• No results found

An Accessible and Unique Insight into Metastasis Mutational Content Through Whole-exome Sequencing of Circulating Tumor Cells in Metastatic Prostate Cancer

N/A
N/A
Protected

Academic year: 2021

Share "An Accessible and Unique Insight into Metastasis Mutational Content Through Whole-exome Sequencing of Circulating Tumor Cells in Metastatic Prostate Cancer"

Copied!
11
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

An

Accessible

and

Unique

Insight

into

Metastasis

Mutational

Content

Through

Whole-exome

Sequencing

of

Circulating

Tumor

Cells

in

Metastatic

Prostate

Cancer

Vincent

Faugeroux

a,b

,

Ce´line

Lefebvre

a

,

Emma

Pailler

a,b

,

Vale´rie

Pierron

c

,

Charles

Marcaillou

d

,

Se´bastien

Tourlet

c

,

Fanny

Billiot

b

,

Semih

Dogan

a

,

Marianne

Oulhen

b

,

Philippe

Vielh

b

,

Philippe

Rameau

b

,

Maud

NgoCamus

e

,

Christophe

Massard

e

,

Corinne

Laplace-Builhe´

b

,

Arian

Tibbe

f

,

Me´lissa

Taylor

b

,

Jean-Charles

Soria

a,e

,

Karim

Fizazi

a,e

,

Yohann

Loriot

a,e,1

,

Sylvia

Julien

c,1

,

Franc¸

oise

Farace

a,b,1,

*

aINSERMU981,InstitutGustaveRoussy,UniversitéParis-Saclay,Villejuif,France;bINSERMUS23AMMICA,InstitutGustaveRoussy,UniversitéParis-Saclay, Villejuif,France;cIPSEN-Innovation,LesUlis,France;dIntegraGenSA,Evry,France;eDepartmentofCancerMedicine,InstitutGustaveRoussy,Université Paris-Saclay,Villejuif,France;fVyCAPBV,Deventer,TheNetherlands

a v a i l ab l e a t w w w . s c i e n c e d i r e c t . c o m j o u r n a l h o m e p a g e : e u o n c o l o g y . e u r o p e a n u r o l o g y . c o m Articleinfo Articlehistory: AcceptedDecember7,2018 AssociateEditor: PaulNguyen Keywords:

Circulatingtumorcells Liquidbiopsy

Prostatecancer

Whole-exomesequencing

Abstract

Background: Genomicanalysisofcirculatingtumorcells(CTCs)couldprovidea uniqueandaccessiblerepresentationoftumordiversitybutremainshinderedby technicalchallengesassociatedwithCTCrarityandheterogeneity.

Objective: ToevaluateCTCsassurrogatesamplesforgenomicanalysesin meta-staticcastration-resistantprostatecancer(mCRPC).

Design,setting,andparticipants: Threeisolationstrategies(filterlaser-capture mi-crodissection,self-seedingmicrowellchips,andfluorescence-activatedcellsorting) weredevelopedtocaptureCTCswithvariousepithelialandmesenchymalphenotypes andisolatethematthesingle-celllevel.Whole-genomeamplification(WGA)andWGA qualitycontrolwereperformedon179CTCsamples,matchedmetastasisbiopsies,and negativecontrolsfrom11patients.Allpatientsbutonewerepretreatedwith enzalu-tamideorabiraterone.Whole-exomesequencing(WES)of34CTCsamples,metastasis biopsies,andnegativecontrolswereperformedforsevenpatients.

Outcome measurements and statistical analysis: WES of CTCs was rigorously qualified in terms of percentage coverage at 10 depth, allelic dropout, and uncoveredregions.SharedsomaticmutationsbetweenCTCsandmatched metas-tasisbiopsieswereidentified.Acustomizedapproachbasedondeterminationof mutationratesforCTCsampleswasdevelopedforidentificationofCTC-exclusive mutations.

Resultsandlimitations: SharedmutationsweremostlydetectedinepithelialCTCs andwererecurrent.Fortwopatientsforwhomadeeperanalysiswasperformed,a fewCTCswere sufficient torepresent halfto one-thirdofthe mutationsinthe

1 Theseauthorscontributedequallytothiswork.

*Correspondingauthor.InstitutGustaveRoussy,Universite´ Paris-Saclay,114rueE´douardVaillant, F-94805Villejuif,France.Tel.:+33142115198.

E-mailaddress:francoise.farace@gustaveroussy.fr(F.Farace).

https://doi.org/10.1016/j.euo.2018.12.005

2588-9311/©2020TheAuthors.PublishedbyElsevierB.V.onbehalfofEuropeanAssociationofUrology.Thisisanopenaccessarticle undertheCCBY-NC-NDlicense(http://creativecommons.org/licenses/by-nc-nd/4.0/).

(2)

1. Introduction

Advances in the development of precision medicine for cancer patients rely on accurate identification of the genomicfeaturesunderlyingapatient’stumor[1].Asthe numberofgenomictargetswithmatchedtherapiesgrows, thecurrenthurdleremainstheavailabilityoftumortissue thatcanreflectaconstantlyevolvingdisease[2–4]. Recog-nition thatmetastatic lesions haveadiscordantgenomic fingerprint compared to primary tumor has led to recommendations for invasive biopsies. However, tumor heterogeneity poses the predicament that single-site biopsiesmaynotcapturethegenomicalterationsrelevant totargetedtherapy[5].Thisisespeciallytrueinmetastatic prostatecancer(mPC),forwhichbonemetastasesarising severalyearsafterdiagnosisandtherapeuticinterventions are either inaccessible or yield insufficient material for genomicprofilingofthedisease[6,7].Inaddition,biopsies of bone metastases are painful and cannot be repeated multipletimes during thedisease course, calling for less invasivemethodsformolecularcharacterizationofmPCand formonitoringdiseaseprogressionduringtherapy.

The development of “liquid biopsies” presents new opportunitiesfornoninvasivemonitoringofclonal heteroge-neity. Circulating tumor cells (CTCs) captured as a “liquid biopsy”arecurrentlyregardedasanoninvasiveand repeat-able source of tumor material that could overcome the samplingchallengesformetastaticdisease[8–11].CTCsare likelytoarisefromdistinctmetastaticsites,andmaybetter representtumorheterogeneityinbothspaceandtime.Liquid biopsiesmight allowgenomiccharacterizationofmPCand routinemonitoringofmetastaticspread,drugresistance,and diseaserelapseduringthecourseoftreatment[9–11]. How-ever,itisstill unknownwhether using liquidbiopsies can provideamorecompleteprofileofmPCclonaldiversity.

CTCs often exhibit combinations of epithelial and mesenchymal traits, highlighting the role of epithelial-mesenchymaltransition(EMT)intheprocessof intravasa-tion and cancer cell dissemination [12]. Therefore, non-epithelial-basedCTCenrichmentmethodsareessentialto capturethephenotypicheterogeneityofCTCsinterms of EMTmarkerexpression[13–15].Reportsofsingle-cell high-dimensional analyses of CTCs are scarce and limited to

EpCAM-positive CTCs [16–18]. The rarity and biological heterogeneityofCTCshaveimposedtechnicalchallengesto their isolation and analyses at single-cell level, and impacted the success of robust processing of complex andcostlydownstreammethodologies.Onceisolatedatthe single-celllevel,CTCsmustundergowhole-genome ampli-fication (WGA), a mandatory process to identify CTC somaticvariantsbutpronetoamplificationbias, polymer-aseerrors,andallelicdropout(ADO)[19].

Ourcentralhypothesiswasthatthegenetic heterogene-ityofCTCsassessedatthesingle-celllevelviawhole-exome sequencing(WES)reflectstheirphenotypicheterogeneity in mPC and may help to resolve the clonal relatedness betweenCTCsandmetastasisinmPC.Here,wereportfor thefirsttimeisolationatthesingle-celllevel,qualification, andWESofCTCs harboringvariousepithelial, mesenchy-mal,and/ormorphologicalcharacteristicsfrom11patients withmetastaticcastration-resistantPC(mCRPC).AllmCRPC patients were included in the MOSCATO01 clinical trial evaluating the potential clinical benefit of screening metastasisbiopsiesviahigh-throughputgenomicanalyses toidentifyactionablealterationsinpatientswithadvanced cancer [1]. Somaticmutations sharedbetween CTCs and matchedmetastaticbiopsies,andCTC-exclusivemutations (exclusivelyidentifiedinCTCsandnotinmatchedbiopsies) were comprehensively explored. Our results reveal that CTCs can be used as surrogate samples for mutational analyses of mCRPC and provide a genomic picture of disseminatingclonesinvolvedinmetastasis.

2. Patientsandmethods

2.1. Patients

PatientswithmCRPCwereenrolledintheMOSCATO01prospectivetrial (IDRCB2010-A00841-40;NCT02613962)[1].Thestudywasauthorized bytheFrenchnationalregulationagencyANSMandapprovedbythe ethicscommittee.BloodwascollectedintoEDTAandCellSavetubes.

2.2. CTCenrichment,detection,andisolation

Isolationbysizeofepithelialtumorcells(ISET)filtration,immuno fluo-rescencestaining,andscanningoffilterswereperformedaspreviously

matchedmetastasisbiopsy.CTC-exclusivemutationswereidentifiedinboth epi-thelialandnonepithelialCTCsandaffectedcytoskeleton,invasion,DNArepair,and cancer-drivergenes.Some41%ofCTC-exclusivemutationshadapredicted delete-rious impact on protein function. Phylogenic relationships between CTCs with distinctphenotypeswereevidenced.

Conclusions: CTCscanprovideuniqueinsightintometastasismutationaldiversity andrevealundiagnosedgenomicaberrationsinmatchedmetastasisbiopsies.

Patient summary: Our results demonstrate the clinical potential of circulating tumorcellstoprovideinsightintometastaticeventsthatcouldbecriticaltotarget usingprecisionmedicine.

©2020TheAuthors.PublishedbyElsevierB.V.onbehalfofEuropeanAssociationof Urology.ThisisanopenaccessarticleundertheCCBY-NC-NDlicense( http://creati-vecommons.org/licenses/by-nc-nd/4.0/).

(3)

reported [20].CTCs and CD45-positivecells were isolated via laser microdissection. CTCs were detected using CellSearch as previously described [10,20] and isolated using self-seeding microwell chips [21]. Enrichment via RosetteSep was performed according to the manufacturer’s protocol(StemCell Technologies,Vancouver,Canada). RosetteSep-enrichedCTCswerepermeabilized,stainedviaimmuno flu-orescence,andisolatedusingfluorescence-activatedcellsorting(FACS). ThemethodsaredescribedintheSupplementarymaterial.

2.3. WGA,qualitycontrols,andpurification

WGAwasperformed usingan Ampli1WGAkitversion1 (Menarini SiliconBiosystems,SanDiego,CA,USA)andevaluatedviaaqualitative polymerasechainreaction(qPCR)assaytodetermineaqualityscore(QS; Supplementarymaterial).

2.4. IsolationofgenomicDNAfrombloodandtumorbiopsies DNAfromformalin-fixed,paraffin-embeddedtumorbiopsyspecimens andconstitutionalDNAwerepurifiedasdescribedintheSupplementary material.

2.5. WESandbioinformaticsanalysis

WES and bioinformatics analysis are described in detail in the Supplementarymaterial.RawdataareavailableonEGAataccession numberega-box-1082.

3. Results

Eleven patients were included in the current analysis (Table 1).At the time of CTC collection, allpatients had metastasesfromCRPCandallbutonehadbeenpreviously treated with either abiraterone or enzalutamide, while threehadbeentreatedwithpriordocetaxel.

WeinitiallydevelopedaCTCenrichmentstrategybased onfiltrationbyISETbeforeamultistepprocessthatincludes immunofluorescentstainingwithepithelialand mesenchy-malmarkersforCTCidentificationandlaser microdissec-tion of individualCTCs (Fig.1A). CTCs fromfive patients (P1–P5)were isolatedinthis way.We detected veryfew

epithelialCTCs,especiallyintwoofthepatients(P2andP3), compared tohigher CellSearch counts,asconfirmedbya recentstudy [13] (Supplementary Table 1). In agreement withpublishedresults[15,22],ISETrevealedhybridcells,as wellaspopulationsoflarge(nuclei 16

m

m)and mesen-chymal candidate CTCs, of possible interest for further analysis by WES (Fig. 1B). We developed two other strategiesto captureepithelial andothercandidateCTCs. In the second strategy, CTCs were enumerated via CellSearchandindividuallyisolatedusingtheself-seeding microwellchipsplatform[21](Fig.1A).Ourthirdapproach involvedhematopoieticblood-celldepletionandindividual CTC isolation via FACS (Fig. 1A). These three strategies allowed identification of five phenotypic subtypes of candidate CTCs on the basis of both epithelial and mesenchymal marker expression and/or morphological characteristics (Fig. 1B). These initial results led us to explore and isolate CTCs in six more mCRPC patients (Table1andSupplementaryTable1).Aglobalworkflowwas establishedbasedonthesestrategies,WGAand qualifica-tion, and WES of CTCs and matched negative controls (CD45+ cells; Fig. 1C). We hypothesized that pools of a limitednumberofindividualCTCscouldresultin higher-qualityWGAandcouldyieldhighersensitivityforvariant detectionthansingleCTCs.Weisolated162poolsoftwoto ten CTCs (each pool containing CTCs of the same phenotype),17singleCTCs,andmatchedCD45+cells.The

WGAqualitywascontrolledusingaqPCRassay,andaQS3 was set as the criterion for WES eligibility, which was reached by 87/179 WGA samples (49%; Supplementary Table2,SupplementaryFig.1AandB).Ashypothesized,the probabilityofreachingQS3washigherinpoolsthanin singleCTCs(Fig.1D).Amongthe87WGACTCsampleswith QS3,31fromsevenpatientswereselectedforWES.Two supplementary epithelial CTC pools (P7-E-7 and P7-E-8) from10CTCsandasingleCTCbearingahybridphenotype, allwithQS<3,wereincludedforpatientP7toaddressCTC heterogeneity(SupplementaryTable3).WESforthe34CTC sampleswassubjectedtorigorousqualitycontrol(Fig.1E,

SupplementaryFig.1).Ofthe34WESsamples,28(82%)had

Table1Clinicalcharacteristicsofpatientswithmetastaticcastration-resistantprostatecancer

Case Age(yr) TNMa Metastaticsites PSAb(ng/ml) Gleasonscore TTB(mo) Biopsysite Previoustreatment

CTx CTN ENZ ABI

P1 77 TxNxM1 BonesandLNs 333 7(4+3) 46 LN No Yes No Yes

P2 77 T1N0M0 BonesandLNs 101 7(3+4) 32 LN No Yes No Yes

P3 73 T3N0M0 LNs 249 7(4+3) 22 LN No Yes No Yes

P4 46 TxN0M1 Bonesandliver 85 8 14 Liver No Yes No Yes

P5 68 T3N0M0 LNs 14 9 24 LN Yes Yes No Yes

P6 65 T3N0M0 LNs 283 8(4+4) 13 LN No Yes No Yes

P7 60 T3N0M0 BonesandLNs 1715 7(4+3) 26 LN Yes Yes Yes Yes

P8 66 T3N0M0 BonesandLNs 51 9 2 LN No Yes No Yes

P9 67 TxN1M1 LNsandbones 27 8(4+4) 12 LN Yes Yes No No

P10 71 TxNxM1 Bones,liver,andLNs 10 NA 72 Liver No Yes Yes No

P11 76 T3N0M0 Bones 163 NA 24 Bone No Yes No Yes

LN=lymphnode;PSA=prostate-specificantigen;NA=notavailable;TTB=timefrommetastaticprostatecancerdiagnosistobiopsy;CTx=chemotherapy; CTN=castration;ENZ=enzalutamide;ABI=abiraterone.

a

Atdiagnosis.

b

(4)

Fig.1–ExperimentalprocessforWESofCTCsatthesingle-celllevel.(A)Schematicofthethreeexperimentalstrategiesusedforenrichment, detection,andisolationofCTCsatthesingle-celllevel.(B)NomenclatureforCTCcandidatesaccordingtoepithelialandmesenchymalmarker expressionandtheexperimentalstrategyusedforisolation.EpithelialCTCs(E)wereCD45-negativeandpositiveforEpCAMandpan-cytokeratin(green staining)andHoechst33342(bluestaining).HybridCTCs(H)wereCD45-negativeandpositiveforEpCAMandpan-cytokeratin,vimentin(redstaining), andHoechst33342.LargeCD45-negative,Hoechst33342-positivecells(L)withanucleus16mmandepithelialandmesenchymalexpressionbelow thethresholdoftheassaywereselectedforWESonthebasisofcytomorphologicalfeaturesreportedinpreviousstudies[20].Largemesenchymal CD45-negative,Hoechst33342-positivecells(LM)withanucleus16mmandvimentinexpressionwereselectedforWESonthebasisof

cytomorphologicalfeatures.CD45-negative,Hoechst33342-positivesmallcellcandidates(S)withcytokeratinexpressionbelowthethresholdofthe assayandcontaininganucleusmorphologicallydistinctfromsurroundingCD45-positivecellsweretestedviaWES.(C)Globalworkflowfor sequencingofmetastasisbiopsies,individualCTCs,andCD45+

controlcells.Somaticmutationswereidentifiedaccordingtofiltersdescribedinthe

Supplementarymaterial.Commonsingle-nucleotidepolymorphisms(SNPs),patientSNPs,andvariantscommonbetweenCTCsandpairedCD45+cell

sampleswereremoved.(D)PercentageofWGAproductswithQS3accordingtothenumberofCTCspersample.CTCsampleswereclassified accordingtothenumberofCTCspersample.ThepercentageWGAwithQS3wasdependentonthenumberofCTCspersample.(E)Coverageat10T sequencingdepthofthe34selectedCTCsamplesfromtheWESexperimentalset.ThenomenclatureforCTCsisdetailedinSupplementaryTable4.Of the34CTCsamples,27(79%)hadmorethan50%coverageat10T.Themeanpercentagecoverageat10Tis54.3%(range12–71%).Thelower percentagecoverageat10TwasbecauseofWGAwithQS<3.WES=whole-exomesequencing;CTCs=circulatingtumorcells;WGA=whole-genome amplification;QS=qualityscore;ISET=isolationbysizeofepithelialtumorcells.

(5)

<30%ADOanduncoveredregions,and27(79%)had>50% coverage at 10 depth (10 was the minimum level of coverageconsidered,butcoveragewasinpracticetypically inexcessof10).

We analyzed mutations shared between CTCs and matched metastasis biopsy samples. Among the 34 CTC samplessequenced,15fromfourpatients(P1,P7,P10,P11) sharedmutationswiththecorrespondingmatchedbiopsy (Fig. 2A). A total of136 sharedmutations for 65 unique mutationsweredetected(SupplementaryFigs.2and3A).Of these65,61(93.9%)weredetectedexclusivelyinepithelial CTCs, while four (6.1%) were detected in CTCs bearing variousphenotypicalcharacteristics(Fig.2B).Forpatients P7andP11,forwhomalargernumberofCTCsampleswith various characteristics were sequenced, 59% and 33%, respectively, of mutations detected in biopsies were identifiedinCTCs(Fig.2C).ForpatientP7,sharedmutations were mostly identified in two CTC pools with high QS (Fig.2D).ForpatientP11,31%ofmutationssharedwiththe matchedbiopsywerepresentinonepooloffive andone singleepithelial CTC with QS 5 (SupplementaryFig. 4). Mostofthesharedmutations,includingthoseinGRM8and TP53, were identified in several epithelial CTC samples (Table2,Fig.2B,SupplementaryTable4).Overall,24/53of shareduniquemutations(45%)hadapredicteddeleterious impacton proteinfunction. Ourresultsdemonstrate that WESofasmallnumberofpoolsorsingleepithelialCTCs, selectedusingarigorousqualificationprocess,can reveal significantmutationalsimilaritiestothemetastasisbiopsy. Shared mutations were recurrent in CTCs, suggesting a possibleimportantroleinmetastaticspread.

ToreliablyidentifysomaticCTC-exclusivemutations,we excluded variants shared between CTCs and matched biopsies and established a customized approach that involves variant classification according to recurrence in CTC samples (Supplementary Fig. 3B). We determined a mutation rate per Mb per CTC sample for criteria of observation in one,two, or three samples, as shownfor patientP11inFig.3A.MutationsidentifiedinCTCsamples withamutationrateperMboflessthanthatformetastasis biopsies were considered as CTC-exclusive mutations. Among the 240 variants identified in two CTC samples, 31 were known mutations with a predicted deleterious impact on protein function, including variants in cancer genessuchasHSP90AB1andKDM5B(SupplementaryFig.2, SupplementaryTable5).Whenvariantswererequiredtobe observedinthreeCTCsamples,themutationrateperMb per samplewaslessthan values for matchedmetastases (Fig.3A).Nineteensomaticvariantswerepresentinatleast threeCTCsamples,ofwhichtenvariantswerereportedin databases(Table2).Sevenofthe19(37%)wereidentifiedin genesinvolvedincytoskeleton(eg,MACF1)orinvasion(eg, NEDD9)andimpactproteinfunction.ForpatientP11,20%of CTC-exclusivevariantswereidentifiedinatleastthreeCTC samples(Fig.3B). Whenweexaminedthedistribution of these19exclusivevariantsaccordingtoCTCphenotype,11 (58%)weredetectedexclusivelyinepithelialCTCs,andeight (42%) were presentin CTCs harboring various character-istics (Fig. 3C). These data demonstrate the existence of

exclusive mutations in CTCs that were not detected in matchedbiopsies.

ToevaluatetherelationshipbetweenCTCswithdistinct characteristics in patients P7 and P11, we performed hierarchicalclusteringforsharedandCTC-exclusive muta-tionsobservedinatleastthreeCTCsamples(Fig.4AandB). The existenceof common mutations between CTCs with distinct characteristics supports their phylogenetic rela-tionshipbutdivergentevolution.

4. Discussion

We present the first genomic analysis atthe single-cell level of CTCs with various phenotypical characteristics. Our primary aim was to determine the feasibility of noninvasivemutationalcharacterizationofmPCpatients and todefine whether the mutation landscape of CTCs reflected thatofmatchedmetastases.Wedemonstrated thatWESofCTCsisfeasiblebytestingdifferentstrategies that enabled us to detect CTCs with multiple cellular phenotypes.Ourfirst CTCisolation strategyfellshortof identifying a fraction of epithelial CTCs, as recently confirmed[14].Thisledustodeveloptwosupplementary enrichment and isolation strategies that were used in conjunction with filtration and laser-capture microdis-sectiontoaddressthechallengeofCTCheterogeneity.Our results show the superiority of strategies 2 and 3 for futureclinicalapplicationsbutdonotallowustoidentify whichis the best. We determinedaglobal workflow to generatehigh-qualitylibrariesforWESinwhichrigorous qualification of WGA uniformity could reliably predict which CTC pool or single CTC was most likely toyield high-quality WES data. Our results differ from those reported by Lohretal. [16], whofoundthat ADO anda lackofsystematiccoverageinWGAproductsfromsingle EpCAM-positiveCTCsnecessitatedWESofmultiplesingle CTCs(n=19)forvariantdetermination.Althoughperhaps less pertinent for exploring tumor heterogeneity, the approach presentedhere,whichlimitedWEStoasmall numberofhigh-qualityCTCsamplesandintegratedCTC pools,istechnicallymorepracticalandcost-effectivefora clinicalsetting.

WeshowedthatWESforaverysmallnumberofpoolsor singleepithelialCTCsyieldedsignificantmutational similari-tiesfortheexomeofmatchedmetastasisbiopsies,including mutations inPCgenessuchasGRM8andTP53.Ourresults indicatethatCTCsequencing canprovideareasonableand feasibleproxyformetastasissamplingindisseminatedcancer. Interestingly,recurrentmutationsweresystematically iden-tified in both CTCs and matched metastasis biopsies, and sequencing more CTCs did not significantly extend the number of mutations identified. However, a fraction of mutations identifiedwithinthematchedmetastasisbiopsy werenotdetectedinCTCs,suggestingthatthesemutations maynothavetheabilitytodisseminateortransitinblood.By analogy, it could be hypothesized that these recurrent mutationsfoundinCTCsandsharedbymatchedmetastasis biopsies could play an important role in metastatic

(6)

progression.Alternatively,wecannotexcludethepossibility thatmutationsidentifiedwithinamatchedmetastasiscould bemissedinCTCsfordifferenttechnicalreasonsrelatedtothe sensitivityofourapproach.

Weestablishedastrictpipelineforvariantcallingandfor greaterreliabilityweonlyconsideredCTC-exclusive muta-tionsobservedinatleastthreeCTCsamples.CTC-exclusive mutationsincludedbothunknownmutationsthatwarrant

Fig.2–CharacterizationofvariantssharedbymetastasisbiopsiesandCTCs.(A)NumberofvariantssharedbymetastasisbiopsiesandCTCs.Fifteen CTCsamplesfromfourofthesevenpatientssharedatotalof136variantswithmatchedmetastasisbiopsysamples,ofwhich65wereunique.Foreach patientthesumofvariantssharedbytheirmetastasisbiopsyandCTCsisrepresentedbyhatchedbars.(B)Distributionofshareduniquevariants accordingtoCTCphenotype.Sixty-onevariants(93.9%)weredetectedinepithelialCTCs(E),twovariants(3.1%)wereonlydetectedinlargeCTCs(L), onevariant(1.5%)wasdetectedinbothinepithelialandlargeCTCs(E+L),andonevariant(1.5%)wasdetectedinepithelial,large,andhybridCTCs(E +L+H).(C)PercentageofvariantssharedbyCTCsandmetastasisbiopsies,oronlypresentinmetastasisbiopsies,forpatientsP7andP11.(D)Heatmap ofvariantsidentifiedinthemetastasisbiopsyandsharedbyCTCsinpatientP7.Some41%ofvariantswereexclusivelyidentifiedinthemetastasis biopsyand59%weresharedwithCTCs.TheP7-E-10pool(QS=7)exhibited51%sharedmutationswiththematchedbiopsy.ThepoolsP7-E-10and P7-E-9(QS=6)hadalmostthesamemutationsandshared54%ofmutationswiththematchedbiopsy.Thepositionsofvariantspresentinthemetastasis biopsyandnotcoveredinCTCsareshown.CTCs=circulatingtumorcells;QS=qualityscore.

(7)

furtherinvestigationandknownmutationswithapredicted deleterious impact on protein function in cancer-driver genessuchasHSP90AB1andKDM5B,aswellasingenesthat might have acrucial rolein thebiological specificity (eg, cytoskeletalremodeling)orsurvival(eg,DNArepair)ofCTCs. These CTC-exclusive mutations were mostly present in epithelialCTCsorinbothepithelialCTCsandCTCswithother morphologicalcharacteristics,aresultthatindirectly vali-datesthetruetumororiginofthesecells.Ourdatasuggest thatCTCsharboringtheseexclusivemutationsderivedfrom distinctmetastaticsites representedminorsubclonesthat wereundetectableinmetastaticbiopsies.

Whiletumortissuebiopsiesremainthegoldstandardfor validating or confirming genetic abnormalities found in CTCsorcirculatingtumorDNA(ctDNA),itisimportantto question the representability of a needle or single-site biopsy specimen. As illustrated here via analysis of CTC-exclusive variants,CTCs canbederived from lesionsthat were not biopsied and may contain a different genetic compositionthanthetumormaterialusedasareference.

Theseresultsdemonstrateforthefirsttimethat sequenc-ing of CTCs at the single-cell level can reveal metastasis mutationalcontentanddiversitythatareotherwise inacces-sible. Exome sequencing of cell-free tumor DNA has also

Table2–ExamplesofvariantssharedbetweenCTCsandmatchedmetastaticbiopsies,andCTC-exclusivevariants

Gene Transcript Database Somaticsingle-nucleotide

variants Prediction (Polyphen2Sift) Numberof CTCsamples PID

dbSNP COSMIC Codonchange AAchange

VariantssharedbyCTCsandmatchedmetastaticbiopsies

GRM8 NM_000845.2 - - c.2207C>T p.Ala736Val D,T 4(P11-E-4c ;P11-E-5b ; P11-E-7b ;P11-E-8b ) P11

TP53 NM_000546.5 - COSM10656 c.742C>T p.Arg248Trp D,D 4(P11-E-4c

;P11-E-5b

; P11-E-7b;P11-E-8b)

P11

PTEN NM_000314.4 - - c.236C>G p.Ala79Gly B,NA 1(P7-E-10c

) P7

CTC-exclusivevariantsobservedinatleast3CTCsamples

PPAPDC1A NM_001030059.1 c.301_305delATTAA p.Ile101fs NA,NA 3(P7-L-2a

;P7-LM-3a ; P7-LM-5a) P7 INO80 NM_017553.2 c.2867C>T p.Pro956Leu B,T 3(P7-L-1a ;P7-L-2a ; P7-LM-3a ) P7 C3orf30 NM_152539.2 rs199919487 c.1369G>A p.Glu457Lys D,D 4(P11-L-2a ;P11-E-4c ; P11-E-5b;P11-E-6b) P11 ANKRD32 NM_032290.3 c.2992G>A p.Glu998Lys B,T 4(P11-L-2a ;P11-E-3c ; P11-E-5b ;P11-E-7b ) P11 EIF4G3 NM_001198801.1 c.2317C>A p.Gln773Lys B,T 3(P11-L-2a ;P11-E5b ; P11-E-8b ) P11

OTUD4 NM_001102653.1 c.1575A>C p.Leu525Phe P,T 3(P11-L-2a

;P11-E-4c

; P11-E-8b

)

P11

ZFYVE1 NM_021260.3 COSM470205 c.488C>A p.Thr163Lys B,T 3(P11-L-2a;P11-E-3c;

P11-E-6b)

P11

CELA3B NM_007352.2 rs202129706 c.803C>A p.Ala268Glu B,T 5(P11-E-3c

;P11-E-4c ; P11-E-5b ;P11-E-6b ;P11-E-8b ) P11

PGBD2 NM_170725.2 c.441_442insT p.Phe149fs NA,NA 4(P11-E-4c

;P11-E-5b

; P11-E-7b;P11-E-8b)

P11

MACF1 NM_012090.5 rs202091916 COSM128604 c.15886A>C p.Thr5296Pro D,NA 4(P11-E-5b

;P11-E-6b ; P11-E-7b ;P11-E-8b ) P11

ABCB11 NM_003742.2 rs200687717 c.1628A>C p.Asp543Ala B,T 3(P11-E-4c

;P11-E-5b ; P11-E-7b ) P11 QPCT NM_012413.3 c.152C>T p.Ser51Leu B,D 3(P11-E-5b ;P11-E-7b ; P11-E-8b ) P11

FAM161A NM_001201543.1 rs200331923 c.1573C>A p.Gln525Lys B,T 3(P11-E-5b;P11-E-7b;

P11-E-8b

)

P11

NEDD9 NM_001142393.1 c.1504A>C p.Thr502Pro D,T 3(P11-E-5b

;P11-E-6b

; P11-E-8b

)

P11

AKNA NM_030767.4 rs200970909 c.3287A>C p.His1096Pro P,D 3(P11-E-5b;P11-E-6b;

P11-E-8b) P11 C2CD3 NM_001286577.1 rs200719890 c.5233T>G p.Tyr1745Asp D,D 3(P11-E-5b ;P11-E-7b ; P11-E-8b ) P11

IFNGR2 NM_005534.3 c.100C>G p.Gln34Glu B,T 3(P11-E-5b;P11-E-7b;

P11-E-8b

)

P11

LANCL2 NM_018697.3 rs201180232 COSM453253 c.953T>G p.Val318Gly D,D 3(P11-E-6b

;P11-E-7b

; P11-E-8b

)

P11

NDUFV2 NM_021074.4 rs72935225 c.401T>C p.Val134Ala B,T 3(P7-E-9c;P7-E-10c;

P7-H-11c

)

P7

CTC=circulatingtumorcells;AA=aminoacid;PID=patientidentity.

a Isolatedusingstrategy1. b

Isolatedusingstrategy2.

c

(8)

Fig.3–CharacterizationofCTC-exclusivevariants.(A)MutationrateperMbinCTCsfrompatientP11forcriteriaofvariantsobservedinone,two,or threeCTCsamples.ForthecriterionofvariantsobservedintwoCTCsamples,themutationratesperMbpersamplewereclosetothevaluesforthe correspondingmetastasis,althoughforsomeCTCsamples,thisvaluewasgreater,indicatingthatWGAerrorscouldbeconserved.Forthecriterionof variantsobservedinthreeCTCsamples,themutationratesperMbpersamplewerelessthanorequaltothevaluesformatchedmetastases.(B) HeatmapofCTC-exclusivevariantsinpatientP11.Of101CTCvariants,81(80.2%)weredetectedintwoCTCsamples,15(14.8%)inthreeCTCsamples, four(4%)infourCTCsamples,andone(1%)infiveCTCsamples.ThepositionsofvariantsnotcoveredinCTCsamplesareshown.(C)Distributionof uniqueCTC-exclusivevariantspresentinatleastthreeCTCsamplesaccordingtotheCTCsamplephenotype.Twenty-twouniqueCTC-exclusivevariants weredetectedinatleastthreeCTCsamples;55%hadanexclusivelyepithelialphenotype,while45%haddifferentphenotypes.CTCs=circulatingtumor cells;WGA=whole-genomeamplification.

(9)

demonstrated concordance of mutations with metastasis biopsies [23]. Although ctDNA analyses may indeed offer advantages interms ofsimplicity, sequencing of CTCs can provideunique additional and complementary information such as EMT status and cancer stem-cell phenotype

[24,25]. The presence of different and multiple genetic

alterations canbe identified withinthe same CTC,offering thepossibilitytoexploreinter-andintratumorheterogeneity andevolution.ItisthereforeanticipatedthatCTCsandctDNA arecomplementaryintheirclinicalutility.Whiletumortissue biopsies remain the gold standard for confirming genetic abnormalitiesfoundinCTCs,itisimportanttoquestionthe representabilityofaneedleorsingle-sitebiopsy.Ourfindings emphasizethepotentialandclinicalutilityofdetectingsuch mutations in CTCs via minimally invasive blood draws for

optimaltherapyselection,precisionmedicine,andtreatment resistance options. A study quantifying digital pathology features ofCTCs recently showedthatlow CTCphenotypic heterogeneitywasassociatedwithbetteroverallsurvival(OS) among patients treated with androgen receptor signaling inhibitors, whereas highheterogeneity wasassociated with betterOSamongpatientstreatedwithtaxanechemotherapy

[26].Beyondidentifyingpotentialtargets,WESatthe single-celllevelcouldquantifyheterogeneityandhelpin decision-makingformCRPCpatients.

5. Conclusions

ThispioneeringstudyemphasizesthepotentialofCTCsto represent metastasis mutational content and tumor

Fig.4–HierarchicalclusteringofCTCsandmatched-metastasisbiopsies.(A)HierarchicalclusteringofCTCandmatchedmetastasisbiopsysamplesin patientP7accordingtomutationalsimilaritiesbetweenCTCsandthematchedmetastasisbiopsy,andbetweendifferentCTCsamplesforvariants observedinatleastthreesamples.Thenumberofsharedvariantsisrepresentedbytheredcolorgradientfromdarkred(highnumberofvariants)to white(nosharedvariant).(B)HierarchicalclusteringofCTCsandmatchedmetastasisbiopsyinpatientP11accordingtothesamerepresentation. CTCs=circulatingtumorcells.

(10)

diversitythatareotherwiseinaccessible.Byoffering real-time monitoring of a constantly evolving disease and detecting potentially critical mutations via minimally invasiveblooddraws,CTCsequencingcanserveanunmet needforoptimaltherapyselectionandprecisionmedicine.

Authorcontributions:FrançoiseFaracehadfullaccesstoallthedatain thestudyandtakesresponsibilityfortheintegrityofthedataandthe accuracyofthedataanalysis.

Studyconceptanddesign:Faugeroux,Massard,Fizazi,Loriot,Julien,Soria, Farace.

Acquisition of data: Faugeroux, Pailler, Marcaillou, Billiot, Oulhen, Rameau,Tibbe.

Analysisandinterpretationofdata:Faugeroux,Pailler,Pierron,Tourlet, Vielh,Loriot,Julien,Farace.

Draftingofthemanuscript:Faugeroux,Lefebvre,Taylor,Farace. Criticalrevisionofthemanuscriptforimportantintellectualcontent:Pailler, Pierron,Tourlet,Taylor,Soria,Fizazi,Loriot,Julien.

Statisticalanalysis:Lefebvre,Dogan.

Obtainingfunding:Pierron,Fizazi,Julien,Farace.

Administrative,technical,ormaterialsupport:Marcaillou,Billiot,Oulhen, NgoCamus,Laplace-Builhé.

Supervision:Farace,Loriot,Julien. Other:None.

Financial disclosures: Françoise Farace certifies that all conflicts of interest, including specific financial interests and relationships and affiliationsrelevanttothesubjectmatterormaterialsdiscussedinthe manuscript(eg,employment/affiliation,grantsorfunding, consultan-cies,honoraria,stockownershiporoptions,experttestimony,royalties, orpatentsfiled,received,orpending),arethefollowing:None. Funding/Supportandroleofthesponsor:Thisstudywassupportedby IPSEN-Innovation,VincentFaugerouxissupportedbyFondationpourla Recherche Médicale (no. FDT20160435543). Céline Lefebvre was supported by SIRIC Socrate. Emma Pailler was supported by LabEx LERMIT (no. ANR-10-LABX-0033-LERMIT) and Fondation pour la RechercheMédicale(no.FDT20150532072).Thesponsorsplayedarole inthedesignandconductofthestudy;dataanalysisandinterpretation; andpreparation,review,andapprovalofthemanuscript.

Conflictofinterestdisclosurestatement:Authorsdisclosenopotential conflictsofinterest.

Acknowledgments:Wethankthepatientsandtheirfamilies,aswellasE. MartinforfruitfuldiscussionandM.Letexierforhelpinbioinformatics analysis.

AppendixA. Supplementarydata

Supplementarydataassociatedwiththisarticlecanbe found,intheonlineversion,atdoi:10.1016/j.eururo.2018. 11.052.

References

[1] MassardC,MichielsS,FerteC,etal.High-throughputgenomicsand clinicaloutcomeinhard-to-treatadvancedcancers:resultsofthe MOSCATO01trial.CancerDiscov2017;7:586–95.

[2] CancerGenomeAtlasResearchNetwork.Themoleculartaxonomy ofprimaryprostatecancer.Cell2015;163:1011–25.

[3]RobinsonD,VanAllenEM,WuYM,etal.Integrativeclinical geno-micsofadvancedprostatecancer.Cell2015;161:1215–28.

[4]WedgeDC,GundemG,MitchellT,etal. Sequencingofprostate cancersidentifiesnewcancergenes,routesofprogressionanddrug targets.NatGenet2018;50:682–92.

[5]BoutrosPC,FraserM,HardingNJ,etal.Spatialgenomic heteroge-neity within localized, multifocal prostate cancer. Nat Genet 2015;47:736–45.

[6]GundemG,VanLooP,KremeyerB,etal.Theevolutionaryhistoryof lethalmetastaticprostatecancer.Nature2015;520:353–7.

[7]HongMK,MacintyreG,WedgeDC,etal.Trackingtheoriginsand driversofsubclonalmetastaticexpansioninprostatecancer.Nat Commun2015;6:6605.

[8]Alix-PanabieresC,PantelK.Challengesincirculatingtumourcell research.NatRevCancer2014;14:623–31.

[9]MiyamotoDT,SequistLV,LeeRJ.Circulatingtumour cells-monitor-ing treatment response inprostate cancer. NatRev Clin Oncol 2014;11:401–12.

[10] AttardG,SwennenhuisJF,OlmosD,etal.CharacterizationofERGAR andPTENgenestatusincirculatingtumorcellsfrompatientswith castration-resistantprostatecancer.CancerRes2009;69:2912–8.

[11] deBonoJS,ScherHI,MontgomeryRB,etal.Circulatingtumorcells predictsurvivalbenefitfromtreatmentinmetastatic castration-resistantprostatecancer.ClinCancerRes2008;14:6302–9.

[12] Nieto MA, Huang RY, Jackson RA, Thiery JP. EMT 2016. Cell 2016;166:21–45.

[13] LecharpentierA,VielhP,Perez-MorenoP,etal.Detectionof circu-latingtumourcellswithahybrid(epithelial/mesenchymal) phe-notypeinpatientswithmetastaticnon-smallcelllungcancer.BrJ Cancer2011;105:1338–41.

[14] MassardC,OulhenM,LeMoulecS,etal.Phenotypicandgenetic heterogeneity of tumor tissue and circulating tumor cells in patientswith metastatic castration-resistant prostate cancer:a report from the PETRUS prospective study. Oncotarget 2016;7:55069–82.

[15] KrebsMG,HouJM,SloaneR,etal.Analysisofcirculatingtumorcells inpatientswithnon-smallcelllungcancerusingepithelial marker-dependent and -independent approaches. J Thorac Oncol 2012;7:306–15.

[16] LohrJG,AdalsteinssonVA,CibulskisK,etal.Whole-exome sequenc-ingofcirculatingtumorcellsprovidesawindowintometastatic prostatecancer.NatBiotechnol2014;32:479–84.

[17] HeitzerE,AuerM,GaschC,etal.Complextumorgenomesinferred fromsinglecirculatingtumorcellsbyarray-CGHand next-genera-tionsequencing.CancerRes2013;73:2965–75.

[18]Polzer B,MedoroG, PaschS,etal.Molecularprofilingofsingle circulatingtumorcellswithdiagnosticintention.EMBOMolMed 2014;6:1371–86.

[19] AdalsteinssonVA,LoveJC.Towardsengineeredprocessesfor se-quencing-basedanalysisofsinglecirculatingtumorcells.CurrOpin ChemEng2014;4:97–104.

[20] Farace F, MassardC, VimondN, et al. A direct comparison of CellSearchandISETforcirculatingtumour-celldetectioninpatients withmetastaticcarcinomas.BrJCancer2011;105:847–53.

[21] SwennenhuisJF,TibbeAG,StevensM,etal.Self-seedingmicrowell chipfortheisolationandcharacterizationofsinglecells.LabChip 2015;15:3039–46.

[22] HofmanV,IlieMI,LongE,etal.Detectionofcirculatingtumorcells asaprognosticfactorinpatientsundergoingradicalsurgeryfor non-small-celllungcarcinoma:comparisonoftheefficacyofthe CellSearchassayandtheisolationbysizeofepithelialtumorcell method.IntJCancer2011;129:1651–60.

(11)

[23] MurtazaM,DawsonSJ,TsuiDW,etal. Non-invasiveanalysisof acquiredresistancetocancertherapybysequencingofplasmaDNA. Nature2013;497:108–12.

[24] BaccelliI, Schneeweiss A,Riethdorf S, et al. Identificationof a populationofblood circulating tumor cells from breast cancer patientsthatinitiatesmetastasisinaxenograftassay.Nat Biotech-nol2013;31:539–44.

[25] PaillerE,AdamJ,BarthelemyA,etal.Detectionofcirculatingtumor cellsharboringauniqueALKrearrangementinALK-positive non-small-celllungcancer.JClinOncol2013;31:2273–81.

[26] ScherHI,GrafRP,SchreiberN,etal.Phenotypicheterogeneityof circulatingtumorcellsinformsclinicaldecisionsbetweenAR sig-nalinginhibitorsandtaxanesinmetastaticprostatecancer.Cancer Res2017;77:5687–98.

Referenties

GERELATEERDE DOCUMENTEN

production during progression and resolution of fibrosis in vivo, and we used precision-cut murine liver slices and fibroblast cell lines to study regulation of hepatic

Facebook… Mijn ouders lopen ook regelmatig te zeuren dat ik vaker het nieuws moet kijken, maar er worden heel veel negatieve verhalen voor verteld, en dat vind ik gewoon niet leuk

Finally, chapter 3 will start discussing the Rijkens group in detail, focussing on the role the group played regarding Dutch governmental policy, the influence it had

Het  Perzisch  Tapijt  op  de  Amerhof  is  geen  gevolg  van  ruimtelijke  ontwikkeling  als  gevolg  van  inspraak,  maar  een  individueel  initiatief  (waar 

Over de steden waar een (?) achter geplaatst is, moet verder epigrafisch bewijs aantonen of deze steden correct zijn toegevoegd aan de lijst.. 50 Verder werden niet alleen

This thesis focusses on the second phase of adoption which is the phase in which the decision whether to adopt a technology (or innovation) or not, will be made. Four

Despite the resistivity of the buffer layer against oxidation at the pressures required for complete oxidation of Ti and Sr, we showed that at least a 0.05 mbar pressure is

Environmental lmpact Assessment (EIA): Cutting Edge for the T7i)enty-First Century. Cambridge, Cambridge University Press. EIA in Brazil: a procedures- practice gap.