• Non ci sono risultati.

Patient-derived xenografts (PDXs) as model systems for human cancer

N/A
N/A
Protected

Academic year: 2021

Condividi "Patient-derived xenografts (PDXs) as model systems for human cancer"

Copied!
6
0
0

Testo completo

(1)

Patient-derived

xenografts

(PDXs)

as

model

systems

for

human

cancer

Federica

Invrea

1

,

Roberta

Rovito

1

,

Erica

Torchiaro

1

,

Consalvo

Petti

1

,

Claudio

Isella

1,2

and

Enzo

Medico

1,2

Patient-derivedxenografts(PDXs)areobtainedby transplantingfragmentsofapatient’stumourinto immunodeficientmice.GrowthandpropagationofPDXs allowscorrelatingtherapeuticresponseinvivowithextensive, multi-dimensionalmolecularannotation,leadingto

identificationofpredictivebiomarkers.PDXsareincreasingly recognisedasclinicallyrelevantmodelsofcancerforseveral reasons,ofwhichthemainisthepossibilityofstudyingthe behaviourofcancercellsinanaturalmicroenvironment,where theyinteractwithstromalcomponentsaccruedfromthemouse host.PDXsmaintainclosesimilaritieswiththetumouroforigin, intermsoftissuearchitecture,molecularfeaturesand responsetotreatments.Indeed,preclinicaltrialsinPDXshave beenshowntomatchandalsoanticipatedataobtainedin patients.Explorationofmorecomplexprocesseslike metastaticevolutionandantitumourimmuneresponsesis activelypursuedwithPDXs,asnewgenerationsofhostmodels emergeonthehorizon.

Addresses

1CandioloCancerInstitute,FPO-IRCCS,stradaProv.142,km3,95, 10060Candiolo(TO),Italy

2UniversityofTorino,DepartmentofOncology,stradaProv.142,km 3,95,10060Candiolo(TO),Italy

Correspondingauthor:Medico,Enzo(enzo.medico@unito.it)

CurrentOpinioninBiotechnology2020,63:151–156

ThisreviewcomesfromathemedissueonSystemsbiology

EditedbyMarcoErcoleVanoniandPasqualePalumbo

https://doi.org/10.1016/j.copbio.2020.01.003

0958-1669/ã2020TheAuthor(s).PublishedbyElsevierLtd.Thisisan openaccessarticleundertheCCBYlicense(http://creativecommons. org/licenses/by/4.0/).

Introduction

Developmentofexperimentalmodelsthatcorrectly

reca-pitulatetumourbiology,geneticheterogeneityanddrug

responsehasbeenakeyobjectiveofcancerresearchsince

itsinception.PDXsaregeneratedbyimplanting

patient-derivedtumourtissueintohost animalmodels,typically

immunocompromisedmice.Afterafirststepof

engraft-mentandadaptationtothenewhost,PDXtumourscan

begrown,explanted,aliquotedandimplantedinfurther

animals,aprocessknownas‘passage’,togeneratecohorts

ofanimalshostingthesametumour,forpreclinical

treat-ment efficacy studies (Figure 1). PDX-derived tumour

tissue canalsobecryopreserved,togenerateofa‘living

tissue biobank’for virtuallyunlimited tissueavailability

forexperimentsandmolecularprofiling.Themain

advan-tage of PDXs versus invitro cancer cell culturesis the

possibilityofstudyingcancercellsintheir

microenviron-mentandassessingtheinvolvementoffibroblasts,

endo-thelial cells and leukocytes in tumour biology and

response to treatments. Over many years, PDXs have

been used to study multiple aspects of the neoplastic

disease [1]. The PDX field isin continuousexpansion,

and large consortia like EurOPDX in Europe (URL:

http://www.europdx.eu/) and PDXNet in the USA

(URL: https://www.pdxnetwork.org/) have been

estab-lished to standardise proceduresandmake PDXs

avail-abletothecancerresearchcommunity.Inthisreview,we

summarisethemostrecentfindingsderivedfromtheuse

of PDXsascancermodels.

Modelling

drug

response

and

resistance

Patient-derived xenografts (PDXs) are widely used to

recapitulatehowthecomplexityoftumourbiologyaffects

drugresponse.Inthelastfewyears,severalstudieshave

been conducted to define pharmacological

vulnerabil-ities. In this context, a large-scale in vivo screen in

1000PDXmodelsassessedthevalueofsuchapproach

in term of reproducibility and clinical translatability, to

identify associations between genotypes and drug

responses [2]. As an alternative to using large PDX

cohorts to establish genotype-response correlations, a

moreempiricalstrategyisbasedonthe‘Avatar’approach:

an individual patient’s tumour tissue is used to derive

PDXs, onwhichmultipletreatments aretested,so that

themosteffectivecanbeadministeredtothepatientof

origin. PDXs can be used to investigate personalised

treatments in real-time (co-clinical trials), as well as to

findand validatenewtargets[3,4].PDX-basedstudies

allowed optimisation of clinical trial designs, providing

strong rationale for new combinatorial regimens in

patients refractory to conventional treatments [5,6].

PDXmodelshavebeenemployedtorecapitulate

mech-anismsofprimaryandacquireddrugresistance[7,8].In

vivo studies highlighted that tumour resistance

mecha-nisms identified in PDXs are also found in theoriginal

patientsamplesresistanttotargettherapy.Thisprovided

potentialtreatmentoptionsinresistanttumours[9]and

(2)

acquired resistance [10]. A limitation of the PDX

approach is the considerable effort and time required

to testinvivo theresponseto multipledrugs. To

over-comethisissue,invivoimplantabledevicesallowtesting

simultaneouslydifferentdrugsinadjacentregionsofthe

same PDX tumour [11]. Moreover, loss-of-function

genetics screenings have been exploited to identify

newtherapeutictargets,employingpooledshorthairpin

RNA(shRNA)libraries,adaptedforinvivoscreensincell

linexenograftsandPDXs[12].

PDX-derived

in

vitro

models

In vivo experiments in PDXs are limited by cost,size,

timeandresources.Reachingtheadequatesamplesizeto

explore inter-patient heterogeneity is therefore quite

challenging. The use of in vitro tests on PDX-derived

cellsfollowedbyinvivovalidation isavalidalternative

and can be achieved in different ways. Short-term 2D

culturescanbeusedforrapid drugtests,thatdisplayed

goodconcordancewithinvivoexperiments[13].

Alter-natively,long-termculturesmaybeemployedto

estab-lishcelllinesretainingthepropertiesofthePDXoforigin

[14].Patient-derivedorPDX-derivedcellscanbegrown

as 3D organoids[15],that retaincertain architectural

featuresandcanbeusedfordrugefficacystudiesandalso

tofurthergeneratePDXswhendirectinvivopropagation

is limiting [16,17]. Cancer-initiating stem cells can be

derived from PDXs and maintained in suspension as

spheroids, a good model for studying the biology and

drugsensitivityofcancerstemcells[18].

Modelling

tumour-stroma

interactions

Itiswidelyrecognisedthatthebiologyofsolidtumours

depends on the ability of cancer cells to recruit and

educatevascular, mesenchymal and immune cells, that

collectively form the tumour microenvironment, or

tumour stroma. During engraftment in a PDX, cancer

cells retainthis ability, so thatwhile thetumour grows

humanstromalcellsarereplacedbymousecounterparts,

thusprovidinganidealexperimentalmodelto

character-isetumour-stromainteractions[19].Indeed,thefactthat

stromal cells in a PDX are of mouse origin has been

considereda limit,potentially affectingtumour biology

andevolution[20].However,thehistologicalarchitecture

ofthetumourtissueismaintainedduringPDX

propaga-tion,suggesting thatthekey mediatorsoftumor-stroma

interactions are functional [21]. Moreover, proteomic

analyses revealed high correspondence of matched

tumourand PDX stromalprofiles, as wellasadaptation

of the stromal proteome upon PDX engraftment, with

distinctprofiles fordifferentPDXsamples[22,23].

Figure1

F0 (Surgery)

“Xen

otr

ial

F1 (Engraftment) F2 (Expansion) Vehicle Drug X Drug Y Drug Z Time (days) % r o m u T e m ul o v F3 (Validation)

Vehic

he

Drug X

Drug Y

Drug Z

Biobanking,

molecular profiling,

cell isolation

Current Opinion in Biotechnology

Generation,expansionanduseofPDXs.Thepatient-derivedtumourisimplantedintoanimmunodeficientmouseand,aftertheengraftment phase,itisexplantedandexpandedinmultiplepassages,enablingthegenerationofcohortsofPDXs,suitableforpreclinical«xenotrials»to evaluatedrugefficacy.PDX-derivedtumoursamplescanbecollectedateverypassagetocreateafrozen,vitaltissuebiobankandtoperform molecularprofilesandexvivoexperiments.

(3)

Tumor-stroma interactions in PDX have been

exten-sively explored as potential therapeutic targets, we

report here the most recent findings. In colorectal

cancer (CRC)PDXs harbouringAPCmutations,

block-ade of the RSPO3 signal from cancer-associated

fibro-blasts (CAFs) to cancer cells enhanced the activity of

paclitaxel-based chemotherapy [24]. Prolonged

treat-ment oflung cancer xenografts with akinase inhibitor

induced ametabolicshift towardincreased lactate

pro-ductionincancercells, thatinturninducedaparacrine

supplyofHGF fromCAFs, promotingresistance. This

adaptive resistance mechanism was also observed in

patients, which showed clinical relevance [25]. In

CRC PDXs, non-canonical TGF-beta signalling from

cancer cells was found to activate CAFs.Inhibition of

this axis led to reduction of metastatic dissemination

and increased sensitivity to therapy [26]. Finally, in

breast cancer PDXs, microvesicle-mediated transfer of

miR-221 from CAFs to cancer cells was found to

pro-moteresistancetohormonaltherapy,highlightinganew

therapeutic avenue [27].

Modelling

metastasis

Thepossibilityofmodellingmetastaticprogressionusing

subcutaneousPDXimplantsisquitelimited.However,a

muchbetter mimicof thisprocessis obtained byimplanting

PDXsinthesametissuefromwhichthepatient’stumour

wasexplanted[28].Thistypeofimplant,calledorthotopic,

in mostcasesrequiresmicrosurgicalcompetencesandin

vivo imaging, for the implant [29] and for subsequent

evaluationof primary tumour and metastasesdevelopment.

Anotableexceptionisbreastcancer,forwhichthestandard

PDX implant in the mammary fat pad is intrinsically

orthotopicandallowsmodellingmetastasis[30].

Severalapproacheshavebeendevelopedandexploitedto

study the metastatic process and its therapeutically

actionabledependenciesinPDXs.Themost

straightfor-wardistheimplantof theprimarytumour inthe

corre-spondingsiteofthemouse.Metastaticbehaviourcanbe

furtherenhancedwhenstromalcellsareaddedtocancer

cells [31]. When the source material is limited (e.g.

metastasis biopsies), a first generation of PDXs can be

derived subcutaneously,and then implanted

orthotopi-cally, retaining the features of the tumour of origin,

includingmetastaticability[17].Humanmetastaseshave

beenimplantedorthotopicallyintheprimarytumoursite,

tomodelatthesametimetumourgrowthattheprimary

site and metastatic propagation [32]. Finally, human

metastases canbe implanted in the primary metastasis

site,asinthecaseofliver-metastaticCRC,torecapitulate

thebehaviourofthediseasewithinitsmetastatic

micro-environment[33].Itisworthnotingthatinallthesecases

morphological,molecularandpharmacologicalfeaturesof

theorthotopicPDXscorrectlyrecapitulatedthoseofthe

tumour oforigin.

Modelling

anticancer

immunotherapy

Toavoidrejectionofhumantumourimplants,PDXsare

invariablygeneratedinimmunocompromisedmice.

Var-ious mouse strains have been developed over several

decades with progressively increasing

immunosuppres-sion, from athymic nude mice in 1966 to non-obese

diabetic-severe combined immunodeficiency

(NOD-SCID) mice in 1995, to NOD-SCID-Gamma-null

(NSG)in2002.Higherlevelsofimmunodeficiencyallow

better engrafting[34],but alsoprogressively reducethe

possibilityto explore therole oftheimmune systemin

tumour biology and response to treatments [35]. This

drawback is particularlycritical for modelling

immuno-therapybasedoncheckpointinhibitors,asitrequiresan

intact human immune system. A wayto overcome this

limitationisthegeneration of‘humanized’mouse

mod-els, byengraftingNSG micewithhuman leukocytesor

hematopoieticstemcells.Patienttumourtissuesarethen

engraftedintothehumanizedmiceandusedforstudying

theefficacyofimmunomodulatorytreatments.However,

thisapproachhasahighriskofgraft-versus-hostdisease

andthereforeallowsonlyshorttermexperiments[36,37].

Mouse‘humanization’hasbeenimprovedbygenetically

insertinghumancytokinegenes(M-CSF,IL-3,GM-CSF

thrombopoietin) at their respective mouse loci, plus

transgenicexpressionofhumanSIRPa28whichenables

mouse phagocytesto toleratehuman cells[38].Indeed,

recent studies with humanizedmouse PDXs have

pro-videdsignificantadvancesin thecomprehensionof

can-cerimmunotherapy [39,40].

ConventionalPDXscanstillhaveagooduseformodelling

another type of immunotherapy, called ‘adoptive

immunotherapy’,inwhichcytotoxicT-lymphocytes,

nat-uralkillercellsorengineeredkillercellsareadministeredto

testtheirdirectanticanceractivity.Asanexample,

Jesper-senandcolleaguesinsertedmelanomatumourcellsandT

cells fromthesame patientintoNSG miceandshowed

tumour inhibition [41].Alternatively, cytokine-induced

killer(CIK)cellscanbegeneratedfromthepatientorfrom

donorsandtestedforanticancerefficacyinPDX,aloneorin

combination with cancer-targeting antibodies [42]. An

interesting researchfrontier in adoptiveimmunotherapy

isthe engineeringofkillercellswithachimeric antigen

receptor,todirectthemagainstaspecificantigenexpressed

bycancercells.Alsothisapproachhasbeensuccessfully

validatedinPDXmodels[43,44].

PDXs

for

biomarker

development

ThefactthatPDXsprovideavirtuallyunlimitedsource

oftumourtissueformulti-dimensionalmolecularprofiles

andallowtestingresponsetomultipledrugsinthesame

model opened a new avenue for biomarker discovery.

Moreover,thefactthatDNAandRNAfromstromalcells

are of mouse origin enables distinguishing cancer

cell-intrinsic (human) from microenvironmental (mouse)

(4)

orsingle-cellsequencing[45].Recentexamplesof

PDX-based biomarker discovery include: (i) identification of

lungcancercell-specificdownregulationof

histocompati-bilityantigens,furtherconfirmedinclinicalsamplesand

associatedtoresistancetoimmunotherapy[46];(ii)lackof

GATA4 expression as a predictor of sensitivity to

TGFBR1inhibition[47];(iii)identificationofp38-alpha

inhibition as sensitizer to taxanes in breast cancer[48].

PDXs havealso been usedto studycirculating tumour

cells (CTCs). In breast cancer, a molecular signature

associated with thepresence of CTC clusters in PDXs

wasassociatedwithworseoutcomeinpatients[49].The

possibilityof performingspecies-specificRNA

sequenc-ing of PDXs to distinguish cancer cell from stromal

transcriptomes hasbeen extensively exploited in CRC,

leadingto theidentificationononesideof apoor

prog-nosisstromalsignature[45],and ontheotherof

cancer-cell-intrinsic molecular subtypes of CRC (CRIS

sub-types) with unprecedented predictive and prognostic

power[50,51].Similarly,acancercell-intrinsic

microsat-elliteinstability transcriptionalsignaturedefinedin

gas-tric cancer PDXs was found to identify patients with

worseprognosis[52].

Conclusion

and

outlook

PDXsofferunprecedented opportunitiesfordeveloping

precisionmedicineapproachestocancertreatment.Vast

PDXcollectionshavebeengeneratedglobally,summing

up to thousands of cases and enabling population-scale

preclinicaltrialsthatcaptureinter-tumourheterogeneity.

Yet,eachindividualmodelrecapitulateswithinitselfthe

complexityofasinglehumantumour,allowingin-depth,

longitudinalexplorationofadaptiveandevolutive

dynam-icsthattypicallyconferresistancetotreatments.ThePDX

communityisfacingimportantchallengestoimprovethe

efficacyofthesemodels.Inparticular,thetime,effortand

costsnecessaryforinvivoexperimentsareurging

research-erstodevelopanduse exvivosystems,exploiting

PDX-derived cells for simpler in vitro experiments before

embarking in PDX tests. Improved ways of modelling

metastatic progression and interactions between cancer

anditsmicroenvironment arecontinuouslybeing

devel-oped,aswellasin-depthmolecularprofilingofalltumour

components.Thetimeinwhichnewlydiagnosedcancer

patientswillfindexistingPDXswithmatchingfeaturesfor

treatmentpersonalizationisnotfar.

Conflict

of

interest

statement

Nothingdeclared.

Acknowledgements

ThisworkwassupportedbyEUH2020ResearchandInnovation Programmeprojectn.731105‘EDIReX’toE.M.;AssociazioneItalianaper laRicercasulCancro5permille2018projectn.21091toE.M.;MyFirst AIRCGrantprojectn.19047toC.I.;andFondazionePiemonteseperla RicercasulCancro-ONLUS5permilleMinisterodellaSalute(year2015to E.M.).

References

and

recommended

reading

Papersofparticularinterest,publishedwithintheperiodofreview, havebeenhighlightedas:

 ofspecialinterest ofoutstandinginterest

1. ByrneAT,Alfe´rezDG,AmantF,AnnibaliD,ArribasJ,BiankinAV, BrunaA,Budinska´ E,CaldasC,ChangDKetal.:Interrogating openissuesincancermedicinewithpatient-derived xenografts.NatRevCancer2017,17:632.

2. GaoH,KornJM,FerrettiS,MonahanJE,WangY,SinghM, ZhangC,SchnellC,YangG,ZhangYetal.:High-throughput screeningusingpatient-derivedtumorxenograftstopredict clinicaltrialdrugresponse.NatMed2015,21:1318-1325.

3. SavaikarMA,WhiteheadT,RoyS,StrongL,FettigN,PrmeauT, LuoJ,LiS,WahlRL,ShoghiKI:SUV(25)andmPERCIST: precisionimagingofresponsetotherapyinco-clinical FDG-PETimagingoftriplenegativebreastcancer(TNBC) patient-derivedtumorxenografts(PDX).JNuclMed2019.pii: jnumed.119.234286.[Epubaheadofprint].

4.

 InoueCurrentA,DeemandfutureAK,KopetzhorizonsS,Heffernanofpatient-derivedTP,DraettaxenograftGF,CarugoA: modelsincolorectalcancertranslationalresearch.Cancers (Basel)2019,11pii:E1321

Thisreviewprovidesinformationonongoingco-clinicaltrialstoevaluate theutilityofPDXmodelsinpredictingdrugresponseinvariousprimary andmetastaticsolidtumoursbeyondCRC.

5. BertottiA,MigliardiG,GalimiF,SassiF,TortiD,IsellaC,Cora` D,Di NicolantonioF,BuscarinoM,PettiCetal.:Amolecularly annotatedplatformofpatient-derivedxenografts

(“xenopatients”)identifiesHER2asaneffectivetherapeutic targetincetuximab-resistantcolorectalcancer.CancerDiscov 2011,1:508-523.

6.

 LonardiSartore-BianchiS,BergamoA,TrusolinoF,ZagonelL,MartinoV,LeoneC,F,BencardinoDepetrisI,MartinelliK, E etal.:Dual-targetedtherapywithtrastuzumabandlapatinibin treatment-refractory,KRAScodon12/13wild-type, HER2-positivemetastaticcolorectalcancer(HERACLES):a proof-of-concept,multicentre,open-label,phase2trial.LancetOncol 2016,17:738-746

SuccessfulclinicaltrialonmetastaticCRCbasedonacombinedtargeted treatmentpreviouslydevelopedandvalidatedinPDXs,seeBertottietal. [5].

7. MarangoniE,LaurentC,CoussyF,El-BottyR,Chaˆteau-JoubertS, ServelyJL,dePlaterL,AssayagF,DahmaniA,MontaudonEetal.: CapecitabineefficacyiscorrelatedwithTYMPandRB1 expressioninPDXestablishedfromtriple-negativebreast cancers.ClinCancerRes2018,24:2605-2615.

8.

 SoleGo´mez-MiragayaA,Pare´ L,Go´mezJ,Mora´nA,SerraS,Calleja-CervantesV,DobroleckiLE,ME,LewisCollado-MT, Diaz-LagaresAetal.:ThealteredtranscriptomeandDNA methylationprofilesofdocetaxelresistanceinbreastcancer PDXmodels.MolCancerRes2019,17:2063-2076

AnexampleofinductionofsecondaryresistanceinvivoinPDXmodels. TheauthorsrenderedPDXsoftriple-negativebreastcancerresistantin vivo to docetaxel, and identified molecular markers of acquired resistance.

9.

 YaoForestYM,A,DonohoNovosiadlyGP,RD,IversenWebsterPW,YW,ZhangEbertY,VanP,BrayHornSRD,etal.: MousePDXtrialsuggestssynergyofconcurrentinhibitionof RAFandEGFRincolorectalcancerwithBRAForKRAS mutations.ClinCancerRes2017,23:5547-5560

ThisPDX-basedtrialrecapitulatedclinicalresultsofcetuximabon color-ectalpatientsanddemonstratedthatconcurrentEGFRandRAFinhibition displayssynergisticantitumoractivityforKRAS-mutantorBRAF-mutant PDXs.

10. KemperK,KrijgsmanO,Cornelissen-SteijgerP,ShahrabiA, WeeberF,SongJY,KuilmanT,VisDJ,WesselsLF,VoestEEetal.: Intra-andinter-tumorheterogeneityina vemurafenib-resistantmelanomapatientandderivedxenografts.EMBOMol Med2015,7:1104-1118.

11. JonasO,LandryHM,FullerJE,SantiniJTJr,BaselgaJ,TepperRI, CimaMJ,LangerR:Animplantablemicrodevicetoperform

(5)

high-throughputinvivodrugsensitivitytestingintumors.Sci TranslMed2015,7:284ra.

12. CarugoA,GenoveseG,SethS,NeziL,RoseJL,BossiD, CicaleseA,ShahPK,VialeA,PettazzoniPFetal.:Invivo functionalplatformtargetingpatient-derivedxenografts identifiesWDR5-Mycassociationasacriticaldeterminantof pancreaticcancer.CellRep2016,16:133-147.

13.

 CriscitielloPunziS,MeliksetianC,OrsiF,SpaggiariM,RivaL,L,MarocchiCasiraghiF,M,PruneriDellaVignaG, Petal.: Developmentofpersonalizedtherapeuticstrategiesby targetingactionablevulnerabilitiesinmetastaticand chemotherapy-resistantbreastcancerPDXs.Cells2019,8pii: E605

Theauthorsderivedshort-terminvitroculturesfrommetastaticbreast cancerPDXsandverified concordantinvitroandinvivoresponseto drugs.

14.

 DurinikovaLazzariL,CortiE,ZanellaG,PiccoER,G,NovaraIsellaL,C,BarbosaMontoneFM,etArcellaal.:Patient-P, derivedxenograftsandmatchedcelllinesidentify pharmacogenomicvulnerabilitiesincolorectalcancer.Clin CancerRes2019,25:6243-6259

Thisworkshowsthatlong-termculturedcelllinesderivedfromPDXs retainthegeneticfeaturesanddrugsensitivityprofileofthePDXsof origin.

15.

 MateosVlachogiannisJ,KhanG,K,HedayatLampisA,S,EasonVatsiouK,HuntingfordA,JaminY,I,Ferna´ndez-BurkeRetal.: Patient-derivedorganoidsmodeltreatmentresponseof metastaticgastrointestinalcancers.Science2018, 359:920-926

Organoidsderivedfromcancerpatientswereusedtogenerate xeno-grafts.Thedrugresponseoforganoidsandderivedxenografts recapi-tulatedthoseobservedinpatients.

16.

 ShiMartins-FilhoR,RadulovichS,RaghavanN,NgC,V,LiuLiN,Q,NotsudaMerASetH,al.:CabaneroOrganoidMN, culturesaspreclinicalmodelsofnon-smallcelllungcancer. ClinCancerRes2019.pii:clincanres.1376.2019.[Epubaheadof print]

Theauthorsderivedorganoidsfrompatients’tumoursorPDXofearly stagenon-smalllungcancer.Theorganoidsrecapitulatedthegenetic, transcriptional andhistologicfeatures oftheparentaltumour,that is, patient tumours or PDX, and tumorigenicityin vivo when implanted subcutaneously.

17. MizukoshiK,OkazawaY,HaenoH,KoyamaY,SulidanK, KomiyamaH,SaekiH,OhtsujiN,ItoY,KojimaYetal.:Metastatic seedingofhumancoloncancercellclustersexpressingthe hybridepithelial/mesenchymalstate.IntJCancer2019.Sep 10.[Epubaheadofprint].

18.

 LuraghiBaccoF,P,IsellaBigattoC,BellomoV,CiprianoSE,E,MedicoReatoEG,etOrzanal.:AF,molecularlySassiF,De annotatedmodelofpatient-derivedcoloncancerstem-like cellstoassessgeneticandnongeneticmechanismsof resistancetoanti-EGFRtherapy.ClinCancerRes2018, 24:807-820

Colon-cancerinitiatingcellsderivedfromPDXsusingastem cell-selec-tivemediumgrowinsuspensionasspheroidcoloniesandsuccessfully generatexenografts.Thislong-termplatformallowsthestudyofthemore resistantsubpopulationofcancerstemcellsresponsibleforresistance andrelapse.

19.

 ChaoGomezC,G,WidenQiuS,SG,ThompsonWoodTG,J,ZatarainSprattHJR,etal.:JohnsonPatient-derivedP,GajjarA, xenograftsfromcolorectalcarcinoma:atemporaland hierarchicalstudyofmurinestromalcellreplacement. AnticancerRes2017,37:3405-3412

Accurateworkdepictingtemporalkineticsofhumanstromalcell replace-mentwithmousecounterpartsduringPDXengraftmentandpropagation.

20. Ben-DavidU,HaG,TsengYY,GreenwaldNF,OhC,ShihJ, McFarlandJM,WongB,BoehmJS,BeroukhimRetal.: Patient-derivedxenograftsundergomouse-specifictumorevolution. NatGenet2017,49:1567-1575.

21. DelittoD,PhamK,VladaAC,SarosiGA,ThomasRM,BehrnsKE, LiuC,HughesSJ,WalletSM,TrevinoJG:Patient-derived xenograftmodelsforpancreaticadenocarcinoma demonstrateretentionoftumormorphologythrough incorporationofmurinestromalelements.AmJPathol2015, 185:1297-1303.

22.

 BlommeOtakaY,A,ShererVanSimaeysF,LovinfosseG,DoumontP,BoutryG,S,CostanzaPalaciosB,APBellieretal.:J, Murinestromaadoptsahuman-likemetabolicphenotypein thePDXmodelofcolorectalcancerandlivermetastases. Oncogene2018,37:1237-1250

Metabolicprofileswereanalysedtoshowthatcancercell-driven repro-grammingofmousestromalcellsinPDXsrecapitulateswhatfoundinthe tumoursoforigin.

23. WangX,MooradianAD,Erdmann-GilmoreP,ZhangQ,VinerR, DaviesSR,HuangKL,BomgardenR,VanTineBA,ShaoJetal.: Breasttumorseducatetheproteomeofstromaltissueinan individualizedbutcoordinatedmanner.SciSignal2017,10pii: eaam8065.

24.

 FischerMurrielC,MM,EvansYeungJW,VP,O’YoungCattaruzzaG,BrunnerF,HusseinAL,WangR,YenMWC,etal.: RSPO3antagonisminhibitsgrowthandtumorigenicityin colorectaltumorsharboringcommonWntpathway mutations.SciRep2017,7:15270

NiceexampleoftheuseofPDXstoidentifyatherapeuticallyactionable paracrinesignalfromCAFstocancercells.

25. ApicellaM,GiannoniE,FioreS,FerrariKJ,Ferna´ndez-Pe´rezD, IsellaC,GranchiC,MinutoloF,SottileA,ComoglioPMetal.: Increasedlactatesecretionbycancercellssustains non-cell-autonomousadaptiveresistancetoMETandEGFRtargeted therapies.CellMetab2018,28:848-865.e6.

26.

 Guille´nCimasFJ,Dı´az-MarotoGarcı´a E,N,Gonc¸Sanz-Pamplonaalves-RibeiroS,R,AlbertBerdiel-AcerN,Garcia-M, Vicie´nG,CapellaG,MorenoVetal.:NoncanonicalTGFb pathwayrelievestheblockadeofIL1b/TGFb-mediated crosstalkbetweentumorandstroma:TGFBR1andTAK1 inhibitionincolorectalcancer.ClinCancerRes2019, 25:4466-4479

Inhibitionofthetumour-stromacross-talkreducedstromalaccrualand sensitizedtochemotherapyinCRCPDXmodels.

27. SansoneP,BerishajM,RajasekharVK,CeccarelliC,ChangQ, StrillacciA,SaviniC,ShapiroL,BowmanRL,MastroleoCetal.: Evolutionofcancerstem-likecellsinendocrine-resistant metastaticbreastcancersismediatedbystromal microvesicles.CancerRes2017,77:1927-1941.

28. HoffmanRM:Patient-derivedorthotopicxenografts:better mimicofmetastasisthansubcutaneousxenografts.NatRev Cancer2015,15:451-452.

29. HayCA,SorR,FlowersAJ,ClendeninC,ByrneKT: Ultrasound-guidedorthotopicimplantationofmurinepancreaticductal adenocarcinoma.JVisExp2019,153http://dx.doi.org/10.3791/ 60497.

30.

 FatimaKuenzingerI,El-AyachiWL,WendI,PlayaP,PenceHC,JC,Alva-OrnelasBrakefieldJA,L,KrutilinaKhalidAB,RIetal.: Simultaneousmulti-organmetastasesfromchemo-resistant triple-negativebreastcancerarepreventedbyinterferingwith WNT-signaling.Cancers(Basel)2019,11

Niceexampleoftherapeutic interferencewiththemetastaticprocess usingbreastcancerPDXsimplantedinthemammaryfatpad.

31. GillsJ,MoretR,ZhangX,NelsonJ,MareshG,HellmersL, CanterD,HudsonM,HalatS,MatranaMetal.:Apatient-derived orthotopicxenograftmodelenablinghumanhigh-grade urothelialcellcarcinomaofthebladdertumorimplantation, growth,angiogenesis,andmetastasis.Oncotarget2018, 9:32718-32729.

32.

 KongChoiSI,SY,JeonYoonAR,KA,KimKohMK,YHLeeetal.:YS,DevelopmentImJE,KohJW,ofpatient-HanSS, derivedpreclinicalplatformformetastaticpancreaticcancer: PDOXandasubsequentorganoidmodelsystemusing percutaneousbiopsysamples.FrontOncol2019,9:875

Theauthorsderivedametastaticmodelofpancreaticcancerbymeansof orthotopic implantation of human liver metastases into the primary tumour site, that is, pancreas. The model recapitulated the genetic and histopathological characteristicsof theprimarytumours, aswell asdistantmetastasesanddrug-responseobservedinpatients.

33. Roque-LimaB,RoqueCCTA,BegnamiMD,PeresiP,LimaENP, MelloCAL,CoimbraFJ,ChojniakR,GossSantosT:Development ofpatient-derivedorthotopicxenograftsfrommetastatic colorectalcancerinnudemice.JDrugTarget2019,27:943-949.

(6)

34. OkadaS,VaeteewoottacharnK,KariyaR:Applicationofhighly immunocompromisedmicefortheestablishmentof patient-derivedxenograft(PDX)models.Cells2019,8.

35.

 LinnebacherMaletzkiC,BockM:NSGS,FruhmiceP,asMaciushostsK,forWittoncologicalA,PrallF,precision medicine.LabInvest2019.[Epubaheadofprint]

Thisstudyisaclearexampleofhowhighimmunodeficiencyresultsina good engraftment but reduced drug tolerance, highlighting how the choiceofthemousestrainaffectssubsequentresults.

36. AliN,FlutterB,SanchezRodriguezR,Sharif-PaghalehE, BarberLD,LombardiG,NestleFO:Xenogeneic graft-versus-host-diseaseinNOD-scidIL-2RgnullmicedisplayaT-effector memoryphenotype.PLoSOne2012,7:e44219.

37. TanaskovicO,VergaFalzacappaMV,PelicciPG:Humancord blood(hCB)-CD34+humanizedmicefailtorejecthumanacute myeloidleukemiacells.PLoSOne2019,14:e0217345.

38. RongvauxA,WillingerT,MartinekJ,StrowigT,GeartySV, TeichmannLL,SaitoY,MarchesF,HaleneS,PaluckaAKetal.: Developmentandfunctionofhumaninnateimmunecellsina humanizedmousemodel.NatBiotechnol2014,32:364-372.

39.

 CapassoDiamondA,JR,LangKopetzJ,PittsS,BarbeeTM,JordanJ,PetersonKR,LieuJetCH,al.:DavisSL, Characterizationofimmuneresponsestoanti-PD-1monoand combinationimmunotherapyinhematopoietichumanized miceimplantedwithtumorxenografts.JImmunotherCancer 2019,7:37

HumanizedmousePDXswereusedtostudyimmunecheckpoint block-adeinbreastandcoloncancer.Tumourgrowthinhibitioncorrelatedwith increasedtumour-infiltratingTcells.

40.

 WangMaAH,M,DeYaoVereLC,WhiteChengRW,M,AirhartCaiD,SMartineketal.:HumanizedJ,PanCX,miceShiW,in studyingefficacyandmechanismsofPD-1-targetedcancer immunotherapy.FASEBJ2018,32:1537-1549

InthisworkhumanizedmousePDXsallowedtodemonstratethat inhibi-tionoftumorgrowthbypembrolizumab(PD-L1inhibitor)insolidtumours isdependentonhCD8+Tcells,asdemonstratedbyantibody-mediated depletion.

41.

 JespersenKellerU,NyH,L,LindbergSvaneIM,MF,NilssonDoniaM,LM,So¨derbergNilssonEMV,JA:ClinicalAndersenR, responsestoadoptiveT-celltransfercanbemodeledinan autologousimmune-humanizedmousemodel.NatCommun 2017,8:707

Inthisworkmelanomatumourcellsandtumour-infiltratingTcellsfrom the same patient were transplantedsequentially in NSG mice. This systemprovidesthepotential frameworktofurthermodel genetically diversehumancancersforassessingtheefficacyofimmunotherapiesas wellascombinationtherapies.

42. GolayJ,MartinelliS,AlzaniR,CribioliS,AlbaneseC,GottiE, PasiniB,MazzantiB,SaccardiR,RambaldiA,IntronaM:Cord blood-derivedcytokine-inducedkillercellscombinedwith blinatumomabasatherapeuticstrategyforCD19(+)tumors. Cytotherapy2018,20:1077-1088.

43.

 TengYangR,WZhaoetal.:J,ChimericZhaoY,GaoantigenJ,LiH,receptor-modifiedZhouS,WangY,SunTcellsQ,LinZ,

repressedsolidtumorsandtheirrelapseinanestablished patient-derivedcoloncarcinomaxenograftmodel.J Immunother2019,42:33-42

This study showed that CAR-T-cell treatment may be a promising approachforsolidtumourclearanceandthatthePDXmodelmaybe usefultoevaluatetheeffectsofCAR-Tcells.

44. MagnaniCF,MezzanotteC,CappuzzelloC,BardiniM, TettamantiS,FazioG,CooperLJN,DastoliG,CazzanigaG, BiondiA,BiagiE:PreclinicalefficacyandsafetyofCD19CAR cytokine-inducedkillercellstransfectedwithsleepingbeauty transposonforthetreatmentofacutelymphoblastic leukemia.HumGeneTher2018,29:602-613.

45. IsellaC,TerrasiA,BellomoSE,PettiC,GalatolaG,MuratoreA, MellanoA,SenettaR,CassentiA,SonettoCetal.:Stromal contributiontothecolorectalcancertranscriptome.NatGenet 2015,47:312-319.

46. PereiraC,Gimenez-XavierP,ProsE,PajaresMJ,MoroM, GomezA,NavarroA,CondomE,MoranS,Gomez-LopezGetal.: Genomicprofilingofpatient-derivedxenograftsforlung canceridentifiesB2Minactivationimpairing

immunorecognition.ClinCancerRes2017,23:3203-3213.

47. GaoL,HuY,TianY,FanZ,WangK,LiH,ZhouQ,ZengG,HuX, YuLetal.:Lungcancerdeficientinthetumorsuppressor GATA4issensitivetoTGFBR1inhibition.NatCommun2019, 10:1665.

48. Ca´novasB,IgeaA,SartoriAA,GomisRR,PaullTT,IsodaM, Pe´rez-MontoyoH,SerraV,Gonza´lez-Sua´rezE,StrackerTHetal.: Targetingp38aincreasesDNAdamage,chromosome instability,andtheanti-tumoralresponsetotaxanesinbreast cancercells.CancerCell2018,33:1094-1110.e8.

49. ThangavelH,AngelisC,VasaikarS,BhatR,JollyMK,NagiC, CreightonCJ,ChenF,DobroleckiLE,GeorgeJTetal.:A CTC-cluster-specificsignaturederivedfromOMICSanalysisof patient-derivedxenografttumorspredictsoutcomesin basal-likebreastcancer.JClinMed2019,8pii:E1772.

50. IsellaC,BrunduF,BellomoSE,GalimiF,ZanellaE,PorporatoR, PettiC,FioriA,OrzanF,SenettaRetal.:Selectiveanalysisof cancer-cellintrinsictranscriptionaltraitsdefinesnovel clinicallyrelevantsubtypesofcolorectalcancer.NatCommun 2017,8:15107.

51.

 AlderdiceMcCorryAM,M,RichmanRoddyAC,SD,VimalachandranGollinsS,StewartD,JP,IsellaHurtCC,etAdamsal.: R, Prospectivepatientstratificationintorobustcancer-cell intrinsicsubtypesfromcolorectalcancerbiopsies.JPathol 2018,245:19-28

ThispapershowsthattranscriptionalclassifiersdevelopedinPDXsand based on cancer cell-intrinsic transcripts have higher concordance acrossmultiplesamplingsitesofthesametumour.

52. CorsoS,IsellaC,BellomoSE,ApicellaM,DurandoS,MiglioreC, UghettoS,D’ErricoL,MenegonS,Moya-RullDetal.:A comprehensivePDXgastriccancercollectioncaptures cancercell-intrinsictranscriptionalMSItraits.CancerRes 2019,79:5884-5896.

Riferimenti

Documenti correlati

Tables II, III and IV show the date and time (UT) of the first event in constituent events in AS cluster, the mean value of the right ascension α and declination δ of AS cluster

For storms with commencement near local noon it has been found that the electric field effect on the vertical drift and peak electron density of the F2 region is pronounced at

Tortola, la più grande delle oltre 40 isole che compongono l'arcipelago delle Isole Vergini Britanniche, vi accoglierà con la sua eleganza, le case bianche che spiccano sul verde

Diversamente da Simon, i ‘planners’ di Penrose sono degli ‘image creators’ più che ‘information processors’; l’analisi parte, infatti, dal loro mondo mentale,

Nella parte iniziale sono state richieste informazioni di carattere generale sulle motivazioni che hanno indotto ad intraprendere il dottorato di ricerca, sull’organizzazione

A way to distinguish the two different materials (aluminium for the plate, air for the radiation domain) with 2D rectangular geometry was not found in COMSOL and a near-field

Mancava quindi spazio nella delega per una modifica sostanziale del sistema di giustizia penale minorile ; tale questione venne risolta solo nel 1982 quando si pensò