• Sonuç bulunamadı

The impact of organic anion-transporting polypeptides (OATPs) on disposition and toxicity of antitumor drugs: insights from knockout and humanized mice

N/A
N/A
Protected

Academic year: 2021

Share "The impact of organic anion-transporting polypeptides (OATPs) on disposition and toxicity of antitumor drugs: insights from knockout and humanized mice"

Copied!
17
0
0

Yükleniyor.... (view fulltext now)

Tam metin

(1)

ContentslistsavailableatScienceDirect

Drug

Resistance

Updates

j ou rn a l h o m e pa g e :w w w . e l s e v i e r . c o m / l o c a t e / d r u p

The

impact

of

Organic

Anion-Transporting

Polypeptides

(OATPs)

on

disposition

and

toxicity

of

antitumor

drugs:

Insights

from

knockout

and

humanized

mice

Selvi

Durmus

a,1

,

Stéphanie

van

Hoppe

b,1

,

Alfred

H.

Schinkel

b,∗

aBilkentUniversity,DepartmentofMolecularBiologyandGenetics,06800Bilkent,Ankara,Turkey

bTheNetherlandsCancerInstitute,DivisionofMolecularOncology,Plesmanlaan121,1066CXAmsterdam,TheNetherlands

a

r

t

i

c

l

e

i

n

f

o

Articlehistory:

Received6February2016

Receivedinrevisedform7May2016

Accepted17June2016 Keywords: OATP1A/1Btransporters OATP1B1 OATP1B3 Drugdisposition Hepaticuptake Drug–druginteractions

a

b

s

t

r

a

c

t

Itisnowwidelyacceptedthatorganicanion-transportingpolypeptides(OATPs),especiallymembers oftheOATP1A/1Bfamily,canhaveamajorimpactonthedispositionandeliminationofavarietyof endogenousmoleculesanddrugs.Owingtotheirprominentexpressioninthesinusoidalplasma mem-braneofhepatocytes,OATP1B1andOATP1B3playkeyrolesinthehepaticuptakeandplasmaclearance ofamultitudeofstructurallydiverseanti-cancerandotherdrugs.Here,wepresentathorough assess-mentofthecurrentlyavailableOATP1AandOATP1Bknockoutandtransgenicmousemodelsaskey toolstostudyOATPfunctionsinvivo.Wediscussrecentstudiesusingthesemodelsdemonstratingthe importanceofOATPs,primarilyintheplasmaandhepaticclearanceofanticancerdrugssuchastaxanes, irinotecan/SN-38,methotrexate,doxorubicin,andplatinumcompounds.Wefurtherdiscussrecentwork onOATP-mediateddrug–druginteractionsinthesemousemodels,aswellasontheroleofOATP1A/1B proteinsinthephenomenonofhepatocytehopping,anefficientandflexiblewayofliverdetoxification forbothendogenousandexogenoussubstrates.Interestingly,glucuronideconjugatesofboththeheme breakdownproductbilirubinandtheproteintyrosinekinase-targetedanticancerdrugsorafenibare stronglyaffectedbythisprocess.TheclinicalrelevanceofvariationinOATP1A/1Bactivityinpatientshas beenpreviouslyrevealedbytheeffectsofpolymorphicvariantsanddrug–druginteractionsondrug toxic-ity.ThedevelopmentofinvivotoolstostudyOATP1A/1Bfunctionshasgreatlyadvancedourmechanistic understandingoftheirfunctionalroleindrugpharmacokinetics,andtheirimplicationsfortherapeutic efficacyandtoxicsideeffectsofanticancerandotherdrugtreatments.

©2016ElsevierLtd.Allrightsreserved.

1. IntroductiontoOATP1A/1Btransportersandgenetically

modifiedmousemodelstostudytheirfunctions

1.1. PropertiesofOATP1A/1Btransporters

Organicanion-transportingpolypeptide(OATP)uptake trans-porterscanplayamajorroleintheuptakeofnumerouscompounds, includingmanyanticancerdrugs,intocellsandorgans.Positioned

Abbreviations: ABC,ATP-bindingcassette;AUC,areaunderthe

concentration-timecurve;DDI,drug–druginteractions;E2G,estradiol17␤-d-glucuronide;i.v.,

intravenous;OATP,organicanion-transportingpolypeptide;TKI,tyrosinekinase

inhibitor;UGT1A1,UDP-glucuronosyltransferase1A1.

∗ Correspondingauthor.

E-mailaddress:a.schinkel@nki.nl(A.H.Schinkel).

1 Thesetwoauthorscontributedequallytothispaper.

in theplasma membrane, thesemultispanning transmembrane proteinscanmediatetheuptakeofastructurallyhighlydiverse rangeofsubstratesintothecell,byasyetincompletelyresolved mechanisms.Asaconsequence,theycanhaveamajorimpacton thepharmacokineticdispositionoftransporteddrugs,determining theiroralavailabilityandplasmaclearance,aswellastheir distribu-tiontoliverandotherorgans,andthemainroute(s)ofelimination (forrecentreviewssee:GongandKim,2013;Konigetal.,2013; Niemietal., 2011;Shitara etal.,2013; Stiegerand Hagenbuch, 2014).OATPscanthereforehaveastrongeffectonthetherapeutic efficacy,butalsothetoxicsideeffectsofsubstratedrugs.Moreover, severalOATPsarevariablyexpressedinarangeofhumancancers. Asthis mayobviouslyinfluencetheeffectiveintracellular expo-sureofthecancercellstoOATPsubstrateanticancerdrugs,this candirectlyaffectthetherapysusceptibilityofthesecancers(for recentreviewssee:NakanishiandTamai,2014;Obaidatetal.,2012; Sissungetal.,2012;Thakkaretal.,2015).Theactivityofthehuman http://dx.doi.org/10.1016/j.drup.2016.06.005

(2)

OATPsthatarethoughttobemostimportantforthegeneral phar-macokineticbehaviorofdrugs,OATP1A2,OATP1B1,andOATP1B3 (aswellaspossiblyOATP2B1,butseebelow),canfurthervary dra-maticallybecauseofgeneticpolymorphismsandmutationsthat affectdrugtransport,butalsobecauseofdrug–druginteractions withavarietyofco-administereddrugs(e.g.Durmusetal.,2015; Frankeetal.,2009;GongandKim,2013;Konigetal.,2013;Niemi etal.,2011;Obaidatetal.,2012;Shitaraetal.,2013;Stiegerand Hagenbuch,2014;vandeSteegetal.,2012).

Giventheirobviousmedicalimportance,itiscrucialtoobtain clearinsightintotheinvivopharmacological,toxicological,and physiologicalfunctionsoftheOATPproteins,especiallythoseofthe OATP1A/1Bfamily.Onewaytoachievethisgoalistogenerateand studymousestrainsthathavethemouseOatp1aandOatp1bgenes knockedout,orthathavereplacedthemouseOatp1a/1bgeneswith oneormoreoftheirhumananalogues(althoughtheformalgene namefortheOATP-encodinggenesisSLCO(forSoluteCarrierof OrganicAnions),forsimplicitywewillmostlyusetheOATP/Oatp nomenclatureinthis review).Thesemouse modelscanthenbe usedtoinvestigatetheimpactofthegeneticmodificationsonthe behaviorof,amongstothers,anticancerdrugs.Thisreviewfocuses onrecentstudiesonsuchmousestrainsandtheinsightsobtained foranumberofanticancerdrugs.Assomeaspectshavealready beenextensivelyreviewedpreviously(Iusufetal.,2012b,c;Sprowl andSparreboom,2014;Tangetal.,2013),wewillonlybrieflytouch uponthose.

1.2. OATP1A/1Bknockoutandtransgenicmousestrains characterizedtodate

Todate, mostcharacterizedknockoutand transgenicmouse modelsconcernmembersoftheOATP1AandOATP1Bsubfamilies, asthesearethoughttobemostrelevantforoverall pharmacoki-neticsinman.SomeinitialstudiessuggestedthathumanOATP1A2 wasexpressedintheintestinalepithelium,whichwould poten-tiallyindicateanimportantroleindrugabsorption(Glaeseretal., 2007).However,manyindependentlaterstudiescouldnot cor-roboratethesefindings,anditisnowprobablysafetoconclude thatnormallythereisnosubstantiallevelofOATP1A2presentin thesmallorlargeintestineofhumans(e.g.Drozdziketal.,2014). Oncurrentdata,OATP1A2issubstantiallyexpressedin cholangio-cytesliningthebileductsintheliver,inthehumanblood-brain barrier, in apical membranes of kidney tubules, and in a vari-etyof humantumors(vandeSteeg etal., 2013and references therein).IncontrasttoOATP1A2,humanOATP1B1andOATP1B3 arehighlyandprimarilyexpressedinthebasolateral(sinusoidal) membraneofhumanhepatocytes,wheretheycanmediatethe hep-aticuptakeofnumeroussubstratecompounds(e.g.Nakanishiand Tamai,2012).Asthesegenesarealsoknowntobesubstantially affectedbygeneticpolymorphismsandmutationsinhumans(e.g. Niemietal.,2011;vandeSteegetal.,2012),theyhaveattracted mostattention.ThefunctionallyrelatedOATP2B1proteinisalsoa broad-specificitymultidrug-uptaketransporter,especiallyatlower pH, and highly expressedin both intestine and the sinusoidal membraneofhepatocytes.Ithasthereforebeensuggestedthatit mightalsohaveconsiderablepharmacokineticimpact(Nakanishi andTamai,2012).However,todatenoOATP2B1mousemodels havebeenpublished,sowewillnotfurthercoverthistransporter here.

AcomplicationinstudyingmousemodelsfortheOATP1A/1B transporters is that there are no straightforward orthologues between the individual mouse and human Oatp1a/1b and OATP1A/1Bgenes.AsindicatedinFig.1,therearenolessthan4 differentOatp1agenesinthemouse,Oatp1a1,Oatp1a4,Oatp1a5 andOatp1a6,inadditionto2Oatp1a-likeelementsthatmaybe pseudo-genes. Thiscompareswiththesingle OATP1A2gene in

Table1

Oatp1a/bknockoutandtransgenicmousemodelsdescribedtodate.

Mousemodels Geneticbackground Primaryreferences

Oatp1a1knockout C57Bl/6N (Gongetal.,2011)

Oatp1a4knockout C57Bl/6N (Gongetal.,2011;

Oseetal.,2010)

Oatp1b2knockout(variant1) C57BL/6 (Luetal.,2008)

Oatp1b2knockout(variant2) DBA1/lacJ (Zaheretal.,2008)

Oatp1a/1bknockout FVB (vandeSteegetal.,

2010)

HumanizedhepaticOATP1A2

transgenic

FVB (vandeSteegetal., 2013)

HumanizedhepaticOATP1B1

transgenic

FVB (vandeSteegetal., 2009)

HumanizedhepaticOATP1B3

transgenic

FVB (vandeSteegetal., 2013)

HumanizedhepaticOATP1B1

andOATP1B3transgenic

FVB (Salphatietal., 2014)

humans.Ontheotherhand,themousehasonlyoneOatp1b2gene, contrastingwith thetwo humanOATP1B1and OATP1B3genes (Fig.1).Although the mouse and humanOATP1A proteinsare obviouslymoresimilartoeachotherthantothemouseandhuman OATP1Bproteins,andviceversa,theaminoaciddivergencewithin eachsubfamilyisstillconsiderable(aslowas67%aminoacid iden-titywithin theOATP1A subfamily, and 65%withinthe OATP1B subfamily).Consequently,withthesebroad-specificitymultidrug transporters,noreliablestatementscanbemadeonoverlapping substratesjustbasedonaminoacidsimilarity.Asthetissue distri-butionisalsonotconservedbetweenmembersofonesubfamily (for instance, mouse Oatp1a1 and Oatp1a4 are present in the sinusoidalmembraneofhepatocytes,whereashumanOATP1A2is not)itisclearthatonecannotusesingle-genemouseOatp1aor Oatp1bknockoutstrainstomakereliablepredictionsontheinvivo behaviorofhumanOATP1A2,OATP1B1,orOATP1B3.Thiswasan importantmotivationtogenerateacompleteOatp1a/1bknockout strain,anduseittospecificallyexpresshumanOATP1A2,OATP1B1, andOATP1B3inthisknockoutbackground(vandeSteegetal.,2012, 2013,2010).

Table1liststhevariousOatp1aandOatp1bknockoutstrains, and OATP1A/1Bhumanized strainsthat havebeendescribed so far.Oatp1a1andOatp1a4knockoutstrainsweredescribedbyOse etal.(2010)andGonget al.(2011).Thesemouse strains, origi-nallymadein129/OlaEScellsbyDeltagen,werebackcrossed10 timestoaC57BL/6background,andsubsequentcharacterization oftheselineswasmainlydoneinthisgeneticbackground.Afew independentOatb1b2knockoutstrainsweregenerated.Luetal. (2008)describedthegenerationandinitialcharacterizationofan Oatp1b2knockoutstraingeneratedin129S1EScells,whichwas backcrossedfor7generationstoaC57BL/6background. Indepen-dently,Zaheretal.(2008)generatedOatp1b2knockoutmiceusing DBA1/lacJEScells,whichwerefurtherkeptandcharacterizedin a DBA1/lacJgeneticbackground.Combined Oatp1a/1bknockout mice,coveringallthemouseOatp1aandOatp1bgenes,were gen-eratedin129/OlaEScells.InitialcharacterizationofOatp1a/1b−/− micewasdoneinamixed(∼50%)129/OlaandFVBgenetic back-ground(vandeSteegetal.,2010),butthisstrainwassubsequently backcrossedforatleast7generationstoanFVBbackground,in which furthercharacterization took place (van de Steeg et al., 2012).TheseFVBbackgroundOatp1a/1b−/−micewerethenused togeneratethreedifferenthumanizedmousestrainswith predom-inantexpressionofhumanOATP1A2,OATP1B1,orOATP1B3cDNA, respectively,intheliverparenchymecells,againallinFVB back-ground(vandeSteegetal.,2012,2013).Moreover,acombined humanizedOATP1B1/OATP1B3strainwascreatedbycrossingthe separatetransgenicstrains(Salphatietal.,2014).

(3)

Fig.1. ComparisonofthehumanandmouseOATP1A/B/Cgeneclusters.ThehumanOATP1A/B/Cgeneclusterislocatedonchromosome12p.OATP1C1isfollowedby

OATP1B3,theOATP1B-likepseudogeneLST-3(OATP1B7accordingtoEnsembl),OATP1B1,andOATP1A2.OATP1A2istranscribedfromtheoppositestrandcomparedtothe

OATP1B-andOATP1C-typegenesinthecluster.ThemouseOatp1a/b/cgenesarelocatedonchromosome6pinasimilarorderasthehumangenes,startingwithOatp1c1

followedbyOatp1b2,Oatp1a-likepseudogeneA(Ensembl:Gm5724),Oatp1a4,Oatp1a1,Oatp1a-likepseudogeneB(Ensembl:Gm6614),Oatp1a6,andOatp1a5.Analogous

tothehumangenecluster,allOatp1a-typegenes(andpseudogenes)areorientedintheoppositetranscriptiondirectionoftheOatp1b-andOatp1c-typegenes.Therelative

sizesanddistancesofthegenesaredepictedroughlytoscale,alsobetweenthemouseandhumanchromosome,butwithoutshowingexon/intronstructures,andarrows

indicatethedirectionoftranscription.Pseudogenesandtheirputativedirectionoftranscriptionarerenderedhatched.Directionsofcentromeresandtelomeresandthe

approximatesizeofthehumanandmousegeneclustersareindicated.

1.3. CaveatsinusingOATP1A/1Bgeneticallymodifiedmouse models

Itshouldbenotedthatthegeneticbackgroundstrainmayat timesbeimportantfor theextenttowhich certainphenotypes are detectablein knockoutand transgenic mice. Although this hasnotbeensystematicallyanalyzedsofarinthevarious Oatp knockoutstrains,wehaveobservedmodestphysiologicalchanges inOatp1a/1b−/− miceinmixedOla/129/FVBbackground,which disappearedupon furtherbackcrossinginto anFVBbackground (unpublisheddata).

Alsotheenvironmentalconditions(e.g.,diet,bedding,intestinal microflora) may at times be important factors in the pene-tranceofcertainphenotypes.Forinstance,wehaveobservedthat FVBbackgroundOatp1a/1b−/− miceanalyzedinourfacility(The NetherlandsCancerInstitute,NKI)hadamarkedinductionof car-boxylesterasegenesrelativetowild-typeFVBmice(Iusuf etal., 2014).However,theverysamestrainofmiceobtainedbyother groups through a commercial supplier (Taconic) did not show adifferenceincarboxylesteraseexpressionwithwild-typemice (Salphati et al.,2014).Further analysissuggested thatthis was becausethewild-typeFVBmiceobtainedfromTaconicalreadyhad amuch higher endogenoushepatic carboxylesteraseexpression comparedtoFVBmicekeptattheNKI,mostlikelybecauseof envi-ronmental differences.The Oatp1a/1b knockoutfrom “Taconic” circumstancesdidnot furtherincrease carboxylesterase expres-sion.Interestingly,humanizingtheOatp1a/1bknockoutmicewith the OATP1B1 transgene did reverse the high carboxylesterase expression in both NKI and Taconic mice (Iusuf et al., 2014; Salphatietal.,2014),indicatingthatthiscouldapparentlyoverrule anyenvironmentalfactorscausingthedifferentcarboxylesterase expressioninFVBwild-typemice.Ontheotherhand,transgenic OATP1B3couldfullyreversethecarboxylesteraseoverexpression

intheNKIfacility,butnotelsewhere.Thisfurthersupportsthat, whateverthefactor(s)pushingupcarboxylesteraseexpressionin theOatp1a/1b−/−(andFVB)mice,theyweremoreprominentinthe otherfacilitiesthanintheNKIfacility.Itisimportanttokeepsuch complicationsinmindwhencomparingtheresultsingenetically modifiedmousestrainsobtainedbydifferentgroups.Moreover,as environmentalconditionsinmousefacilitiescannotalwaysbekept undercompletecontrol,itisalsopossiblethatshiftsinphenotypes occurovertimeevenwithinonefacility.

Thirdly,alsogendermaysubstantially affectresultsobtained withOatp1a/1bgeneticallymodifiedmice.Forinstance,Oatp1a1, Oatp1a4,andOatp1b2areallsubstantiallyexpressedinthe baso-lateral(sinusoidal)membraneofthemouseliver.JudgedbyRNA level,Oatp1a1ismoreabundantinmalethaninfemaleliver(about 2-fold),andOatp1a4infemaleliver(alsoabout2-fold),whereas Oatp1b2issimilarlyexpressedinbothgenders(Chengetal.,2005). Thiscandirectlyaffectwhethercertainpharmacokineticeffectscan bedetectedinknockoutstrains. Forinstance,Gongetal.(2011) foundthatOatp1a1deficiencysubstantiallyreducedtheclearance ofthediagnosticdyedibromosulfophthaleininmalemice,butnot infemalemice.

Insomecases,authorshaveassertedthattheknockoutofmouse Oatp1b2,asthesinglemouserepresentativeofthemammalian OATP1Bsubfamily(Fig.1),mightbemoreorlessequivalenttothe deficiencyofOATP1B1andOATP1B3inhumans(e.g.,Zaheretal., 2008).However,formanyOATPsubstratesthisassumptionclearly doesnotapply.Inhumans,OATP1B1andOATP1B3aretheonly membersoftheOATP1A/1Bfamilythatarepresentinthe basolat-eralmembraneofliverparenchymecells,andthusinvolvedinthe uptakeofsubstratesfrombloodintotheliver.HumanOATP1A2is alsoexpressedintheliver,butonlyfoundinthecholangiocytes, andthereforeirrelevantforthedirectuptakeofcompoundsfrom bloodintotheliver.Incontrast,inthemousetherearetwoOATP1A

(4)

proteinsthataresubstantiallypresentinthesinusoidalmembrane ofliverparenchymecells,Oatp1a1andOatp1a4.Thismeansthat foranysubstratethatissubstantiallytransportedbybothOatp1b2 andeitherorbothofOatp1a1orOatp1a4,resultsobtainedin sin-gleOatp1b2−/− micemayunderestimatetheeffectsofOATP1B1 and OATP1B3deficiency inhumans. Acase in point is therole ofOATP1B1andOATP1B3intheuptakeofconjugatedbilirubin. WhereassingleOatp1b2−/−miceshowedonlyamarginalincrease inplasmatotalandconjugatedbilirubinlevels(Luetal.,2008;Zaher etal.,2008),infullOatp1a/1b−/− micetherewasapronounced increaseinplasmabilirubinglucuronidelevels(vandeSteegetal., 2010).Thelatterobservationpointedtotheprominentrolethat humanOATP1B1andOATP1B3playinthereuptakeofconjugated bilirubininhumanliver,andprovidedthemechanistic explana-tionforthehumanRotorsyndromecausedbyfullOATP1B1and OATP1B3deficiency(vandeSteegetal.,2012).Clearly,the redun-dantroleofOatp1a1andOatp1a4relativetoOatp1b2inhepatic uptakeofconjugatedbilirubininthemouseobscuredthe impor-tantcontributionofthehumanOATP1Bproteinsinthisprocess. Greatcaremustthereforebetakenwhenextrapolatingfromresults inOatp1b2−/−micetothefunctionalroleofthehumanOATP1B1 andOATP1B3proteinsinliver.Forpharmacokineticstudies,given the very substantial but incomplete and unpredictable overlap in substratespecificity betweenthevarious mouse and human OATP1A/1Btransporters,everydrugshouldbecarefullyassessed initsownright,andgreatcautionshouldbeusedin extrapolat-ingfromresultsobtainedinsingleknockoutstrainstothehuman situation.

Keepingthesecaveatsinmind,inthisreviewwewillfocuson recentstudiesperformedwiththesemouse modelsfora range ofanticancerdrugs.Fig.2providesanoverviewofthestructures ofthesedrugs,alsoillustratingtheirstructuraldiversity.Theaim ofsuchstudieswastoobtainbasicinsightintothehandlingof the anticancer drugs by OATP1A/1B proteins, in the hope that thisknowledgemayultimatelybeusedtoimprovecurrent anti-cancerdrugtreatmentregimens,byenhancingtherapeuticefficacy ofthesedrugs,reducingtoxicsideeffects,andpossiblyboth.

2. RecentpharmacologicalandtoxicitystudieswithOATP

knockoutandhumanizedmice

2.1. Taxanes

Thetaxanespaclitaxelanddocetaxelarecytotoxicanticancer drugsthatbindtomicrotubulesanddisrupttheirfunctionby stabi-lizingGDP-boundtubulin,thusinterferingwithpropercelldivision. Paclitaxelanddocetaxelarecurrentlyappliedintravenously(i.v.) inthetreatmentofseveraltypesofcancer,suchasnon-smallcell lungcancer,ovarian,breast,gastric,prostate,andhead-and-neck cancer(GligorovandLotz,2004;Koolenetal.,2010).Both pacli-taxelanddocetaxelarequitelarge,veryhydrophobic,uncharged molecules(Fig.2).Itwasthereforethoughtthattheycouldpass cellmembranesprimarilybypassivediffusion,soitwasinitiallya bitofasurprisethatthesemoleculesweresubstantiallytakenup intocellsbyOATP1B1andOATP1B3inseveral(butnotall,seealso Section4)invitroexpressionsystems(Bakeretal.,2009;deGraan etal.,2012;Nieuweboeretal.,2014;Smithetal.,2006;Svoboda etal.,2011).Toassessthepossibleinvivorelevanceofthisuptake transport,anumberofstudieswithpaclitaxelanddocetaxelwere performedinOATP/Oatpknockoutandtransgenicmice.

2.1.1. Paclitaxel

VandeSteegetal.(2011)foundthatthepaclitaxelplasmaAUC wasmorethan2-foldincreasedinOatp1a/1b−/−relativeto wild-type miceafter i.v.administration at 10mg/kg. Conversely,the

liverAUCwas2-foldlowerintheOatp1a/1b−/−mice.Clear dif-ferencesinplasma andliverpaclitaxelconcentrationswerenot yetapparentat3.5min,butemergedfrom7.5minafter admin-istration.Thissuggeststhattheveryearlypaclitaxeldistribution, whenplasmaconcentrationswereveryhigh(>20mg/l),wasnot muchdependentonOatps.However,withplasmaconcentrations below20mg/l,paclitaxelliveruptakeandhenceplasmaclearance becamestronglydependentonOatp1a/1btransporters.This sug-gestedthatatplasmalevelsabove20mg/lthesinusoidalOatp1a/1b proteinsweresaturated,andotheruptakeprocesses,perhaps pas-sivediffusion,dominatedtheliveruptakeofpaclitaxel.Thiswas supportedby limited paclitaxeldistributionstudies ata higher dosage (50mg/kg), which yielded reduced differences between Oatp1a/1b−/− andwild-typemicein plasmaconcentration (1.7-fold) and liver concentration (1.5-fold) 30min after paclitaxel administrationcomparedtothe10mg/kgdose(1.9-foldand 2.2.-fold,respectively).Overall,thedataindicateasubstantialroleof hepatic sinusoidalOatp1a/1bproteinsintheclearance of pacli-taxelfromplasma,atplasmaconcentrationsthatcanalsooccur in patients. Followinghigh-dose paclitaxelchemotherapy, peak plasma concentrations range from 2 to 11mg/l (Rowinsky and Donehower, 1995), i.e. well below the saturation level of the Oatp1a/1b proteins. A later, independent study byNieuweboer etal.(2014)foundquantitativelysimilareffectsofasingleOatp1b2 knockoutontheplasmaAUCandliveraccumulationofi.v. pacli-taxeldosedat5mg/kginmice.Thiscouldindicatethatmostof theeffectsonpaclitaxelobservedintheOatp1a/1b−/−micewere primarilymediatedbyOatp1b2deficiency,althoughdifferencesin geneticbackgroundstrain(DBA/1lacJvs.FVB),exactpaclitaxel for-mulationused,laboratoryandevenperiodofexperimentation,may allaffectthedetailedoutcome ofsuchstudies(e.g.Nieuweboer etal.,2014;SparreboomandMathijssen,2014).

Ina follow-upstudy,thepharmacokineticsofpaclitaxelwas analyzed in “humanized” Oatp1a/1b−/− mice with transgenic expression of human OATP1B1, OATP1B3, or OATP1A2 in the liver parenchyme cells (van de Steeg et al., 2013). The trans-genicpromoter/enhancerusedwaschosentoobtainpreferential expressionofthehumanproteinsinliverparenchymecells,which wassuccessful,althoughminorOATP1B1andOATP1B3transgene expressionwasalsofoundinkidney,butnotsmallintestine.Based onproteinimmunoblotanalysisandproteinmassspectrometry, thehepaticlevel oftransgenicOATP1B1wasinthesameorder asthatobservedinhumanliversamples,andthatoftransgenic OATP1B3 somewhat higher, with estimated humanized/human ratiosof0.5- to1-foldand ∼3-foldfor OATP1B1and OATP1B3, respectively(Higginsetal.,2014;Salphatietal.,2014).Transgenic OATP1A2expressionwasmuchhigherthaninhumanliver,but thisrelatesmostlytothefactthatOATP1A2inhumanliverisonly expressedincholangiocytes,whereasinthetransgenicmiceitis expressedinthefarmoreabundantliverparenchymecells.This alsomeansthattheOATP1A2humanizedmousestraindoesnot representaphysiologicallycorrectmodelofthenormalOATP1A2 functionin humanliver.However,it doesallowanassessment of the in vivo functioning of OATP1A2in uptake of drugs and othercompoundsfromplasma.Thismay,interalia,berelevant asOATP1A2issubstantiallyexpressedinthehumanblood-brain barrier,inapicalmembranesofkidneytubules,andinavarietyof humantumors(vandeSteegetal.,2013andreferencestherein). Immunohistochemically, transgenic OATP1B1 was foundin the sinusoidalmembraneofhepatocytesandexpressedthroughoutthe liverlobule,albeitwithstrongerstainingaroundtheportalveinin humanliver,whereasthistransporterisvariouslyreportedtobe expressedthroughouttheliverlobule,orprimarilyincentrilobular hepatocytes(vandeSteegetal.,2009,2013).TransgenicOATP1B3 waslikewisefoundinthehepatocytesinusoidalmembrane, show-ingsomewhatdisperseddistributionthroughouttheliverlobules

(5)

Fig.2. StructuresofanticancerdrugsclearlyaffectedbyOatp1a/1bactivityinvivo.Drugsthatarestructurallycloselyrelated,butthatarenotappreciablyaffectedby

Oatp1a/1b-mediatedtransportinvivoarenamedinitalicsandmarkedwithanasterisk.Whilepaclitaxelanddocetaxelareclearlytransported,cabazitaxel,whichdiffersin

justtwomethoxygroupsfromdocetaxel,isnotappreciablytransportedbyOatp1a/1binvivo.Oatp1a/1bactivityinmiceclearlyaffectsdispositionofbothirinotecanand

itsactivemetaboliteSN-38.SorafenibitselfisnotnoticeablytransportedbyOatp1a/1binvivo,butitsglucuronateconjugatesorafenib-␤-D-glucuronideis.Forcisplatinit

shouldbenotedthatthisdrugishighlyreactive,andonlytotalPtlevelsaremeasuredininvivoexperiments.Itisthereforepossiblethatstructurallydifferentcomplexes

(6)

(vandeSteegetal.,2013).InhumanliverOATP1B3ispreferentially foundinthesinusoidalmembranesofcentrilobularhepatocytes. AlthoughthetransgenicOATP1B1andOATP1B3thereforedonot exactlyreflectthelobularsubdistributionofOATPsinhumanliver, subsequentstudiesrevealedgoodfunctionalityoftheseproteins (aswellastransgenicOATP1A2)intheliveruptakeandplasma clearanceofarangeofcompounds.

Strong support for the functional activity of the hepatic OATP1B1,-1B3and -1A2transportersinthehumanized strains camefromthereversalofthehighlyincreasedplasmabilirubin levelsfoundintheOatp1a/1b−/−micerelativetowild-typemice (vandeSteegetal.,2013).Theincreasedplasmalevelsof biliru-binmonoglucuronideanddiglucuronidewerereversedtonearly wild-typelevels(>15-foldand>7-foldreduction,respectively)by bothOATP1B1andOATP1B3expression,whereasOATP1A2caused amoremodest2-foldreduction.Interestingly,transgenicOATP1A2 wastheonlytransporterthatcouldsignificantlyreducethe2-fold increasedlevelsofunconjugatedbilirubinbacktowild-typelevels. ThissuggeststhatOATP1A2is arelativelymore efficient trans-porterofunconjugatedbilirubincomparedtoconjugatedbilirubin, whereastheinverseistrueforOATP1B1andOATP1B3.Whatthis meansforthenormalbiologicalfunctionofhumanOATP1A2in cholangiocytes is as yet unclear.Perhaps it plays a role in the resorptionofhighlyinsolubleunconjugatedbilirubininadvertently formedinbile,thusreducingthechanceofformationof bilirubin-containinggallstones,butforthemomentthispossibilityremains speculative.

A limited pharmacokinetic study of paclitaxel in the OATP humanizedmicerevealedamodest,∼1.6-fold,buthighly signifi-canteffectofOATP1B3andOATP1A2inenhancingtheliveruptake ofpaclitaxeldosedi.v.at2mg/kgcomparedtothatinOatp1a/1b knockoutmice.Asmallereffectwasobservedat10mg/kgi.v. pacli-taxel(onlysignificantforOATP1A2).TransgenicOATP1B1didnot elicitsignificantchangesrelativetoOatp1a/1b−/− miceateither dose(vandeSteegetal.,2013).Collectively,thesedatasuggestthat OATP1B3andperhapsOATP1B1mayonlyhavemodesteffectson paclitaxelliveruptakeandclearanceinhumans.Itshouldbekeptin mind,though,that,unlikethesituationinsinglehumanizedmice, inhumanliverOATP1B1andOATP1B3usuallyfunction simulta-neously,whichcouldwellenhancetheiroverallpharmacokinetic impact by additiveeffects. Such an additiveeffect was indeed observed for another drug in recently generated OATP1B1/1B3 double-transgenicmice,wheresingleOATP1B1orOATP1B3 trans-genesdidnot markedlyreducetheAUCofpravastatinafteri.v. administration,butthecombinationreversedtheAUCtocloseto thatseeninwild-typemice(Salphatietal.,2014).

2.1.2. Docetaxel

Althoughquantitativelydivergentresultshavebeenobtained for the impact of Oatp1b2 and Oatp1a/1b knockouts, and of various OATP1A2/1B1/1B3 humanized transgenes on docetaxel pharmacokinetics, all studies to date support a role of these OATP/Oatpsindocetaxelclearance.DeGraanetal.(2012)reported that after i.v. administration of docetaxel at 10mg/kg to wild-typeandOatp1b2−/−mice,theplasmaAUCwas26.3-foldhigher in Oatp1b2−/− mice, indicating a very large effect of Oatp1b2 removal.Asexpected,theliver-to-plasmaAUCratiowasreduced inOatp1b2−/−mice,by6.2-fold.However,somewhatsurprisingly, theliverdocetaxelAUCitselfwassubstantiallyhigher(4.3-fold)in Oatp1b2−/−comparedtowild-typemice,ratherthanlower(or per-hapsequal,seebelow).Thisresultwasunexpectedincasetheliver uptakeofdocetaxelwasstronglyreducedintheknockoutmice. Ananalysisofpotentialcompensatoryexpressionchangesof func-tionallyrelatedgenesintheknockoutmicedidnotyieldobvious alternativecausesofthechangesindocetaxelpharmacokinetics.A simplemechanisticexplanationoftheobservedincreasedliverAUC

ofdocetaxelintheOatp1b2−/−miceisforthemomenttherefore stilllacking.

Thepotentialexperimentalvariabilityofthesedocetaxel phar-macokineticstudieswasillustratedbytheoutcomeofverysimilar experimentsthatwereperformedafewyearslaterbythesame group,inthesamemousestrains,underapparentlythesame con-ditions(Huetal.,2014;SparreboomandMathijssen,2014).Instead ofa 26-foldhigherplasma AUCof docetaxelintheOatp1b2−/− mice,onlya1.6-foldincreasewasobserved,whichwas,however, stillstatisticallysignificant.Themaindifferencewasnotachange overtimeintheplasmaAUCoftheOatp1b2−/−mice(∼7400vs. ∼8800ngxh/ml),butalmostentirelyattributabletoamuchhigher plasmaAUCofdocetaxelinthewild-typemicecomparedtothe ear-lierexperiments(4500vs.336ngxh/ml).Despiteextensivelater analysesbythesamegroup,thecauseofthesedivergentresultsis stillunclear(SparreboomandMathijssen,2014).Althoughinour experienceitisnotuncommontoseesomevariationinabsolute druglevels(AUC)measuredinpharmacokineticstudiesperformed afewyearsapartunderotherwiseseeminglyidenticalconditions, thisusuallyconcernslessthanabout2-folddifferences.However thismaybe,collectively,thedatastillindicatearoleforOatp1b2in theclearanceofi.v.docetaxel,mostlikelybymediatinguptakeof docetaxelfrombloodintotheliver.Thiswasfurthercorroborated byindependentstudiesbytwoothergroups.

UsingthesamestrainofOatp1b2−/−mice,Leeetal.(2015)very recentlyreporteda limited,30minpharmacokineticstudywith i.v.[3H]-docetaxeldosedat1mg/kg.Bydeterminingplasmaand liverradioactivity,theyfounda5.5-foldhigherplasmaAUCof[3 H]-docetaxelequivalentsinOatp1b2−/−mice(340±149vs62±8ngx h/ml,P<0.05),nosignificantdifferenceinliverconcentrations,but a3-folddecreasedliver-to-plasmaratio(P<0.05).While concern-ingtotalradioactivitymeasurements,whichmaybecomplicated byextensiveandpossiblydifferentialdocetaxelmetabolism,these resultsqualitativelysupportthefindingsofHuetal.(2014)andDe Graanetal.(2012),indicatingaroleofOatp1b2inplasmaclearance ofdocetaxelbymediatinguptakeintotheliver.

Iusuf et al. (2015) studied docetaxel pharmacokinetics in Oatp1a/1b−/− miceof anFVB geneticbackground,usinga low-polysorbate80formulationtominimizepossibleinhibitoryeffects onOatps(deGraanetal.,2012; Nieuweboeret al.,2014).After i.v.administrationofdocetaxelat10mg/kg,theplasmaAUCwas 2.9-foldhigherintheOatp1a/1b−/−mice(609vs212␮gxmin/ml, P<0.001).SimilartothefindingsofLeeetal.(2015),theliver expo-surewasnotsubstantiallyalteredintheOatp1a/1b−/−mice,butthe liver-to-plasmaratiowasmarkedlyloweratallexcepttheearliest timepoints(atleast3-foldormoreafter15min).Alimited pharma-cokineticstudy(15–60min)ofi.v.docetaxel(dosedat10mg/kg) inOATP1B1,OATP1B3,andOATP1A2humanizedmiceindicated a4-foldincreaseindocetaxelplasmaAUCinOatp1a/1b−/− com-pared to wild-type mice, which was largely reversed in both OATP1B1- and OATP1A2-transgenic mice (P<0.001), and more modestly(P<0.01)inOATP1B3-transgenicmice(Iusufetal.,2015) (Fig.3AandB).InaccordancewiththeunalteredliverAUCinthe Oatp1a/1b−/− mice,theliverAUCs inallthehumanized strains werenotsignificantlydifferentfromthoseinthewild-typeand Oatp1a/1b−/−mice.

TheseunalteredliverAUCdataarereadilyexplainedbya phys-iologicallybasedpharmacokineticmodeldevelopedbyWatanabe etal.,2009,2010).Thisshowsthatastrongreductioninhepatic uptake of drugs that have little alternative extrahepatic clear-ance(e.g.,renalclearance),willoftenresultinmarkedlyincreased plasma levels of the drug, but only very small changes in the liverAUC.Accordingly, liver-to-plasmaconcentrationratioswill decrease,butmainlyduetotheincreasedplasmaconcentrations ofthedrug.Thisisexactlythebehaviorthatwasobservedfor doce-taxelinthestudiesofIusufetal.(2015)andLeeetal.(2015),butalso

(7)

Fig.3.ImpactofmouseandhumanOATP1A/1Btransportersontheplasmaandliverdispositionofvariousanti-cancerdrugs.Plasmadisposition(A,C,andE)and

liver-to-plasmaratios(B,D,andF)ofdocetaxel,SN-38anddoxorubicinareshowninthefigureatvarioustimepointsafteri.v.administrationof10mg/kgdocetaxel,10mg/kg

irinotecan(pro-drugofSN-38)or5mg/kgofdoxorubicin,respectively.Drugswereadministeredtowild-typeandOatp1a/1b−/−mice,andtoOATP1B1-,OATP1B3-,and

OATP1A2-transgenicmiceinanOatp1a/1b−/−background.Dataarepresentedasmean±SD[*P<0.05;**P<0.01;***P<0.001whencomparedwithwild-type;#P<0.05;

##P<0.01;###P<0.001whencomparedwithOatp1a/1b−/−mice].Notethatdifferentunitsareusedtoindicatetheplasmaconcentrationsofeachdrug.Thisfigurewas

modifiedwithpermissionfrompreviouslypublishedexperimentaldataofIusufetal.(2015,2014)andDurmusetal.(2014).

earlierforrosuvastatin,anotherOATPsubstrate(Iusufetal.,2013). OnlytheearlierdataofDeGraanetal.(2012),whichappearedto showa>4-foldincreaseinliverAUCofdocetaxelinOatp1b2−/− micearenotyetadequatelyexplained.

Nonetheless,thecollectivedatainOatpknockoutandvarious OATP1B-humanizedstrainsprovide strongsupportforthe con-ceptthatmouseOatp1b2andhumanOATP1B1andOATP1B3in thesinusoidalmembraneofthelivercancontributesubstantially totheliveruptakeofdocetaxel,andthusitsplasmaclearanceas showninFig.3AandB.Itisfurthernoteworthythatthe contribu-tiontodocetaxelclearanceofOATP1B1andOATP1B3inthehuman liverwilllikelybeatleastadditive.Itcouldthereforebethat co-administrationofdocetaxelwithstronginhibitorsofOATP1B1and OATP1B3mightresultinunwarrantedoverexposureofpatientsto docetaxel,whichonlyhasanarrowtherapeuticwindow.

2.1.3. Cabazitaxel

Thetaxanecabazitaxelwasrecentlyintroducedintheclinicas a second-linetreatmentof hormone-refractory prostate cancer. Amongstothersthiscompoundislessaffectedbymultidrug resis-tancecausedbytheMDR1P-glycoproteinthandocetaxel(Figgand Figg,2010;Kathawalaetal.,2015;PallerandAntonarakis,2011) However,recentstudiesdemonstratedthatstepwiseselectionof MCF-7cellstocabazitaxelresultedinP-gpoverexpression(Duran etal.,2015).(Interestingly,cabazitaxelhasaverysimilarstructure as docetaxel, withthe only difference being that two hydroxy groups in docetaxel are replaced by methoxy groups (Fig. 2). However,inspiteofthisgreatsimilaritytodocetaxel,cabazitaxel appearsnottobetransportedbyhumanOATP1B1andOATP1B3 ormouseOatp1b2inHEK293cells,orinvivoinOatp1b2knockout mice(Nieuweboer etal., 2014).Thisillustrateshow apparently minor structural modifications can sometimes have dramatic effects onwhethera compound is transported by OATPs,as is alsothecaseforMDRtransportersoftheABCsuperfamilysuchas ABCG2(Brametal.,2009).Onthepositiveside,thisprobablyalso meansthatforthisdrug,variationsinOATP1Bactivityinpatients duetogeneticpolymorphisms,drug–druginteractionsorvariable

expression in tumors, are less likely to affect the therapeutic efficacyortoxicity.

2.2. Irinotecan/SN-38

Theanticancerdrugirinotecanis atopoisomeraseIinhibitor widelyusedinthetreatmentofcolorectal,ovarianandlungcancer. Itstherapeuticindexisquitelow,inpartowingtoitscomplex phar-macokineticsinvolvingavarietyofmetabolicenzymesanddrug transporters.Irinotecan,essentiallyaprodrug,ishydrolyzedtoits primarypharmacodynamicallyactivemetabolite,SN-38,by vari-ouscarboxylesterasesoccurringinliver,intestine,and,inmice,also inplasma(Innocentietal.,2009;Mathijssenetal.,2002).Severe toxicsideeffectsofirinotecantherapyincludediarrheaand neu-tropenia,andthesegenerallycorrelatewiththesystemicexposure toSN-38(Smithetal.,2006).Low-activitypolymorphicvariantsof OATP1B1,whichmayleadtoimpairedhepaticclearanceof irinote-canand/orSN-38,areassociatedwithincreasedsystemicexposure toSN-38and life-threateningtoxicity (Han etal.,2008;Takane etal.,2009;Xiangetal.,2006).Itwasthereforeofgreatinterest toimproveourunderstandingoftheinvivoimpactofOATPson irinotecan/SN-38pharmacokineticsandtoxicityusingapanelof Oatp/OATPknockoutandtransgenicmice.Asitturnedout, how-ever,thisstudyalsorevealedapotentiallyimportantconfounder instudieswiththesemousestrainsfordrugsthatcanbeaffected byplasmacarboxylesterases.

Initialstudiesofi.v.administeredirinotecan(10mg/kg)in wild-typeandOatp1a/1b−/− miceshowedconsistentlyhigherplasma levelsofirinotecan(AUC1.7-foldhigher)andSN-38(AUC2.9-fold higher)intheknockoutmice,and roughlysimilarliver concen-trations,resulting in clearlydecreased liver-to-plasma ratiosof bothcompoundsintheknockoutsatvirtuallyalltimepoints(Iusuf etal.,2014).Thisisconsistentwithreduceduptakeofboththese compoundsfrombloodintotheliverintheabsenceofOatp1a/1b transporters,causinghigherplasmalevels.Theamountofplasma SN-38 as a fraction of plasma irinotecan was ∼10% in wild-typemice,and∼23%inknockoutmice.Amedium-termtoxicity

(8)

experiment,withdailyi.v.administrationsofirinotecan(30mg/kg) over 6 days resulted in more extensive weight loss in the Oatp1a/1b−/−mice,andamorepronouncedreductioninthewhite bloodcellcountthaninthewild-typemicemeasuredattheendof theexperiment(day7).Assessedatthissameday7,therewassome toxicitytobonemarrow,thymus,andsmallintestineinwild-type mice,butthiswasmuchmorepronouncedinOatp1a/1b−/−mice (Iusuf etal.,2014).Thesedatawereinlinewitha muchhigher plasmaconcentrationofSN-38intheOatp1a/1b−/−micemeasured 24hafterthelastirinotecanadministration,whereastheirinotecan concentrationsatthistimepointwerenotsignificantlydifferent fromwild-typemice.It seemsvery likelythatthehigher expo-suretoSN-38intheOatp1a/1b−/−micewasthemaincauseofthe increasedtoxicity.Themostobviousexplanationforthehigher SN-38exposurewasstronglyreducedhepaticclearanceofSN-38,and likelyalsoofitsprecursoririnotecan,byeliminationofthemouse Oatp1a/1btransporters.

2.2.1. UpregulationofplasmacarboxylesteraseinOatp1a/1b−/− mice

Furtheranalysesrevealed,however,thatthehigherSN-38 expo-surewas not caused solely by a decreased clearance of SN-38 directlyduetoadeficiencyofOatp1a/1btransporters.Itturnedout that,testedexvivo,plasmaofOatp1a/1b−/−micecontainedahighly increasedlevelofirinotecanhydrolaseactivity,whichcausedafar morerapidformationofSN-38fromirinotecanthanobservedin wild-typeplasma. Thisdifference wasmostlikely theresult of thehighlyincreasedexpressionofanumberofcarboxylesterase (Ces)genesintheliverofOatp1a/1b−/−mice,namelyCes1b,Ces1c, Ces1dandCes1e.Ces1c,whichisrelativelyabundant,displayed an80-foldincreaseinRNAlevels(Iusufetal.,2014).Interestingly, themouseCes1cenzyme isknowntobemainlysecretedfrom liverintoplasma,duetotheabsenceofa C-terminal endoplas-micreticulumretentionsignalinitsaminoacidsequence(Holmes etal.,2010).Applicationofthecarboxylesteraseinhibitor bis(4-nitrophenyl)phosphate(BNPP)supportedthatincreasedplasma carboxylesterase activity was responsible for the hydrolysis of irinotecaninOatp1a/1b−/−plasma.Moreover,variousothertested Ces1,Ces2,andCes3familygenes,andgenesforothercandidate plasmaesteraseenzymes likebutyrylcholinesterase,Aadac, and Pon1-3,werenotupregulatedintheOatp1a/1b−/−liver.Altogether, Ces1cupregulationisthusthemostlikelycauseoftheincreased hydrolysisofirinotecaninOatp1a/1b−/−mice.

ThattheincreaseinCes1cexpressionwasadirectconsequence of the functional Oatp1a/1b deficiency wasstrongly supported byanalysisof Cesgene expressionin OATP1B1-and OATP1B3-humanizedtransgenicderivativesofOatp1a/1b−/−mice:thehighly increasedliverexpressionofCes1c,butalsoofCes1b,Ces1dand Ces1e,weremarkedlyreduced,forCes1candCes1dtoevenbelow thelevelofexpressionseeninwild-typemice,bybothtransgenic OATP1B1andOATP1B3(Iusufetal.,2014).Thisindicatesthatthe functionoftheremovedOatp1a/1bgenesresponsiblefor keep-ingliverCes1expressionlowcouldbetakenoverbybothhuman OATP1B1andhumanOATP1B3.Still,theexactmechanism under-lyingtheseCes-regulatoryfunctionsofOatp/OATP1proteinsisas yetunclear.Anobviouspossibilityappearstobeadetoxifying func-tionoftheseproteins,normallyinvolvedinremovingoneormore Ces-inducingcompoundsfromthebody.Suchcompoundsmight beofendogenousorexogenousorigin(dietary,ormetabolitesor evenprimaryproductsformedbytheintestinalmicroflora),or pos-siblyboth.Theirprolongedretentioninthebodymightincrease theoverallliverexposureandthusCesinductionintheknockout strain.Furthersupportfortheinvolvementofadetoxifyingroleof theOatp1a/1bproteinsinCesgeneexpressionemergesfromthe findingthattheknockoutofanumberofotherbroad-specificity detoxifyingproteinsinFVBmice,includingthemultidrugefflux

transportersAbcb1a/1bandAbcg2,andthemultidrug metaboliz-ingCyp3acomplex,causesimilarlevelsofupregulationofhepatic Ces1b,Ces1c,Ces1d,andCes1eexpression(Lagasetal.,2012;Tang etal.,2015,2014).Alternatively,afunctionoftheOATPsinliver uptakeofCes-repressingcompoundsisalsoapossibility.Ifthese compoundsthengetefficientlyclearedthroughalternativeroutes, mayberenalorbymetabolism,theeffectiveliverexposuretothese compoundsmight bereducedin theOatp1a/1bknockoutmice. However,forthemomenttheseoptionsremainspeculative. 2.2.2. IrinotecanstudiesinOATP1B1andOATP1B3humanized mice

Whatever themechanismof Cesupregulation,theincreased plasmalevelsofCes1cinOatp1a/1b−/−micemightconfound inter-pretation of the irinotecan and SN-38 disposition and toxicity resultsinthesemice.Theincreasedconversionofirinotecan to SN-38 could reduceplasma irinotecan levels,and increase SN-38levelsindependentofdirectOatp-mediatedtransport.Forthis reason,amongothers,i.v.irinotecanstudieswerealsoperformed intheOATP1B1andOATP1B3humanizedmice,asthesehavea mostlynormalizedplasmaCes1cactivity.Theresultsobtainedfor irinotecan and SN-38 were strikingly different: the humanized miceshowedhigherplasmalevelsofirinotecancomparedto wild-typemice,butsimilarliverconcentrations,resultinginmarkedly reducedliver-to-plasmaratiosofirinotecan.Asthesedifferences cannotbeattributedtoCesupregulation,theresultssuggestthat thetransgenicOATP1B1andOATP1B3arefarlessefficient(ifat all)intakingupirinotecanintotheliverthanthemouseOatp1a/1b proteins(whichareabsentfromthehumanizedmice).Thisalso explainsthehigherplasmalevelsofirinotecaninthehumanized micecomparedtowild-typemice.

In contrast,for SN-38afteri.v. irinotecan administrationwe foundthattheplasmalevelsinthetransgenicmiceweremarkedly reducedcomparedtoOatp1a/1b−/−mice,butsimilartothosein wild-typemice(Fig.3CandD).Intheorythiscouldeitherbecaused bytheup-anddown-regulationofCes1c,orbychangesinSN-38 clearance.Interestingly,theliverlevelsinalltestedstrainswere thesame,andtheliver-to-plasmaratiosofSN-38wereidentical betweenthewild-typeandtransgenicstrains,butmarkedlyhigher thaninOatp1a/1b−/−mice.Thelatterresultstronglysuggeststhat theliveruptakeofSN-38wascompromisedintheOatp1a/1b−/− mice,andmostlyrestoredbyOATP1B1andOATP1B3inthe human-izedstrains(Fig.3CandD).

2.2.3. SN-38dispositioninOatp1a/1b−/−mice

Inanalternativeapproachtocircumventthecomplicationsof Cesupregulationin theOatp1a/1b−/− mice,SN-38was directly administeredi.v.tothesemice,at1mg/kginviewofthe consider-ablesolubilityandformulationlimitationsofSN-38.Thisresulted insignificantlyhigherplasmalevelsofSN-38shortlyafter admin-istration,andsignificantlylowerliverconcentrationscomparedto levelsinwild-typemice.Areducedshort-termliveruptakeof SN-38wasfurthersupportedbyatwo-folddecreaseinbiliaryexcretion ofSN-38intheOatp1a/1b−/−mice(Iusufetal.,2014).

Collectively,themousestudiesindicatethatbothirinotecanand SN-38areclearedfromplasmabyuptakeintotheliverthrough oneormoreofthemousehepaticOatp1a/1btransporters.In con-trast,thetransgenichumanOATP1B1andOATP1B3canmediate substantialliveruptakeandplasmaclearanceofSN-38,butnotof irinotecan.Theseresultsfitwellwithinvitrostudies,whichfound thatSN-38isreadilytransportedbyhumanOATP1BandOATP1B3, whereasirinotecanisnot(Nozawaetal.,2005;Oostendorpetal., 2009; Yamaguchiet al.,2008).The resultsindicatespecies dif-ferencesinsubstratespecificitybetweenthemouseandhuman hepaticOATP1A/1Bproteins,asexpressedinvivoinmouse hepa-tocytes.Theirinotecan/SN-38toxicitydatainthemousestrainsare

(9)

moredifficulttointerpretinviewofthealteredCes1clevelsand rateofSN-38formationinOatp1a/1b−/−mice.However, extrapo-latingfromthedelayedhepaticSN-38clearanceinOatp1a/1b−/− miceanditsreversalbyhumanOATP1B1andOATP1B3,itseems verylikelythatalsoinhumanstheseproteinsareinvolvedinthe detoxification ofSN-38. Thiswould be in linewiththeclinical observationsthatpartialdeficienciesinOATP1B1duetogenetic polymorphismscorrelatewithincreasedirinotecan/SN-38toxicity (Hanetal.,2008;Takaneetal.,2009;Xiangetal.,2006).Thedataof Iusufetal.(2014)stronglysuggestthatthishasprimarilytodowith delayedSN-38clearanceinthesepatientsduetothecompromised functionofOATP1B1.

2.3. Methotrexate

Methotrexate is a folate antimetabolite that is used in the treatmentofseveralimportantcancertypes(breastcancer,lung cancer,head-andneckcancer,non-Hodgkin’slymphoma)(Assaraf, 2007;GonenandAssaraf,2012).Itisalsoused,albeitatusually lower dosage,and mostly orally, totreat non-malignant disor-derssuchasrheumatoidarthritisandpsoriasis(vanOutryveetal., 2002;Wesselsetal.,2008).Methotrexateisabicarboxylicorganic anion,that isknowntobetransported invitrobya number of OATP1A/1Bproteins, includinghuman OATP1B1,OATP1B3, and OATP1A2(Abeetal.,2001;Badagnanietal.,2006;Sasakietal., 2004).Inaddition,agenome-wideassociationstudyshowedthat SNPsinSLCO1B1associatedwithreducedtransportactivitywere linkedtodecreasedplasmaclearanceanddecreased gastrointesti-naltoxicityinchildrenwithacutelymphoblasticleukemia(ALL) treatedwithhigh-dosei.v.methotrexateinfusions(Trevinoetal., 2009).

2.3.1. MethotrexatepharmacokineticsinOatp1a/1bknockout mice

Initialcharacterizationofi.v. methotrexatepharmacokinetics inOatp1a/1b−/−micerevealeddramaticeffectsonplasma clear-anceandliveruptakeofthedrug(vandeSteegetal.,2010).The plasmaAUCwasincreased5-foldinOatp1a/1b−/−mice,andliver concentrationswereabout20-foldreduced.Hepatic methotrex-ateuptakewasveryrapid,with>50%ofthedoseaccumulatingin thewild-typeliverwithin3.5minafteri.v.dosing,whereasonly ∼2%ofthedrugaccumulatedintheliverofOatp1a/1b−/−mice. Ascouldbeexpectedfromthedramaticallyreducedliveruptake ofmethotrexate,thetotalintestinalcontentofi.v.methotrexate, whichmostly derivesfrombiliaryexcretion,wasabout17-fold reduced in the Oatp1a/1b−/− mice. Accordingly, theamount of unchangedmethotrexateexcretedinthefeceswasreducedfrom ∼20%to∼2% ofthedoseadministered.In contrast,theurinary excretionrosefrom∼40%ofthedoseinwild-typemiceto∼100%in Oatp1a/1b−/−mice,indicatingasubstantialreroutingfrom hepa-tobiliarytorenalexcretion.

AssomeOatp1a/1bproteinsarealsoexpressedinthemouse smallintestine(Slco1a4,andtoalowerextentSlco1a5andSlco1a6 RNAweredetected(Chengetal.,2005),anoralmethotrexatestudy wasalsoperformedtoassesspossiblechangesinoralavailability. However,whereasthesystemicplasmaandliverconcentrationsof methotrexatefollowedsimilarpatternsasseenafteri.v. adminis-tration,short-termhepaticportalveinsamplingdidn’tprovideany indicationforareducedrateofintestinaluptakeofmethotrexatein theOatp1a/1b−/−mice.Apparentlyotherintestinaluptakesystems areprimarilyinvolvedintheuptakeofthisbi-anioniccompound.

Infact,despiteextensiveefforts,noonehasthusfardirectly demonstratedforanyOATPsubstratedrugthatitsintestinaluptake is detectably dependentonOatp1a/1b proteins. Directly tested drugsinclude,inadditiontomethotrexate,fexofenadine, pravas-tatin,androsuvastatin(Iusufetal.,2012a,2013;vandeSteegetal.,

2010),aswellasanumberofother,asyetunpublished,drugs.This raisesthebroaderquestionwhetherornotOatp1a/1bproteinsplay anysignificantroleintheintestinaluptakeoforallyadministered substratedrugs.However,therecanbemanycausesfornegative resultsinthisrespect,includingthepossibilityofextensive redun-dancywithoneormoreotherintestinaluptaketransportsystems. Furtherexperimentation willbe neededto resolve the intrigu-ingquestionwhattransportsystemsareprimarilyinvolvedinthe intestinaluptakeofaspectrumofrelativelypolarorchargeddrugs. TheOatp1a/1b−/−micecanalsobeusedtoassesstheefficacy andspecificityofOATP-inhibiting drugs,forinstanceasacause ofdrug–druginteractions. Forthis purposerifampicin,aknown OATPinhibitor,wasadministeredi.v.3minbeforei.v. methotrex-atetowild-typeandOatp1a/1b−/−mice,andmethotrexateplasma and liverconcentrations weredetermined 15minlater (vande Steegetal.,2010).The methotrexateplasmaconcentrationwas increasedthreefoldbyrifampicintreatmentinwild-typemice,to thesamelevels asseen inOatp1a/1b−/− mice.In theknockout mice,rifampicintreatmenthadnoeffectontheplasmalevelsof methotrexate.Theliverconcentrationofmethotrexateinwild-type micewas4-folddecreasedbyrifampicin,toabout9%ofthe admin-istereddose.TheliverconcentrationinOatp1a/1bknockoutmice wasstilllower(∼1%ofthedose),andnotaffectedbyrifampicin treatment.Theseresultsindicatethatrifampicincouldlargely,but notcompletely,inhibitOatp1a/1b-mediatedmethotrexateuptake intotheliver,andthusitsassociatedplasmaclearance.Thelack ofeffectofrifampicinonmethotrexatepharmacokineticsinthe Oatp1a/1b−/−miceindicatesthatitdidnotsignificantlyaffectother methotrexate clearance mechanisms, attesting to its specificity undertheseconditions.

2.3.2. MethotrexatepharmacokineticsinOATP-humanizedmice In a follow-up study, methotrexate pharmacokinetics was analyzed in OATP1B1-, OATP1B3- and OATP1A2-humanized Oatp1a/1b−/−mice(vandeSteegetal.,2013).Asexplained else-wherein thisreview (Section2.1.1),thesethree transgenesare primarilyexpressedinliverparenchymecellsinthesemice,andthe proteinsaresituatedinthebasolateral(sinusoidal)membrane.For OATP1B1andOATP1B3thisisthephysiologicallyrelevant local-ization,butfor OATP1A2it isnot,asin humanliveritis found primarilyincholangiocytes,theepithelialcellsliningthebileducts. Still, inclusionof thelatterstrain allowsanalysisof theinvivo functioningofOATP1A2indruguptake,whichcanberelevantfor assessmentofanydruguptakethatitmaymediateinothertissues andintumorcells.

Methotrexatewasadministeredi.v.attwodifferentdoses(10 and2mg/kg)towild-type,Oatp1a/1b−/−,andthethree human-izedmousestrains, andplasma,liver,andintestine (tissueplus contents) levels of methotrexate were measured 15min after administration(vandeSteegetal.,2013).Asobservedpreviously, at10mg/kgintheknockoutmice,methotrexateplasmalevelswere increased5-fold,whereasliverlevelsweredecreased24-fold,and smallintestinelevels20-fold(Table2).Eachofthethreehumanized transgenespartiallyreversedallofthethreemeasured parame-ters,albeitnottothelevelsseeninwild-typemice.Plasmalevels ofmethotrexatewerereduced∼2-fold,andliverlevelsincreased by4-to9-fold.Smallintestinallevelswere2-to4-foldincreased. Qualitativelyvery similarresults wereobtained atthe 2mg/kg methotrexatedose(Table2).Ingeneral,transgenicOATP1B3and OATP1A2causeda∼2-foldmoreeffectivereversalofliverandsmall intestinalconcentrationsthanOATP1B1,whereastheplasma rever-saleffectsweresimilarbetweenthethreetransgenes.Thepartial reversalbythehumanizedOATP1B1andOATP1B3proteins com-paredtowild-typeparameterscouldrelatetospeciesdifferencesin substratepreferencebetweenmouseandhumanOatp/OATP pro-teins,but alsoto differencesin effective expressionlevel. Also,

(10)

Table2

ImpactofOATPtransportersinOatp1a/1bknockoutandOATP1A/1BtransgenicmiceonplasmaconcentrationandtissuedistributionofintravenouslyadministeredMTX(at

10and2mg/kg,15minafteradministration).

Dose Mousestrains Plasmaconcentration(␮g/ml) Liveramount(%ofdose) Smallintestineamount(%ofdose)

10mg/kg WT 1.55±0.24 30.0±4.1 20.9±1.4 Oatp1a/1b−/− 7.59±0.76 1.24±0.14 0.94±0.17 Oatp1a/1b−/−;1B1tg 4.66±0.38 4.54±1.24 2.48±0.60 Oatp1a/1b−/−;1B3tg 4.35±0.52 9.62±0.69 5.21±1.03 Oatp1a/1b−/−;1A2tg 4.03±1.00 11.3±2.1 5.81±2.56 2mg/kg WT 0.50±0.07 33.0±5.1 12.0±0.6 Oatp1a/1b−/− 1.90±0.29 1.65±0.14 1.62±0.26 Oatp1a/1b−/−;1B1tg 0.93±0.18 5.09±0.41 2.56±0.60 Oatp1a/1b−/−;1B3tg 1.11±0.14 10.2±1.2 3.93±0.51 Oatp1a/1b−/−;1A2tg 0.88±0.11 9.07±0.96 3.76±0.51

in humanliver OATP1B1 and OATP1B3 would presumably act additivelytowardsmethotrexate,andthusmaycauselarger phar-macokineticeffectsthansuggestedbytheeffectsseeninthesingle transgenicstrains.Regardless,thedataclearlyshowthathuman OATP1B1andOATP1B3canhavemajoreffectsontheplasmalevel, hepaticuptakeclearance,andsubsequenthepatobiliary/intestinal excretionofmethotrexate.Moreover,asOATP1B3isalsoexpressed invariousgastrointestinal,hepatocellular,breast,andlungcancers, itmayfurtheraffectsusceptibilityof thesecancerstoOATP1B3 substratedrugs(Abeetal.,2001;Cuietal.,2003;Monksetal., 2007;Mutoetal.,2007).ThedataobtainedforhumanOATP1A2 suggestthatthisproteincouldsubstantiallyaffectmethotrexate uptakeinvivoinotherrelevanttissuesandbarriers,suchasthe blood-brainbarrier,kidneytubules,andtumorcellsthatexpress OATP1A2(Gaoetal.,2000;Leeetal.,2005).

2.3.3. Impactofrifampicinandtelmisartanco-administrationon OATP-mediatedmethotrexatedisposition

Thehumanizedmousestrainsweresubsequentlyusedto fur-therinvestigateOATP-dependentdrug–druginteractions,starting with the inhibitory effect of rifampicin on in vivo methotrex-atetransport by humanOATP1B1 and OATP1B3. Using various humanOATP-overexpressingHEK293cells,Durmusetal.(2015) showedthat rifampicin inhibited methotrexateuptake invitro, withIC50 levelsrangingfrom0.3to0.9␮M.Inthemouse mod-els,rifampicin(20mg/kg,i.v.)substantiallyinhibitedbothmouse andhumanOATP-mediatedhepaticuptakeandplasmadisposition ofmethotrexate(10mg/kg,i.v.)atclinicallyachievable concentra-tionsforeachofthedrugs.AsshowninTable3,liver-to-plasma ratiosofmethotrexateweredecreased6-to8-foldbyinhibitingthe mouseOatp1a/1bproteins,∼4-foldbyinhibitinghumanOATP1B1 and11-to18-foldbyinhibitinghumanOATP1B3.Thisalsoledto increasedplasmalevelsofmethotrexateby4-to5-foldinmouse Oatp1a/1bbackgroundandupto2-foldinhumanizedOATP1B1or OATP1B3background(Table3).

Althoughthisspecificcombinationofdrugs(methotrexateand rifampicin)wouldberareintheclinic,theseresultsstillsuggest thatmorethanoneOATP-substrateor-inhibitordrugsmightbring theriskofdrug–druginteractionsatthesystemicandhepaticlevel. Thiswouldbeespeciallyimportantforpatientschronicallytaking OATP-interactingdrugs, suchasstatinsandhypertensiondrugs. Moreimportantly,peoplewhoalreadyhaveactivity-reducing poly-morphismsintheirOATPgenes(NakanishiandTamai,2012)might beatincreasedriskforaltereddrugdispositionduetothistype ofdrug–druginteractions,andconsequentlyineffectivetreatment and/orincreasedtoxicity.

Thepossibledrug–druginteractioneffectofaclinicallymore commondrug combination wasinvestigated using a hyperten-sivedrug,telmisartan,andmethotrexate.Thisalsoallowedfurther evaluationoftheapplicabilityofknock-outandhumanizedmouse strainsin drug–druginteractionstudies.Asimilarset-uptothe

rifampicin and methotrexate experiments was chosen, but the doseof telmisartanhad tobekeptlower(7mg/kg)due to sol-ubility issues. In vitro cellular uptake studies resulted in fairly butnotverylowIC50levelsfortelmisartaninhibiting methotrex-ateuptake (<11␮M),and in vivostudies in the mouse models showedthat telmisartanpre-treatmentdidnot yieldimportant changesinmethotrexatedisposition.Therewasonlyaweak inhi-bitionofOATP1B1-mediatedhepatic uptakeofmethotrexateby telmisartan,leadingto∼2-folddecreasedliver-to-plasmaratiosof methotrexate.However,comparingthesystemictelmisartan lev-els(40–200nM)inpatients(Stangieretal.,2000)withthelevels achievedin mouseplasma (7–13␮M)suggestsonly avery low risk ofadverse OATP-mediateddrug–druginteractions between telmisartanandmethotrexateintheclinic.Thesefindingsare help-fulforpatientswhoaresimultaneouslytreatedwithmethotrexate andtelmisartan.However,additionalclinicallyrelevant combina-tionsofdrugsthatareOATP-substratesand/or-inhibitorsshouldbe testedtoevaluatetherisksofOATP-mediateddrug–drug interac-tionsintheclinic,especiallyforthesystemicandhepaticeffectson drugconcentrationsduetothefunctionofOATPsinliveruptakeof theirsubstrates.Wesuggestthatthehumanizedtransgenicmouse modelswithliver-specificOATPexpressioncouldbeusedtoobtain amorerealisticviewofthehumansituation.However,itshouldbe notedthatthesemousemodels,whileveryusefultosupportthe developmentofbasicmechanisticinsights,shouldnotbeusedas simpleone-to-onemodelsofdrugbehaviorinhumans.

2.4. Doxorubicin

Doxorubicin,animportanttopoisomeraseIIinhibitordrugused incancertreatment,wasrecentlyshowntobeanOATPsubstrate (Durmusetal.,2014).Doxorubicinisananthracyclineantibiotic, commonly used in the treatment of several cancers, includ-ingHodgkin’sandnon-Hodgkin’slymphoma,multiplemyeloma, leukemia,sarcomaandbreast,ovarian,thyroid,gastricandlung cancers(Cortes-FunesandCoronado,2007;Gewirtz,1999). Vari-ousmodesofactionhavebeendescribedfordoxorubicin,ofwhich twomajormechanismsareintercalationofDNAthatleadsto inhi-bitionofthetopoisomeraseIIenzyme,requiredforsmoothDNA transcriptionandreplication,and,perhapsmorecontroversially, generationoffreeradicalsthatleadstoDNAandcellmembrane damage (Gewirtz,1999).Cardiotoxicity is the majorand dose-limiting side effect of doxorubicin both in adult and pediatric cancerpatients(GrenierandLipshultz,1998).Thus,recenteffortsto improvedoxorubicintreatmentfocusonimprovedanti-tumor effi-cacyanddecreasedcardiotoxicitybydevelopingtumor-targeting strategiessuchasliposomalformulationsofdoxorubicinor devel-opingnewdoxorubicinanalogs(Cortes-FunesandCoronado,2007; Minottietal.,2004).Anotherstrategyforloweringthetoxicityand increasingtheefficacyofdoxorubicinistounderstandthefactors affectingitstissuedisposition,suchasitsinteractionswithdrug

(11)

Table3

Impactofrifampicinpre-treatmentontheplasmalevelsandliver-to-plasmaratiosofmethotrexateinWT,OatpknockoutandOATP1Bhumanizedtransgenicstrains.

Mousestrains

Timepoint Parameter Rifampicin WT Oatp1a/1b−/− Oatp1a/1b−/−;1B1tg Oatp1a/1b−/−;1B3tg

5min

Plasmaconcentration(nmol/ml) − 11.9±1.7 46.6±7.4 49.0±15.3 32.2±3.7

+ 50.2±7.8 67.7±3.5 53.6±9.4 62.3±6.9

Rifampicin-inducedfoldincrease 4.2 1.5 1.1 1.9

Liver-to-plasmaratio − 15.5±2.0 0.2±0.0 0.5±0.1 1.7±0.3

+ 1.9±0.3 0.1±0.0 0.2±0.0 0.2±0.0

Rifampicin-inducedfolddecrease 8.2 2.0 2.5 8.5

15min

Plasmaconcentration(nmol/ml) − 6.6±2.7 31.2±13.4 43.1±4.2 19.5±10.3

+ 30.7±12.9 43.9±14.1 46.4±10.0 43.1±18.8

Rifampicin-inducedfoldincrease 4.7 1.4 1.1 2.2

Liver-to-plasmaratio − 27.0±5.9 0.2±0.0 1.1±0.2 5.1±1.4

+ 4.7±2.5 0.2±0.0 0.3±0.1 0.3±0.0

Rifampicin-inducedfolddecrease 5.7 1.0 3.7 17.0

transporters,andutilizetheseinsightstomodifythe pharmacoki-netics,possiblywithtransporterinhibitors.Althoughdoxorubicin hasbeenusedintheclinicforaverylongtime,informationonits interactionwithdruguptaketransportershasbeenverylimited. Okabeetal.(2005)suggestedforthefirsttimethatdoxorubicin mightentercellsviaorganiccationtransporter(OCT6)-mediated activetransport.

Very recently, it was shown that both mouse and human OATP1A/1Bfamilymemberscanmediatecellularuptakeof doxo-rubicin(Durmus etal., 2014).This wasrathersurprising, asits structure,withpropertiesofaweakbase,wasnotconsidereda typ-icalOATPsubstrate.InhumanOATP1A2-overexpressingHEK293 cells,theuptakeofdoxorubicinwas∼2-foldincreasedcompared tocontrolcells,buttherewasnonoticeabletransportbyhuman OATP1B1orOATP1B3invitro.MouseOatp1a/1bproteins trans-porteddoxorubicininvivo,asevidencedbyupto∼2-foldincreased plasmaconcentrationsandupto4-folddecreasedliver-to-plasma ratiosinOatp1a/1b−/−miceafteri.v.doxorubicinadministration (Fig. 3E and F) (Durmus et al., 2014).Moreover,the doxorubi-cin levelsin thesmall intestinal content were alsoreduced in theknockoutmice,suggestingadecreasedbiliaryoutput, proba-blyduetothelowerliverlevels.Interestingly,additionofoneof thehumanOATP1Aor1Btransportersintransgenicmice(on a mouseOatp1a/1bknockoutbackground)rescuedthealtered lev-els seen in the knockout mice to various extents (Fig. 3E and F).Transgenicliver-specificexpressionofhumanOATP1A2could completelyreverse thehepatic uptake and intestinal excretion ofdoxorubicinbacktowild-typelevels,whereasthatofhuman OATP1B1orOATP1B3resultedin partial,but substantialrescue of these phenotypes (Durmus et al., 2014). Only the OATP1B1 orOATP1B3transportersalonewerepresentinthesetransgenic mouselivers,insteadofbothsimultaneouslyasinhumans.The substantialimpactofeithertransporteralonethereforesuggests an even stronger impact of OATP1Bs on doxorubicin pharma-cokineticsin humans,especially affectingplasmaclearance and hepaticuptakecharacteristics.Obviously,fora drugwitha nar-rowtherapeuticindexlikedoxorubicin,thismightbecriticalforits therapeuticefficacyandtoxicityinpatients.Itmaythereforealso beofinteresttoinvestigatepossibleassociationsbetweenOATP transportactivityanddoxorubicinpharmacokinetics,therapeutic efficacy,andtoxicityinpatientcohorts.

2.5. Platinumchemotherapeutics

Cisplatinand morerecent platinum chemotherapeuticssuch ascarboplatinandoxaliplatinareusedtotreatawidespectrum of cancers, including testicular,bladder, lung, ovarian, colorec-tal,cervical,andbreastcancers,aswellassomelymphomasand sarcomas.InaCOMPAREanalysisof60humantumorcelllines(the

well-characterizedNCI-60panel),Lancasteretal.(2013)foundthat highOATP1B3expression,asjudgedbyReal-timeRT-PCR,was sig-nificantlylinkedwithsensitivitytothecytotoxicityof9anticancer drugs,amongstwhichcisplatin,carboplatin,andaplatinum com-poundstructurallyrelatedtooxaliplatin.UnlikeOATP1B1,which wasfoundtobeonlyexpressedinliverand livertumortissue, OATP1B3wasfoundexpressedinarangeofdifferenttissuesand derivedtumortissuesamples,suggestingthatitmightplayarole intumorsensitivitytoplatinumcompounds.

SubsequentinvitrostudiesrevealedthathumanOATP1B3and OATP1B1 canmediate thecellular uptake of Ptupon exposure to cisplatin, and additionally OATP1B3 can mediate cellular Pt uptakeuponexposuretocarboplatinandoxaliplatin.Invivo stud-ieswithcisplatinadministeredintraperitoneallytowild-typeand Oatp1b2−/−miceshowedupto35%reducedliverlevelsofPtinthe knockoutmiceshortlyafteradministration,and2.5-foldhigher uri-naryexcretionofPt,thelatteramountingtomorethan60%ofthe administereddose(Lancasteretal.,2013).Thedatasuggestarapid andsubstantialliveruptakeofafractionofPtbeingmediatedby Oatp1b2inwild-typemice.IntheOatp1b2−/−mice,thisPtfraction mayinsteadbeexcretedprimarilyintotheurine.Asaconsequence, theplasmalevelsofPtarenotmuch differentbetweenthetwo strains.Itshouldbenoted,however,thatcisplatinishighlyreactive, forinstancewithcarbonate,andtheOatp1b2-mediatedPtuptake maythereforewellrepresentuptakeofvariousresultingnegatively chargedPtcomplexesinsteadofcisplatinitself.Cisplatinalsobinds irreversiblytoplasmaproteins,furthercomplicating pharmacoki-netic analyses.Howeverthat maybe, invivo Oatp1b2function doesclearlyaffectthepharmacokinetics ofcisplatin-derived Pt. Extrapolatingtohumanpatients,thiscouldmeanthatvariationsin OATP1B1andOATP1B3expressionandtransportactivityinliver,in othertissues,andespeciallyintumors,mightaffecttheefficacyand toxicsideeffectsofcisplatin-basedchemotherapy.Itwilltherefore beofinteresttoextendpreclinicalstudiesofcisplatinto OATP1B1-and OATP1B3-humanized Oatp1a/1b knockoutstrains, and also includestudies ofcarboplatin andoxaliplatin. It willfurtherbe worthwhiletoassesswhetherpartofthehighinter-individual vari-ationinefficacyandtoxicityofthesedrugsseeninpatientscanin partbeattributedtovariationsinOATPfunction.

2.6. Tyrosinekinaseinhibitors

Tyrosinekinaseinhibitors(TKIs),withtheprimeexample imat-inib (Gleevec), have revolutionized the treatmentof a number ofmalignancies (Drukeretal.,2001).Asof 2015,more than28 TKIs have been FDA-approved (Wu et al., 2015). These ratio-nallydesigned,targetedanticancerdrugscanmakeuseofspecific vulnerabilities of tumor cells, for instance bytargeting specific oncogenicmutationsinproteintyrosinekinases,thusincreasing

(12)

disease-specificityandreducingtheriskoftoxicsideeffects. Under-standably,therehasbeensignificantinterestinwhetherTKIsare OATPtransportsubstrates,asthiscouldaffecttheiraccumulation intumorcellsaswellasinvariousimportanttissues,andcontrol theirplasmapharmacokineticsandelimination.Suchparameters canbedecisiveintheoveralltherapeuticefficacyandtolerability ofanticancerandothertherapeuticagents.

Whilemany TKIs havenowbeen testedfor interactionwith OATPs,the invivo relevance for transport of the unconjugated parentcompoundsappearstobelimited.InRNA-injected Xeno-pusoocytes,humanOATP1A2mediatesmodestimatinibuptake, OATP1B3alittle,andOATP1B1none(Huetal.,2008).Initialtests failedtoshowsignificantuptakeofsorafenibandsunitinib medi-atedbyOATP1A2,OATP1B1,andOATP1B3inRNA-injectedXenopus laevisoocytes(Huetal.,2008).However,laterstudieswithhighly OATP-overexpressingHEK293cellsreportedsignificantuptakeof crizotinib, nilotinib,pazopanib, and sorafenib by OATP1B1 and OATP1B3ofimatinib,gefitinib,dasatinib,vandetanib,and vemu-rafenib by OATP1B3 but not by OATP1B1; and of sunitinib by OATP1B1butnotbyOATP1B3(Zimmermanetal.,2013).Itshould benoted,however,thatoftenrelativelevelsofuptakeofTKIs medi-atedbyOATPin thevariousinvitromodelsystemsweresmall comparedtothoseoftheanionicOATPcontrolsubstrates. Mod-erntargetedTKIsarerarely,ifatall,designedtobeanionic,given theusualrequirementsofgoodoralavailability(intestinal absorp-tion)andefficienttumorcellpenetrationofthesecompounds.A possiblefurthercauseofdiscrepanciesbetweeninvitroandinvivo OATP-mediatedtransportresultsforcertaindrugsisdiscussedlater inSection4ofthisreview.

2.6.1. Invitroandinvivostudieswithsorafenib

TheTKI sorafenibwas furtherinvestigatedin vivoin mouse models because it was readily transported by both OATP1B1 andOATP1B3invitro.Unlikeitsconjugate,sorafenib-glucuronide, which will be discussed later, the plasma pharmacokinetics of parent sorafenib, or its relative accumulation in the liver,was notsubstantially altered in eitherOatp1b2−/− mice(DBA back-groundstrain),orinOatp1a/1b−/−mice(FVBbackgroundstrain) afteradministrationof10mg/kgoralsorafenib(Zimmermanetal., 2013).Apartialexplanationmightbethatinvitrosorafenibwas poorlytransportedbymouseOatp1b2.However,sorafenib trans-portbythetwootherprominentsinusoidaluptake transporters inthemouseliver,Oatp1a1andOatp1a4,wasnottestedinvitro, soitremainsuncertainwhetherornotthereisaninvitro/invivo discrepancy(Zimmermanetal.,2013).

SomeTKIshavebeenfurtherevaluatedfortheirabilitytoinhibit humanOATP1B1invitroandinvivo(Huetal.,2014).These stud-iesweretriggeredbyclinicalobservationsthatco-administration ofseveralTKIscanincrease thesystemic exposuretodocetaxel incancerpatients.Theunderlyingmechanismwaspoorly under-stood,but,asdocetaxelclearance mayin partdependonOATP function(seeSection2.1.2),itmightinvolveinhibitionofOATPsby theseTKIs(Huetal.,2014).Nearlyall16testedTKIsinhibitedE2G uptakebyhumanOATP1B1invitrowhenappliedat10␮M,and4 oftheseTKIs(axitinib,nilotinib,pazopanib,andsorafenib)bymore than10-fold.Interestingly,threeofthese(axitinib,pazopanib,and sorafenib) are known to increase the docetaxel AUC by up to 50%ormorewhenco-administeredinpatients(clinicaldatafor nilotinib/docetaxel co-administrationare not available).Further analysisofsorafenibshowedthatitinhibiteddocetaxeluptakeby OATP1B1invitrowithanIC50below100nM,andveryextensively whenappliedat10␮M.MouseOatp1b2showedsimilarinhibition ofE2Ganddocetaxeltransportbysorafenibinvitro.Invivo, how-ever,nosignificanteffectofhigh-dose(60mg/kg)oralsorafenib co-administrationcouldbedemonstratedon10mg/kgi.v. doce-taxelplasmaCmaxorAUCinwild-type,Oatp1b2−/−,Oatp1a/1b−/−,

or in OATP1B1-humanized mice. Moreover, multiple sorafenib administrationsdidnotelicitsignificanteffects(Huetal.,2014). Ofnote,inthesespecificexperiments(seealsoSection2.1.2),the effectsofOatp1b2knockoutorOATP1B1transgenicsondocetaxel AUCwererelativelymodest(about2-fold).Thismayhaverendered theseexperimentslesssensitivetopickingupsmall pharmacoki-neticeffectsofsorafenib-mediatedinhibition.Theabsenceofan obviouspharmacokineticinteractionbetweensorafeniband doce-taxelinthevariousmousestrainssuggeststhattheremaybeother factorsinmice,perhapsalternativeorcompensatorymechanisms that can sufficientlyoffset anychanges in OATP1B-like activity towardsdocetaxelclearance.Heretoo,onehastoconsiderthe pos-sibilitythatinteractionsofOATPswithcertaindrugsmaydepend onthecellularcontextinwhichtheyareexpressed(inthiscase mousehepatocytes).Howeverthatmaybe,atthepresenttimeit remainsuncertainwhetherornottheobservedclinical pharma-cokineticinteractionbetweensorafenibanddocetaxelismediated throughOATP1B1,orratherbysomeothermechanism(s).

3. Hepatocytehoppingofaconjugatedanticancerdrug:

Thecaseofsorafenibglucuronide

In contrast to the nearly absent effect of Oatp1b2 or Oatp1a/1bdeficiencyonthepharmacokineticsoforally adminis-teredsorafenib,inthesameexperimentsapronouncedincrease in plasma levels of sorafenib glucuronide was observed, with a 5.5-fold increased AUC in Oatp1b2−/− mice, and 29-fold in Oatp1a/1b−/−mice;althoughthebiggerrelativeincreaseinthe lat-tercasemainlyarosefromasubstantiallylowerAUCofsorafenib glucuronideinwild-typeFVBmicethaninwild-typeDBAmice, withtheplasmaAUCsinboththeknockoutstrainsbeingsimilar (Zimmerman et al., 2013). The plasma levels of sorafenib glu-curonidein theknockoutstrainsequaled orsurpassed thoseof parentalsorafenib itself, indicating thequantitativeimportance of this process. Subsequent invitro experiments indicated that sorafenibglucuronidewasefficientlytakenupbymouseOatp1b2 and humanOATP1B1 and OATP1B3expressed inHEK293 cells. Furthermore,transgenicexpressionofeitherhumanOATP1B1or OATP1B3intheliverof“humanized”Oatp1a/1b−/−miceresulted inapartialreversaloftheincreasedplasmasorafenibglucuronide levels(Zimmermanetal.,2013).Thisconfirmedaprominentrole ofthemouseandhumansinusoidalOATPtransportersinhepatic uptakeofsorafenibglucuronide.

An important question wasthe origin of the sorafenib glu-curonide,asit isknownthat both enterocytesandhepatocytes can have substantial drug glucuronidation capacity, and oral sorafenib might undergo rapid glucuronidation in the intesti-nalwalluponabsorption.However,mouseintestinalmicrosomes showedonlymarginal sorafenibglucuronidationcapacity, com-paredtomouselivermicrosomes(>50-folddifference),suggesting that most sorafenib glucuronide had been formed in the liver (Zimmerman et al., 2013). Taken together, these findings sug-gestedstronglythatundernormalconditionsasubstantialpartof sorafenibglucuronideformedintheliverisextrudedacrossthe sinusoidalmembraneintotheblood,andthentakenupagaininto theliverbyOatp1b2,OATP1B1,andOATP1B3.

3.1. Hepatocytehoppingofbilirubinglucuronide

This process inferred to explain the behavior of sorafenib glucuronidewashighly reminiscent ofthepreviously proposed “hepatocyte hopping” process for the endogenous metabolite bilirubinglucuronide(Iusufetal.,2012c;vandeSteegetal.,2012, 2010).The hydrophobic,very poorly water-soluble,and poten-tiallyhighlytoxicunconjugatedbilirubin,theprimarybreakdown

Şekil

Fig. 1. Comparison of the human and mouse OATP1A/B/C gene clusters. The human OATP1A/B/C gene cluster is located on chromosome 12p
Fig. 2. Structures of anticancer drugs clearly affected by Oatp1a/1b activity in vivo
Fig. 3. Impact of mouse and human OATP1A/1B transporters on the plasma and liver disposition of various anti-cancer drugs
Fig. 4. Schematic diagram of hepatocyte hopping of sorafenib glucuronide and possible recirculation of sorafenib

Referanslar

Benzer Belgeler

Millî marş temposu o hafız ağzı ses cilveleri arasında şöyle böyle belirir gibi oluyordu.. Medet, aman, yar yar, hey gibi san’ at inceliklerini de katsaydı,

The Hacı Bektash Veli Ocak, is at the top of the ocak hierarchy in the Seyyid Ali Sultan Ocak, sürek of the Çamlıca Region.. Ercan Ordukaya (1983) and his spouse,

This work contributes to the development of the Regional Security Complex theory by providing evidence of the important role played by the United Nations Office for

Oysa, ~ngi- lizler tüm alanlarda faaliyet gösteriyorlar ve Vikers Arm-strong veya Brassert gibi büyük firmalar birinci s~n~f teknisyenlerini de içeren heyetleri

Ardında daha o zaman büyük bir askerî mazi bulunan Mustafa Kemal, 1920 de Ankara’da yerleştiği ve Büyük Millet Meclisini teşkil ettiği zaman, yalnız bir

Since research regarding the effect of portfolio application on young learners in L2 writing classes in the Turkish context is scarce, the purpose of the study is to explore the

Valinin bu nazik zi­ yaretine kurucumuz Habib Edib Törehan kısa bir hitabe ile teşek­ kür etmiş, V ali de bu hi­ tabeye mukabelede bulunarak basını daime bir

Buna göre primer karaciğer, dalak ve mezenterik kist hidatik- lerin yırtılması sonucu, kist içeriği batın içine yayılmakta, daha sonra primer kistler teşhis yöntemleri