the most effective interventions for reducing methamphetamine use. It has been more than ten years since the last systematic review of contingency management for methamphetamine use disorder. Since then, an additional ten randomized controlled trials and a variety of other studies have been completed. The present systematic review includes 27 studies. Several factors, most notably problem severity, appear to predict treatment outcome. However, the effectiveness of CM has been demonstrated in studies restricted to MSM, studies restricted to implementation in community programs, and in studies of the general population of methamphetamine users conducted in research treatment programs. There appear to be broad benefits of contingency management intervention, including greater drug abstinence, higher utilization of other treatments and medical services, and reductions in risky sexual behavior. Twenty of the twenty-one studies that reported abstinence outcomes showed an effect of contingency management on abstinence, and seven of the nine studies that reported sexual risk behavior outcomes showed an effect of contingency management in reducing risky sexual behavior. Taken together, recent evidence suggests strongly that outpatient programs that offer treatment for methamphetamine use disorder should prioritize adoption and implementation of contingency management intervention.
Article
Objective To conduct a systematic review of modern‐era (post‐millennium) clinical studies assessing the therapeutic effects of serotonergic psychedelics drugs for mental health conditions. Although the main focus was on efficacy and safety, study characteristics, duration of antidepressants effects across studies, and the role of the subjective drug experiences were also reviewed and presented. Method A systematic literature search (1st Jan 2000 to 1st May 2020) was conducted in Pubmed and Psychinfo for studies of patients undergoing treatment with a serotonergic psychedelic. Results Data from 16 papers, representing 10 independent psychedelic‐assisted therapy trials (psilocybin=7, ayahuasca = 2, LSD=1) were extracted, presented in figures and tables, and narratively synthesized and discussed. Across these studies, a total of 188 patients suffering either anxiety and/or depressive symptoms associated with cancer (C‐RPD), major depressive disorder (MDD), obsessive compulsive disorder (OCD) or substance use disorder (SUD) were included. The reviewed studies established feasibility and evidence of safety, alongside promising early data of efficacy in the treatment of depression, anxiety, OCD, and tobacco and alcohol use disorders. For a majority of patients, the therapeutic effects appeared to be long‐lasting (weeks‐months) after only 1 to 3 treatment session(s). All studies were conducted in line with guidelines for the safe conduct of psychedelic therapy and no severe adverse events were reported. Conclusion The resurrection of clinical psychedelic research provides early evidence for treatment efficacy and safety for a range of psychiatric conditions, and constitutes an exciting new treatment avenue in a health area with major unmet needs.
Article
Drug repositioning has gained strategic value as a reaction to high attrition rates of new drugs as they pass through the clinical development process. The 5-HT2C receptor agonist lorcaserin (Belviq®), and the selective NA reuptake inhibitor atomoxetine (Strattera®) represent two drugs FDA approved for obesity and ADHD respectively. Although both drugs are of differing pharmacological class, each share a property of regulating impulsive behaviours in preclinical studies, and thus represent candidates for consideration in clinical conditions labelled as ‘impulsive-compulsive disorders’. The present studies investigated both drugs, as well as the highly selective 5-HT2C agonist CP-809101 in two tests of compulsive action: schedule-induced polydipsia (SIP) and increased perseverative [PSV] (and premature [PREM]) responses emitted during an extended ITI 5-choice task. While lorcaserin (0.06–0.6 mg/kg), CP-809101 (0.1–1 mg/kg) and atomoxetine (0.1–1 mg/kg) each reduced both PREM and PSV measures in the 5-choice task, at equivalent doses only lorcaserin and CP-809101 affected excessive water intake in the SIP task, atomoxetine (0.1–2 mg/kg) was essentially ineffective. Further evidence supporting a role of the 5-HT2C receptor as an important regulator of impulsive-compulsive behaviours, the selective antagonist SB-242084 produced the opposing effects to lorcaserin, i.e promoting both impulsive and compulsive behaviours. The profile of atomoxetine may suggest differences in the nature of compulsive action measured either as non-regulatory drinking in the SIP task, and PSV responses made in a 5-choice task. These studies support the consideration of 5-HT2C receptor agonists, typified by lorcaserin, and atomoxetine as potential treatments for clinical conditions categorised as ‘impulsive-compulsive disorders’. This article is part of the special issue entitled ‘Serotonin Research: Crossing Scales and Boundaries’.
Article
Background Alcohol use disorders (AUD) are a major socioeconomic burden on society and current pharmacotherapeutic treatment options are inadequate. Aberrant alcohol use and seeking alters frontostriatal function. Methods We performed genome-wide RNA sequencing, and subsequent qPCR and receptor binding validation in the caudate-putamen of human AUD samples to identify potential therapeutic targets. We next back-translated our top candidate targets into a rodent model of long-term alcohol consumption to assess concordance of molecular adaptations in the rat striatum. Finally, we adopted rat behavioural models of alcohol intake and seeking to validate a potential therapeutic target. Results We found G protein-coupled receptors (GPCRs) were the top canonical pathway differentially regulated in individuals with AUD. The M4 muscarinic acetylcholine receptor (mAChR) was down regulated at the gene and protein level in the putamen, but not caudate, of AUD samples. We found concordant downregulation of the M4 mAChR, specifically on dopamine D1 receptor-expressing medium spiny neurons in the rat dorsolateral striatum. Systemic administration of the selective M4 mAChR positive allosteric modulator, VU0467154, reduced home cage and operant alcohol self-administration, motivation to obtain alcohol and cue-induced reinstatement of alcohol seeking in rats. Local microinjections of VU0467154 in the rat dorsolateral striatum reduced alcohol self-administration and cue-induced reinstatement of alcohol seeking. Conclusions Collectively, these results identify the M4 mAChR as a potential therapeutic target for the treatment of AUD and D1 receptor positive medium spiny neurons in the dorsolateral striatum as a key site mediating the actions of M4 mAChR in relation to alcohol consumption and seeking.
Article
Cocaine use disorder (CUD) is a major public health challenge for which there are no pharmacotherapeutics approved by the United States Food and Drug Administration (FDA). The propensity to relapse in CUD involves several vulnerability factors including sensitivity to cues associated with cocaine-taking. Serotonin (5-hydroxytryptamine, 5-HT) neurotransmission, particularly through the 5-HT2A receptor (5-HT2AR) and 5-HT2C receptor (5-HT2CR), is mechanistically linked to cocaine-seeking in preclinical models. In the present experiments, we employed self-administration assays in male rats to investigate whether acute and/or repeated administration of the FDA-approved selective 5-HT2AR antagonist/inverse agonist pimavanserin, selective 5-HT2CR agonist lorcaserin or their combination would alter cocaine intake and/or cocaine-seeking behavior. We found that acute administration of lorcaserin, but not pimavanserin, attenuated cocaine intake while pimavanserin plus lorcaserin did not impact cocaine self-administration. In contrast, 10-days of repeated administration of pimavanserin, lorcaserin, or pimavanserin plus lorcaserin during forced abstinence from cocaine self-administration, blunted cocaine-seeking, similar to the acute administration of each ligand. Taken together, these data reveal the efficacy of repeated treatment with pimavanserin plus lorcaserin to attenuate factors important to relapse-like behaviors in rodent models of CUD. This article is part of the special issue entitled ‘Serotonin Research: Crossing Scales and Boundaries’.
Article
Background: Lorcaserin, a high-affinity 5-HT2C receptor agonist approved for treating obesity, decreased self-administration of oxycodone and cue-induced reinstatement of drug-seeking behavior in preclinical studies. The current investigation is the first clinical trial to evaluate the ability of lorcaserin to alter the reinforcing and subjective effects of oxycodone. Methods: In this 7-week inpatient trial, 12 non-treatment-seeking volunteers (11 males) with moderate-to-severe opioid use disorder were detoxified from opioids. In a randomized cross-over fashion, participants were first stabilized on lorcaserin (10 mg BID) or placebo (0 mg BID). Participants underwent a two-week testing period during which the reinforcing and subjective effects of intranasal oxycodone were examined in verbal choice, cue-exposure, and progressive-ratio choice sessions. The two testing weeks were identical with the exception that during the first week, active oxycodone (10 mg) was available during verbal choice (self-administration) sessions, and during the second week placebo oxycodone was available. Subsequently, participants were stabilized on the other medication condition (placebo or lorcaserin) and underwent the same testing procedures again. Results: Lorcaserin did not alter oxycodone self-administration. However, lorcaserin had a trend to increase "wanting heroin" when oxycodone was available, and to accentuate oxycodone-induced miosis. Conclusion: Under the current experimental conditions, lorcaserin at a dose of 10 mg BID did not reliably decrease the abuse liability of oxycodone, even though the study was sufficiently powered (≥80 %) to detect clinically meaningful differences in the main outcome variables between the placebo and active lorcaserin condition. Future research could explore a wider dose range of lorcaserin and oxycodone.
Article
The Concise Guide to PHARMACOLOGY 2019/20 is the fourth in this series of biennial publications. The Concise Guide provides concise overviews of the key properties of nearly 1800 human drug targets with an emphasis on selective pharmacology (where available), plus links to the open access knowledgebase source of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. Although the Concise Guide represents approximately 400 pages, the material presented is substantially reduced compared to information and links presented on the website. It provides a permanent, citable, point-in-time record that will survive database updates. The full contents of this section can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.14748. G protein-coupled receptors are one of the six major pharmacological targets into which the Guide is divided, with the others being: ion channels, nuclear hormone receptors, catalytic receptors, enzymes and transporters. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. The landscape format of the Concise Guide is designed to facilitate comparison of related targets from material contemporary to mid-2019, and supersedes data presented in the 2017/18, 2015/16 and 2013/14 Concise Guides and previous Guides to Receptors and Channels. It is produced in close conjunction with the International Union of Basic and Clinical Pharmacology Committee on Receptor Nomenclature and Drug Classification (NC-IUPHAR), therefore, providing official IUPHAR classification and nomenclature for human drug targets, where appropriate.