Targeting the Microenvironment in Advanced Colorectal Cancer

publicité
TargetingtheMicroenvironmentinAdvancedColorectalCancer
1
2
3
DanieleV.F.Tauriello1andEduardBatlle1,2
4
5
1.InstituteforResearchinBiomedicine(IRBBarcelona).TheBarcelonaInstituteofScience
6
andTechnology.BaldiriiReixac10,08028Barcelona,Spain
7
2.InstitucióCatalanadeRecercaiEstudisAvançats(ICREA),Barcelona,Spain.
8
9
10
11
12
13
14
15
16
17
Correspondence:[email protected]&[email protected]
18
19
20
21
Keywords:stroma,cancerecology,molecularsubtypes,pre-clinicalmousemodels,
22
23
immuneoncology,TGF-β
1
1
Abstract
2
Colorectal cancer (CRC) diagnosis often occurs at late stages when tumour cells have
3
already disseminated. Current therapies are poorly effective for metastatic disease, the main
4
causeofdeathinCRC.Despitemountingevidenceimplicatingthetumourmicroenvironmentin
5
CRCprogressionandmetastasis,clinicalpracticeremainspredominantlyfocussedontargeting
6
the epithelial compartment. As CRCs remain largely refractory to current therapies, we are
7
compelled to devise alternative strategies. TGF-β has emerged as a key architect of the
8
microenvironment in poor prognosis cancers. Disseminated tumour cells show a strong
9
dependencyonaTGF-β-activatedstromaduringtheestablishmentandsubsequentexpansion
10
of metastasis. Here, we will review and discuss the development of integrated approaches
11
focusedontargetingtheecosystemofpoorprognosisCRCs.
12
13
Trends
14
•
15
16
In the search for new paradigms on which to base novel therapeutic strategies for
advancedCRC,focushasincreasinglybeencentredonthetumourmicroenvironment.
•
Thereisanemergingnotionthatinteractionsbetweenepithelialcancercellsandtheir
17
environmentcanbeunderstoodapplyingaconceptualframeworksimilartothatused
18
tostudyecosystems.
19
•
20
21
In CRC patients, a stromal-expressed gene programme enriched for TGF-β and
downstreamtargetshasbeenlinkedtopoorprognosisandmetastasisformation.
•
TGF-β signalling plays key roles in instructing the tumour microenvironment of late
22
stageCRCs,yetinhibitionofTGF-βsignallingasatherapeuticstrategyremainsscarcely
23
exploredintheclinic.
2
1
Thetumourepitheliumasprimarytargetofstandardtherapies
2
Colorectal cancer (CRC) originates from benign lesions known as adenomas: localised
3
glandular overgrowths in the epithelial lining of the large bowel. Over time, some adenomas
4
accumulate mutations in signalling pathways critical to stem cell maintenance, cellular
5
proliferation, and tumour suppression [1, 2] and they can evolve into invasive CRCs that may
6
ultimatelyspreadtodistantorgans.Thisprocesscalledmetastasis(seeBox1)occursinabout
7
40-50%ofpatientsandconfersalowprobabilityofsurvival.
8
In the clinic, patients are classified in 4 stages (see Glossary). Treatment involves
9
aggressive surgical resection of the primary tumour, which cures a large proportion of early
10
stage patients. However, some stage II and many stage III patients will relapse, frequently in
11
theformofmetastasis,asaconsequenceoftumourcellsthatdisseminatedbeforeresection.
12
Unliketheprimarytumour,metastasesarelessfrequentlyremovedbysurgeryunlesslimitedin
13
number and extent [3]. Therefore patients that present with metastases at the time of
14
diagnosis(stageIV)andthoseatperceivedriskofrelapsereceivecytotoxicchemotherapy:in
15
most cases a combination of folinic acid, 5-fluorouracil, and oxaliplatin or ironotecan. This
16
strategyaimstokillhighlyproliferativecancercellsandhasbeenastapleinthetreatmentof
17
solidcancersfordecades[4].AlthoughadjuvantchemotherapyisbeneficialtostageIIIpatients
18
and has modest potential for improved survival in stage II [5, 6], it performs poorly in the
19
metastaticsetting,almostinvariablygivingrisetodrugresistanceanddiseaseprogression.
20
In second-line treatment, chemotherapy is increasingly combined with targeted therapy,
21
designed to intervene with specific signalling pathways or cellular mechanisms [4]. A key
22
exampleinCRCistheuseofantibodiestargetingtheepidermalgrowthfactor(EGF)receptor,
3
1
often exploited by cancer cells to stimulate proliferation. Apart from eventual resistance, the
2
mainproblemwithtargetedtherapiesisthevariableresponsesinpatients,requiringabetter
3
griponpredictivebiomarkers[7].Forinstance,mutationsthatactivatetheMAPK/ERKpathway
4
downstream of EGFR, such as those frequently occurring in the KRAS and NRAS GTPases or
5
BRAF,canrenderanti-EGFRtherapyineffective[8].
6
A better understanding of advanced cancer may lead to more effective therapies for
7
metastaticCRC,butisalsohighlyrelevanttoimprovethemanagementofearlierstagesofthe
8
disease.ArguablythemostimportantquestionforstageI-IIIpatientsiswhetherornottotreat
9
at all and which therapeutic strategy will be beneficial to prevent recurrence. Assessment of
10
probability of relapse after therapy on the individual level remains a major challenge [9].
11
Anatomicalandhistopathologicalfeaturesofthetumoursuchasextentofinvasion(T),number
12
of lymph node metastases (N), bowel perforation or obstruction, the presence of
13
lymphovascularinvasion,andapoorlydifferentiatedhistologyareusedtoidentifyCRCpatients
14
atriskofrecurrence.However,theseparametersonlyholdamoderatepredictivepowerand
15
donothelpselectoptimaltreatmentoptions.Furthermore,withtheexceptionofchromosomal
16
or microsatellite instability (MSI) [10, 11] and mutations in the BRAF oncogene [12], no
17
molecularfeatureisrobustlyassociatedwithprognosisandthereforeusedroutinelyinclinical
18
practice[9,13].
19
20
4
1
TranscriptomicsredefiningCRCclassification
2
Toimprovepatientstratificationandidentifypotentialmoleculartargetsfortherapy,the
3
field has generated large collections of transcriptomic datasets from tumour samples, which
4
have enabled the identification of CRC subtypes based on distinctive global gene expression
5
profiles[14-19].Inanattempttoconsolidatethesedata,arecentmeta-analysisdividedCRCs
6
into 4 defined consensus molecular subtypes (CMS), representing an MSI-like class (CMS1), a
7
canonical WNT/MYC class (CMS2), a metabolically dysregulated class (CMS3), and a
8
mesenchymalclass(CMS4)[20].Thelattersubtype,towhichabout1inevery4CRCsbelongs,is
9
ofparticularinterestasitrelapseswithhigherfrequencythantheothers.
10
Theelevatedexpressionofmesenchymalgenesinanepithelialcancerledtotheproposal
11
that epithelial-to-mesenchymal transition (EMT) characterizes poor prognosis in CRC [21].
12
However,thetranscriptomeofatumoursamplenotonlyreflectstheexpressionprogrammeof
13
epithelial cancer cells but also the profile of mesenchymal cells present in the tumour
14
microenvironment(TME).Indeed,ourgroupandthegroupofEnzoMedicorecentlydiscovered
15
thatexpressionofmesenchymalgenesintranscriptomicCRCdataislargelycontributedbycells
16
oftheTME,mainlybycancer-associatedfibroblasts(CAFs),ratherthanbycancercells[22,23].
17
In agreement with these observations, CMS4 cancers show a higher degree of stromal
18
infiltration than the other subtypes [20]. Furthermore, our analyses indicate that a large
19
proportion of these CAF-expressed genes are strongly associated to cancer relapse and poor
20
prognosisinCRCcohorts[22,23].Ofnote,thefactthattheexpressionofmesenchymalgenes,
21
includingmanyEMTmasterregulatorssuchSNAI1,TWISTandZEB1,iscontributedbystromal
5
1
cellsintranscriptomicprofilesdoesnotruleoutthepossibilitythatsubsetsoftumourcellsmay
2
undergoEMT,particularlyatinvasionfronts.
3
4
TreatingCancerasanEcosystem
5
Focussing on the tumour stroma is not novel. At the end of the 19th century, English
6
physician Stephen Paget – observing preferential patterns of metastatic dissemination to
7
particularorgans–proposedthe‘seedandsoil’hypothesistoexplainthisphenomenonasthe
8
combination of the colonizing cancer cells’ adaptability and a congenial microenvironment in
9
thedistantorgan[24].HesuggesteditwouldbeusefultostudythepropertiesoftheTME,as
10
wellasitscrosstalkwithcancercells.Indeed,theroleoftheTMEbothattheoriginalsiteandin
11
thedistantorganhasbecomeatopicofintenseinvestigationinrecentdecades[25].Similarto
12
howintestinalcryptstemcellsareregulatedbyaspecializedstemcellmicroenvironmentcalled
13
aniche,theTMEcanfostercancerstemcellsandthusdriverecurrenceandmetastasis(Box1)
14
[26,27],aswellasprovidemechanismsfordrugresistance[28,29].Itisnowacceptedthatthe
15
TMEisanactiveplayerintumourmalignizationandthatitprovidesafertilegroundtodelvefor
16
therapeutictargets[30,31].
17
The study of complex interactions between cancer cells and their environment inspires
18
parallelswiththeinterrelationshipsrecognizedinanecosystem.Theapplicationofecological
19
conceptstooncologymayhelpexplainwhyarigidfocusonepithelialcancercellsalonehasled
20
toahighnumberoffailednewdrugsandclinicalstrategies.Justasecologyistheframeworkof
21
choice for understanding the mechanisms of organismal evolution through natural selection,
22
ecology working on the level of cellular populations serves to clarify the dynamics of cancer
23
evolution. This includes natural selection due to limited resources as well as the artificial
6
1
selectionofsubclonesresistanttotherapy.Hence,thefitnessofacellularpopulation(orofa
2
set of cancer mutations) should be considered in the context of its local environment, which
3
consistsofcellularinteractions,physicalparameters(oxygenlevels,pH,mechanicalpressure),
4
andmoleculeslikeextracellularmatrixcomponents,growthfactorsandcytokines.
5
Moreover,ecologicaltypesofinteractioncandescribevariouslevelsofcrosstalkintumour
6
dynamics. Examples of competitive and cooperative interactions have been reported both
7
betweendifferentepithelialsubclonesandbetweenepithelialcancercellsandthestroma[32-
8
34].Additionally,theimmunesystemcanbeconvertedfromapredatortoanaccomplice(see
9
Box2).Indeed,thestromalenvironmentisnotstaticandco-evolutionisacompellingconcept:
10
parallel evolution of neoplastic epithelial cancer cells and cells in the microenvironment,
11
subject to epigenetic and gene expression changes [35]. Furthermore, the stromal
12
compartment contributes significantly to intratumoral heterogeneity, with important
13
implications for disease progression and therapeutic responses [36]. Thus, cancer ecology
14
integrates complex interactions between epithelial cancer cell populations and their
15
environmenttopromiseadeeperunderstandingofthebiologyofcancer[37,38].
16
17
ModulatingtheCRCecosystemfortherapeuticpurposes
18
The composition of the TME during colorectal tumorigenesis and in advanced cancers is
19
subject to intensifying inquiry (for a recent overview see refs: [39, 40]) and is found to be
20
different from normal intestinal stroma (see Figure 1, Key Figure). As recently reported, the
21
majorityofgenesthatpredictcancerrecurrenceinCRCpatientsareexpressedinCAFsandnot
22
bythecancercells[22,41].ThestrikingprognosticpoweroftheTMEraisestheopportunityto
23
more accurately estimate the risk of relapse of any given stage I-III patient. It also paves the
7
1
wayforthedevelopmentofadditionallyrefinedmolecularclassificationsbasedonthecontents
2
of the ecosystem. A relevant early example of such an ecological classification of CRC was
3
establishedbyGalonandothers[42-44],whoobservedthatinfiltrationofparticularsubsetsof
4
Tcellsinthetumourmasspredictslongerdiseasefreesurvivalintervalsaftertherapy.
5
Moreover,novelCRCclassificationsbasedonthetypeandfeaturesofstromalcellswillbe
6
key to stratify patients for treatments that target the TME. Based on the idea that CRC cells
7
dependonparticularstromalfactorstoeffectivelydisseminate,therapeuticsthatmodulatethe
8
CRCecosystemmaybeeffectiveintreatingorpreventingmetastasis.Anadditionaladvantage
9
of targeting the TME is its genetic stability, making drug resistance less likely to occur.
10
Furthermore,thecommonalityofmostelementsinthestromaacrosscancertypessuggestsa
11
broad applicability of effective therapies. A handful of therapies based on the above concept
12
have already been developed and are currently being applied. For instance, blood vessel
13
formation and endothelial cells have been successfully targeted by anti-angiogenic therapy
14
[45]. Indeed, a monoclonal antibody against vascular endothelial growth factor (VEGF, i.e.
15
bevacizumab) inhibits angiogenesis and has been shown to improve survival for stage IV
16
patients[46,47].Additionally,metronomicchemotherapy–aimingtoforestalldrugresistance
17
and suppress neo-angiogenesis – has shown promise for prolonging survival and limiting
18
metastasisinpreclinicalmodels,andisbeingtestedinclinicaltrials[48,49].
19
AnotherprimeexampleoftherapydirectedtotheTMEisimmunotherapy(Box2).Ithas
20
beenproposedthatseveral(chemo-)therapiesthatdonotdirectlytargetimmunity,suchas5-
21
fluorouracil and platinum compounds, in fact do rely on immune components. These
8
1
conventional treatments are effective at least in part by reinforcing tumour
2
immunosurveillance,especiallywhentheyinduceimmunogeniccancercelldeath[50].
3
4
TGF-βasamainorganizerofthemetastaticCRCecosystem
5
TGF-β has a dual role in CRC, with tumour suppressive functions in early stages and
6
promotingdiseaseprogressioninadvanceddisease(seeBox3).Ourgroupdemonstratedthat
7
elevatedTGF-βsignallingintheTMElinkstopoorprognosisinCRC[22,41].ThestromalTGF-β
8
responseencodesageneprogrammethatincludesaplethoraofcytokines,growthfactorsand
9
extracellularmatrixproteins,manyofwhichhavebeenshowntoplaykeyrolesduringdisease
10
progressionandmetastasisinseveralcancertypes.Amongthesepotentialtherapeutictargets
11
isinterleukin-11(IL-11),acytokinesecretedbyTGF-β-stimulatedfibroblaststhatinducespro-
12
survival response in CRC cells during cancer progression and metastatic colonization [41, 51].
13
The picture that emerges is that key functions required to complete the formation of
14
metastasisareprovidedbytheTGF-β-activatedmicroenvironment(Figure1).
15
Because disseminated tumour cells thrive on a TGF-β-activated environment, this
16
dependencycouldbeexploitedintheclinicalsettingtoimprovepatienttreatment;ourgroup
17
showedforthefirsttimethatpharmacologicalinhibitionofstromalTGF-βsignallingiseffective
18
in blocking metastatic colonization in mice [22, 41]. Besides activating a pro-metastatic
19
programme in CAFs [52], stromal TGF-β signalling induces a pro-tumorigenic phenotype in
20
neutrophils and macrophages [53-55]. In addition, TGF-β directly suppresses the anticancer
21
response of the adaptive immune system, including the deregulation of cytotoxic T
22
lymphocytes[56-58].Also,neutralizingtheTGF-βpathwayinTcellsledtoimmune-mediated
9
1
eradicationoftumoursinamodelofmelanoma[59].ThatthedualroleofTGF-βsignallingin
2
instructing immune cells and CAFs might be more related than is immediately obvious, is
3
suggestedbyreportinwhichCAFswereshowntohaveimmunesuppressiveeffects[60,61].
4
Thus, it will be interesting to assess the efficacy of pharmacological intervention in this
5
pathway on the CRC ecosystem. Several therapeutic strategies targeting TGF-β are in clinical
6
development (see Box 4) [62-64]. This spurs the hope that if we can block TGF-β
7
pharmacologically, we may prevent a vicious cycle of stroma activation, expression of pro-
8
tumorigenic secreted factors, and the generation of an immunosuppressive environment
9
(Figure 1). Indeed, TGF-β inhibition may work as (or synergize with other types of)
10
immunotherapy(Box2).
11
12
CaveatsaboutthedualroleofTGF-βsignallinginCRC
13
DuetothetumoursuppressivefunctionofTGF-βsignallinginepithelialcomponentofCRC
14
(see Box 3), elevated TGF-β levels necessary to instruct the poor prognosis-associated
15
microenvironmentmightnotbecompatiblewithtumourgrowth.Wespeculatethatthelossof
16
TGF-βresponseintheepithelialcomponentofthecancermayfacilitatetheelevationofTGF-β
17
levels during tumour progression. A functional link between these two events has been
18
observedinexperimentalmodels [65].InabiobankofCRCorganoids,independenceofTGF-β-
19
mediatedgrowthinhibitionwasassociatedwithadvancedstagesofthedisease[66],whereas
20
lossofSMAD4inepithelialCRCcellscorrelatedwithpoorprognosisinseveralstudies[67,68].
21
Yet,abrogationofthecytostaticTGF-βresponseinCRCcellsisnotexclusivelyexplainedbyloss-
22
of-function mutation in pathway components suggesting additional mechanisms of TGF-β
10
1
resistance [66]. Also, a proportion of TGF-β-reactive CRCs may respond to the cytokine by
2
undergoing EMT while bypassing the cytostatic effect. Apparently, this effect is relevant to
3
trigger invasion of a particular subset of early lesions named sessile serrated adenomas [69],
4
whichmightthereforebenefitfromanti-TGF-βtherapy.Conversely,itremainsunclearwhether
5
anti-TGF-β therapy would be safe for patients with cancer cells that have an intact TGF-β
6
pathway–eveniftheyrepresentasubclone–andarekeptincheckbyitscytostaticeffects.
7
Although the TGF-β pathway has emerged as a powerful architect of the pro-metastatic
8
poorprognosisTME,itseffectonindividualstromalcelltypesispleiotropicandincompletely
9
charted. Accordingly, we propose that, to treat the cancer ecosystem with more integrated
10
approaches, we need to study and understand its complexity in greater detail. As such,
11
manipulatingamasterregulatorlikeTGF-βinsufficientlycomplexmodelsystemsofmetastatic
12
CRCshouldnotonlyprovidetheneededrationaleforclinicaltranslation,butwouldadditionally
13
giveusrelevantparameterstohelpmapthecrosstalkinthecancerecosystem.
14
15
Needforbetterpreclinicalmodelsmovingforward
16
To study the efficacy of above-mentioned and other stroma-directed therapies in the
17
preclinicalsetting,thereisademandformorecomprehensive,predictivemodels.Althoughthe
18
fieldhasmovedfromsubcutaneouslyinjecting2D-culturedhumanCRCcelllinestoexploiting
19
3Dorganoidcultureandpatient-derivedxenografts(PDX)–approachesthatpromisetoretain
20
tumour heterogeneity in genetics, architecture and drug responses [70-72] – these in vivo
21
models lack an intact TME. This is due to a mismatch between some murine and human
22
signallingmoleculesandbecauseoftheabsentorabrogatedimmunesystemrequiredforthe
11
1
engraftment of human tumour tissue in murine hosts. To address these problems, mouse
2
modelsarebeinggeneratedthatcorrectknownsignallingdeficitsaswellasfeaturehumanized
3
immune system components, potentially leading to patient-specific immune environments.
4
Theseadvancespromisetostronglyenhancecurrentmodelsforcancerbiologyandpreclinical,
5
personalizedoncology.
6
An alternative approach to study the CRC TME and cancer immunity involves genetically
7
modified mouse models (GEMMs), which feature an intact, immunocompetent TME yet
8
typicallyfailtocaptureadvancedCRC.Thisleavesagap:totestcurrentideasanddiscoverand
9
develop novel therapeutic strategies we are in need of CRC models that are humanlike in
10
molecular characteristics, genetics and histopathology, and that are metastatic in a fully
11
immunocompetentenvironment.Theidealscenariowouldincludeatransplantable(organoid)
12
system in the commonly used C57BL/6 strain, so that it can be leveraged across a wealth of
13
existinggeneticmodelstodissectspecificfunctionsofstromalpathways.Furthermore,sucha
14
systemwouldbecriticaltotestingnoveltherapiesthattargetthetumourecosystem.
15
16
Concludingremarks
17
The CRC oncology field has evolved in the past decades. From histological analyses to
18
molecularsubtyping,fromcytotoxicchemotherapytopersonalizedcombinationswithtargeted
19
therapy.Whathasnotchanged,unfortunately,isthatbesidessurgerythereisverylittlewecan
20
dotoimproveoverallsurvivalforadvanceddisease.
21
However, the paradigm shift that points to the TME as a critical factor in determining
22
metastatic success opens the door to a more integrated understanding of the biology of
12
1
metastasis. One that invites us to consider cancer as a complex ecosystem rather than as a
2
tenacious weed. In this context, therapeutic targeting of stromal TGF-signalling might be
3
expectedtoreprogrammethepoorprognosiscancerecosystem(Figure1),withthepotential
4
to make metastatic lesions unsustainable or to prevent metastatic initiation altogether. We
5
hopethatthisemergingconceptwillfindatimelytranslationtotheclinic.Acknowledgingthe
6
many challenges ahead (see also Outstanding Questions), we need more sophisticated and
7
predictive CRC models to show proof-of-principle results that may catalyse the effort of
8
targetingtheTME.
9
10
OutstandingQuestions
11
• WhattriggerstheupregulationofTGF-βinsometumoursbutnotothers?
12
• What cell type is (or which cell types are) the source of secreted and activated TGF-β
13
thatreprogramstheCRCecosystem?
14
• As not all CRCs have known mutations in the TGF-β pathway, and some tumours may
15
haveco-existingmutantandsensitivesubclones,towhatextentistheresponsivenessof
16
epithelial cells to TGF-β (mainly associated to early disease stages) still relevant in
17
advancedCRC?
18
• Andwithinthiscontext,isTGF-βtherapysafeforallpatients?
19
• Given the dependency of metastatic initiation on a TGF-β-activated TME, is the TGF-β
20
therapeuticwindowlimitedtoadjuvanttherapy–toinhibitmetastaticcolonization–or
21
couldTGF-βtherapyalsotreatestablishedmetastases,e.g.asimmunotherapy?
22
13
1
Box1.CRCmetastasis:biologyandtherapeuticscope
2
Initial transformation of the colon epithelium and subsequent transitions through the
3
adenoma-carcinoma sequence are thought to require successive genetic alterations in 4
4
signalling pathways: WNT, EGFR, TGF-β and P53 [1, 2]. In the healthy colonic mucosa, these
5
signallingpathwaysregulatethebehaviourofintestinalstemcells(ISCs).Therefore,duringCRC
6
progression,tumourcellsbecomeindependentfromthesecryptnichesignals[66].About40%
7
of all CRC disseminate to other organs. Metastasis can either be present during diagnosis or
8
reveal itself months or years after surgery as a recurrence. In many cases, distant recurrence
9
targets the liver, at least in part because intestinal blood vessels drain into the portal vein,
10
whichconnectstotheliver.Metastasisisamultistepprocessthatrequirescellstodetachfrom
11
their epithelial neighbours, invade the submucosa, intravasate and survive in the vasculature
12
(bloodorlymphaticvessels),extravasateandsurviveinanalienorgan,toeventuallyreinitiatea
13
thrivingtumour[73,74].Thereismuchofthebiologyofmetastasisthatwedon’tunderstand,
14
butfromwhatweknow,itisaveryharshandinefficientprocess[75,76].Arguably,theprocess
15
selectsforthefiercestandmostresilientcancer(stem)cellsanddiscoveringwhatmakesthem
16
successfulmightuncovertherapeutictargets.Importantly,themetastaticprocesshasnotbeen
17
linked to specific genetic alterations within epithelial CRC cells [77]. Therefore, studies have
18
shiftedfocusontotheroleoftheTMEandthemechanismbywhichthemetastaticcellexploits
19
the TME for survival, immune evasion, and growth stimuli [78, 79]. Perhaps, success in
20
metastasisisdefinedaswhocanbestremodeltheirmicroenvironment.
21
14
1
Box2.Cancerinflammation,immunityandimmunotherapy
2
Inflammation is a well-described risk factor in gastrointestinal tumorigenesis [80], and
3
encompassesdisparatesignallingpathwaysbyavarietyofimmunecellsthatimpacteverystep
4
of cancer progression and metastasis [81]. Although the immune system can be a powerful
5
tumour suppressor by the coordinated elimination of aberrant cells, tumours find a way to
6
surviveinspiteofimmunosurveillance.Thereisevidenceforimmunoselectioninmanycancers,
7
includingCRC–wherethenumberofobservedneoantigens(inremainingsubclones)islower
8
thanexpectedbasedonmutationrates–andcancercellscanacquireresistancetoimmunity
9
bye.g.abrogatingantigenpresentation[82,83].
10
Moreover,ithasbecomeincreasinglyclearthatinflammatorymechanismsintumourscan
11
alsohelpdrivecancerprogression[81].Bystudyinghowprogressingcancerssuppresstheanti-
12
tumour response and subvert immune regulation to foster immune suppressor cells and
13
produce pro-tumorigenic factors that support cancer cells, the expansive field of cancer
14
immunology has introduced a multitude of novel therapeutic strategies [84, 85]. On the
15
conceptuallevel,manysuchtherapiesaimtoenhanceendogenousanticancerimmunitythatis
16
somehowinsufficientordysfunctional[86].Toachievethiseffect,atherapycanreducecellular
17
or molecular crosstalk mechanisms of immune suppression and/or increase T cell survival,
18
proliferation,infiltration,andactivationintheTME[87].
19
The promise of immuno-oncology has recently been demonstrated by long-lasting
20
responsestoimmunecheckpointinhibitors.Theseprovedbeneficialinvarioustypesofcancer
21
by unleashing T cells that were crippled by signalling either from immune suppressor cells or
22
cancer cells hijacking anti-inflammatory mechanisms [88, 89]. It is still unclear whether these
15
1
therapieswillbroadlybenefitCRCpatients.However,onestudyonmismatchrepair-deficient
2
(MSI) cancers, including of the colon, did show therapeutic responses [90]. This supports the
3
concept that tumour cells with higher frequency of neoantigens, such as in microsatellite
4
instable cancers, are generally more vulnerable to T cell-mediated anti-tumour immunity,
5
possiblyexplainingtheirrelativelygoodprognosis[91,92].Ifcurrentimmunotherapiesturnout
6
nottohaveabroadapplicabilityassingleagentsinCRC,thereisstillhopethatacombination
7
of treatments that both enhances intratumoral immune infiltration as well as activation will
8
farebetter[93,94].
9
10
Box3.Thedual,biphasicroleofTGF-βinCRC
11
Thepro-metastaticeffectsofTGF-βsignallingonthetumourmicroenvironmentcanoccur
12
independently of signalling in epithelial cancer cells, as many cancers silence the epithelial
13
pathwaysoastoprogress.TGF-βsignallingisconsideredatumoursuppressorpathwayincolon
14
carcinogenesisbecauseittriggersapotentcytostaticresponseinepithelialcells[22,65,95-97].
15
Indeed, about 40% of CRCs display loss-of-function mutations in TGF-β pathway components,
16
which are acquired around the adenoma carcinoma transition [2, 98]. An early clue for a
17
positive role for TGF-β signalling in disease progression was found by Kawata and colleagues
18
whoreportedthatserumlevelsofTGF-β1predictrelapseinCRCpatients[99].Later,ourgroup
19
showedthatTGFB1,TGFB2andTGFB3mRNAlevelsassociatewithshorterdiseasefreesurvival
20
intervalsaftertherapyinstageI-IIIpatients[41].ElevatedTGFB1-3expressioncorrelateswith
21
upregulationofTGF-βresponsegenesignatures(TBRS)incellsoftheTME,specificallyinTcells,
22
macrophages, endothelial cells and most prominently CAFs. These TBRSs hold a striking
16
1
predictivepowerforcancerrecurrenceinCRC[41]anddetectionbyimmunohistochemistryof
2
severalproteinsupregulatedinTGF-β-activatedCAFsidentifiespatientsathighriskofrelapse
3
[22].Furthermore,comparisonofthemolecularsubtypesrevealedthatthemainsignallingaxis
4
deregulated in the stromal/mesenchymal subtype (CMS4) is the TGF-β pathway [20]. A
5
significant proportion of the genes differentially expressed in CMS4 cancers that predict a
6
higherprobabilityofrecurrenceareincludedintheTBRSsandcorrespondtogenesinducedby
7
TGF-β in CAFs. The role of a TGF-β-activated ecosystem in disease progression has been
8
confirmed in experimental models of advanced CRC, where enforced TGF-β signalling in the
9
TMEstronglyincreasesmetastaticburden[41].
10
11
Box4.TGF-βtherapeuticsinclinicaldevelopment
12
Several methods of targeting the TGF-β pathway have been described. These include
13
antisense oligonucleotides to TGF-β ligand mRNA, small molecule inhibitors of the receptor
14
kinasedomains,andmonoclonalantibodiesagainstligandsorreceptors.Inaddition,thereare
15
vaccines and adoptive cell transfer strategies that target TGF-β signalling as part of their
16
mechanism. For an overview of all (pre-) clinical strategies, see references [62-64]. Here, we
17
selectedTGF-βtargetingtherapiesthatarecurrentlyinclinicaltrialsforCRC(TableI).
18
19
20
17
1
TableI.TGF-βtargetedtherapiesinclinicaltrialsforCRC
Type
Drug
MoA
Antisenseoligos
Trebedersen
ASO to TGF-β2
(AP12009 Antisense mRNA
Pharma)
Receptorinhibitors
Kinase
Galunisertib
TβRI(ALK5)kinase
inhibitors
(LY2157299EliLilly) inhibitor
MAbs
TEW-7197,
MedPactor
IMC-TR1
(LY3022859EliLilly)
PF-03446962
(Pfizer)
Setting(Phase)
TrialIdentifier
Melanoma,
pancreas,CRC(I)
NCT00844064
Rectalcancer(II)
Study
cancer
immunology in solid
tumours(I)
Checkpoint inhibitor
combination in solid
tumours(I/II)
TβRI(ALK5)kinase Solidtumours(I)
inhibitor
TβRII neutralizing Solidtumours(I)
MAb
ALK-1 neutralizing Metastatic CRC (I)
MAb
Solidtumours(I)
NCT02688712
NTT02304419
NCT02423343
NCT02160106
NCT01646203
NCT02116894
NCT00557856
2
3
4
5
Immunotherapy
Vaccines
FANG
vaccine shRNA
against MetastaticCRC(II)
NCT01505166
(Gradalis)
furin*
TAGvaccine
TGF-β2-antisense/ Metastatic
solid NCT00684294
GMCSF modified tumours(I)
autologous
tumourcells
MoA:Modeofaction.ASO:antisenseoligo.ALK1:Activinreceptor-likekinase1(non-canonical
TGF-βreceptortypeIinendothelialcells).*AspartofitsMoA,autologoustumourcellscarry
bifunctionalshRNAblockingfurin,leadingtolowerTGF-β1andTGF-β2levels.
6
7
18
1
2
3
Glossary
Adjuvantchemotherapy:chemotherapygivenafterprimarytreatment(surgeryinCRC)to
lowertheriskofthecancercomingback.
4
Cancer evolution: the theory that within a tumour, accumulating mutations and/or
5
localizedselectivepressureleadstothegenerationofsubclonesthatcompetewitheachother
6
forlimitedresources.Furthermore,atthetimeofdiagnosisa(minority)subclonemayalready
7
carryamutationthatrendersit(partially)resistanttochemotherapy.Boththisconceptandthe
8
realization that this theory helps explain the unique nature of each individual tumour make
9
cancerevolutionahighlyrelevantconceptthatformsthebasisformoreintegrated(ecological)
10
therapeuticstrategies.
11
CRC staging: The American Joint Committee on Cancer (AJCC) has established a general
12
cancerstagingprotocolcalledtheTNMStagingSystem.Tstandsfortumourandevaluatesthe
13
original (primary) tumour in grades of aggressiveness; N stands for lymph Node involvement
14
andqualifiesthepresenceofcancercellsspreadingtonearbylymphnodes.Maddressesthe
15
presence or absence of distant metastasis (spreading of cancer to distant organs). Based on
16
these3categories,patientsaregroupedintofourstagesindicatedbyRomannumerals(I,II,III
17
and IV). While statistically representing significantly different risk groups for recurrence and
18
cancer-relateddeath,thestagingsystemdoesnotaccuratelypredictonthelevelofindividual
19
patients.
20
21
Cytokines: large class of small secreted signalling proteins involved in cellular crosstalk,
especiallyrelevantfortheorchestrationofimmuneresponsesandcancer.
22
Ecology: the comprehensive study of the distribution, abundance and dynamics of
23
organisms, their interactions with others and with their physical environment. It is most
24
frequently applied on the organism level, where populations and their environment form an
25
ecosystemandthelargestsuchsystemstudiedistheecosphereearth,yetitcanalsobeapplied
26
to cells in a tissue or in a cancer. Central concepts in ecology include cooperation and
27
competition,parasitismandpredation,andevolution.
28
Immunosurveillance:theprocessbywhichmalignantcellsarekeptincheckbyanticancer
29
immunity. Cancer immunity is understood to have an initial elimination phase before the
19
1
balance is tipped and tumour cells start escaping from immunosurveillance. Between
2
eliminationandescape,theremaybeaprolongedstateofequilibriumbetweencancergrowth
3
andimmunity-relatedkilling.
4
Relapse/Recurrence:thereturnofatumouraftersurgicalremovaloftheoriginaltumour
5
(whichoftenentailedremovalofpartsorthewholeofthecolon).Thetumourcanreappear,
6
oftenasadistantmetastasis.
7
Metronomicchemotherapy:frequentorcontinuousadministrationoflownon-toxicdoses
8
with no interruptions for long periods to prevent resistance and target both epithelial cancer
9
cellsandthegrowingtumourvasculature.
10
MicroenvironmentorStroma:supportiveconnectivetissuewithstructuralandfunctional
11
roles both during homeostasis and in disruption such as wounding or disease. The stroma
12
includesfibroblasts,bloodandlymphaticvessels,immunecells,andtheextracellularmatrix.In
13
thecontextofcancer,thestromaisincreasinglyunderstoodasacomplex,dynamicentitythat
14
istransformedbyandco-evolveswithcancercellsandcandrivemalignantprogression.
15
Microsatellite instability (MSI): due to a deficiency in DNA mismatch-repair genes, MSI
16
patients accumulate spontaneous errors in regions of their genome that are repetitive and
17
therebychallengingtoreplicate.
18
20
1
Figurelegend
2
Figure 1 (Key Figure). The CRC ecosystem and the progression-driving role of TGF-β.
3
Besides fibroblasts (CAF), endothelial cells, pericytes and mesenchymal stem cells (MSC),
4
colorectal cancers are infiltrated by a variety of innate and adaptive immune cells, including
5
lymphocytes (T cells, B cells, natural killer cells), monocytes, macrophages, dendritic cells,
6
granulocytes (neutrophils, basophils, eosinophils, and mast cells), and myeloid-derived
7
suppressorcells(MDSC).
8
TGF-β has emerged as a central architect that drives malignization of the cancer ecosystem,
9
activating stromal cells towards pro-tumorigenic differentiation, inducing the expression of
10
secretedfactors,andfacilitatingtheaccumulationofimmunesuppressiveanddrugresistance
11
functionalities.EffectiveecologicaltreatmentmightincludeacombinationofTGF-βinhibition
12
withotherstroma-directedtherapies,suchaschemotherapyandtargeted(immuno-)therapy.
13
14
Acknowledgements
15
Workintheauthors’labhasbeensupportedbygrantSAF2014-53784_RandaJuandela
16
Cierva fellowship (D.V.F.T) from the Spanish Ministry of Economy and Competitiveness
17
(MINECO),andbytheDr.JosefSteinerFoundation.IRBBarcelonaistherecipientofaSevero
18
OchoaAwardofExcellencefromtheMINECO.WewouldliketothankElenaSanchoforcritical
19
readingofthismanuscript,aswellastheBatllelabforfruitfuldiscussions.
20
21
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
References
1Fearon,E.R.andVogelstein,B.(1990)Ageneticmodelforcolorectaltumorigenesis.Cell61,
759-767
2Fearon,E.R.(2011)Moleculargeneticsofcolorectalcancer.AnnuRevPathol6,479-507
3Kopetz,S.,etal.(2009)Improvedsurvivalinmetastaticcolorectalcancerisassociatedwith
adoptionofhepaticresectionandimprovedchemotherapy.JClinOncol27,3677-3683
4VanCutsem,E.,etal.(2014)Metastaticcolorectalcancer:ESMOClinicalPracticeGuidelines
fordiagnosis,treatmentandfollow-up.AnnOncol25Suppl3,iii1-9
5Gray,R.,etal.(2007)Adjuvantchemotherapyversusobservationinpatientswithcolorectal
cancer:arandomisedstudy.Lancet370,2020-2029
6Andre,T.,etal.(2009)Improvedoverallsurvivalwithoxaliplatin,fluorouracil,andleucovorin
asadjuvanttreatmentinstageIIorIIIcoloncancerintheMOSAICtrial.JClinOncol27,31093116
7Sinicrope,F.A.,etal.(2016)MolecularBiomarkersinthePersonalizedTreatmentofColorectal
Cancer.ClinGastroenterolHepatol14,651-658
8Bertotti,A.,etal.(2015)ThegenomiclandscapeofresponsetoEGFRblockadeincolorectal
cancer.Nature526,263-267
9Labianca,R.,etal.(2013)Earlycoloncancer:ESMOClinicalPracticeGuidelinesfordiagnosis,
treatmentandfollow-up.AnnOncol24Suppl6,vi64-72
10Carethers,J.M.andJung,B.H.(2015)GeneticsandGeneticBiomarkersinSporadicColorectal
Cancer.Gastroenterology149,1177-1190e1173
11 Mouradov, D., et al. (2013) Survival in stage II/III colorectal cancer is independently
predicted by chromosomal and microsatellite instability, but not by specific driver mutations.
AmJGastroenterol108,1785-1793
12 Samowitz, W.S., et al. (2005) Poor survival associated with the BRAF V600E mutation in
microsatellite-stablecoloncancers.CancerRes65,6063-6069
13 Jass, J.R. (2007) Classification of colorectal cancer based on correlation of clinical,
morphologicalandmolecularfeatures.Histopathology50,113-130
14 De Sousa E Melo, F., et al. (2013) Poor-prognosis colon cancer is defined by a molecularly
distinctsubtypeanddevelopsfromserratedprecursorlesions.NatMed19,614-618
15 Marisa, L., et al. (2013) Gene expression classification of colon cancer into molecular
subtypes:characterization,validation,andprognosticvalue.PLoSMed10,e1001453
16 Sadanandam, A., et al. (2013) A colorectal cancer classification system that associates
cellularphenotypeandresponsestotherapy.NatMed19,619-625
17 Roepman, P., et al. (2014) Colorectal cancer intrinsic subtypes predict chemotherapy
benefit, deficient mismatch repair and epithelial-to-mesenchymal transition. Int J Cancer 134,
552-562
18 Budinska, E., et al. (2013) Gene expression patterns unveil a new level of molecular
heterogeneityincolorectalcancer.JPathol231,63-76
19Schlicker,A.,etal.(2012)Subtypesofprimarycolorectaltumorscorrelatewithresponseto
targetedtreatmentincolorectalcelllines.BMCMedGenomics5,66
22
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
20Guinney,J.,etal.(2015)Theconsensusmolecularsubtypesofcolorectalcancer.NatMed
21,1350-1356
21Kalluri,R.andWeinberg,R.A.(2009)Thebasicsofepithelial-mesenchymaltransition.JClin
Invest119,1420-1428
22 Calon, A., et al. (2015) Stromal gene expression defines poor-prognosis subtypes in
colorectalcancer.NatGenet47,320-329
23 Isella, C., et al. (2015) Stromal contribution to the colorectal cancer transcriptome. Nat
Genet47,312-319
24Paget,S.(1889)TheDistributionofSecondaryGrotwhinCanceroftheBreast.Lancet133,
571-573
25 Fidler, I.J. (2003) The pathogenesis of cancer metastasis: the 'seed and soil' hypothesis
revisited.NatRevCancer3,453-458
26 Biswas, S., et al. (2015) Microenvironmental control of stem cell fate in intestinal
homeostasisanddisease.JPathol237,135-145
27 Medema, J.P. and Vermeulen, L. (2011) Microenvironmental regulation of stem cells in
intestinalhomeostasisandcancer.Nature474,318-326
28 Tredan, O., et al. (2007) Drug resistance and the solid tumor microenvironment. J Natl
CancerInst99,1441-1454
29Klemm,F.andJoyce,J.A.(2015)Microenvironmentalregulationoftherapeuticresponsein
cancer.TrendsCellBiol25,198-213
30 Fang, H. and Declerck, Y.A. (2013) Targeting the tumor microenvironment: from
understandingpathwaystoeffectiveclinicaltrials.CancerRes73,4965-4977
31Quail,D.F.andJoyce,J.A.(2013)Microenvironmentalregulationoftumorprogressionand
metastasis.NatMed19,1423-1437
32Hanahan,D.andCoussens,L.M.(2012)Accessoriestothecrime:functionsofcellsrecruited
tothetumormicroenvironment.CancerCell21,309-322
33 Marusyk, A., et al. (2014) Non-cell-autonomous driving of tumour growth supports subclonalheterogeneity.Nature514,54-58
34Cleary,A.S.,etal.(2014)Tumourcellheterogeneitymaintainedbycooperatingsubclonesin
Wnt-drivenmammarycancers.Nature508,113-117
35 Polyak, K., et al. (2009) Co-evolution of tumor cells and their microenvironment. Trends
Genet25,30-38
36 Junttila, M.R. and de Sauvage, F.J. (2013) Influence of tumour micro-environment
heterogeneityontherapeuticresponse.Nature501,346-354
37Merlo,L.M.,etal.(2006)Cancerasanevolutionaryandecologicalprocess.NatRevCancer
6,924-935
38Korolev,K.S.,etal.(2014)Turningecologyandevolutionagainstcancer.NatRevCancer14,
371-380
39Peddareddigari,V.G.,etal.(2010)Thetumormicroenvironmentincolorectalcarcinogenesis.
CancerMicroenviron3,149-166
40 Quante, M., et al. (2013) The gastrointestinal tumor microenvironment. Gastroenterology
145,63-78
41 Calon, A., et al. (2012) Dependency of colorectal cancer on a TGF-beta-driven program in
stromalcellsformetastasisinitiation.CancerCell22,571-584
23
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
42Naito,Y.,etal.(1998)CD8+Tcellsinfiltratedwithincancercellnestsasaprognosticfactor
inhumancolorectalcancer.CancerRes58,3491-3494
43Galon,J.,etal.(2006)Type,density,andlocationofimmunecellswithinhumancolorectal
tumorspredictclinicaloutcome.Science313,1960-1964
44Galon,J.,etal.(2014)Towardstheintroductionofthe'Immunoscore'intheclassificationof
malignanttumours.JPathol232,199-209
45 Carmeliet, P. and Jain, R.K. (2011) Molecular mechanisms and clinical applications of
angiogenesis.Nature473,298-307
46Mathonnet,M.,etal.(2014)Hallmarksincolorectalcancer:angiogenesisandcancerstemlikecells.WorldJGastroenterol20,4189-4196
47 Hurwitz, H., et al. (2004) Bevacizumab plus irinotecan, fluorouracil, and leucovorin for
metastaticcolorectalcancer.NEnglJMed350,2335-2342
48 Hackl, C., et al. (2013) Metronomic oral topotecan prolongs survival and reduces liver
metastasis in improved preclinical orthotopic and adjuvant therapy colon cancer models. Gut
62,259-271
49Pasquier,E.,etal.(2010)Metronomicchemotherapy:newrationalefornewdirections.Nat
RevClinOncol7,455-465
50 Zitvogel, L., et al. (2013) Mechanism of action of conventional and targeted anticancer
therapies:reinstatingimmunosurveillance.Immunity39,74-88
51 Putoczki, T.L., et al. (2013) Interleukin-11 is the dominant IL-6 family cytokine during
gastrointestinaltumorigenesisandcanbetargetedtherapeutically.CancerCell24,257-271
52 Calon, A., et al. (2014) TGF-beta in CAF-mediated tumor growth and metastasis. Semin
CancerBiol25,15-22
53Fridlender,Z.G.,etal.(2009)Polarizationoftumor-associatedneutrophilphenotypebyTGFbeta:"N1"versus"N2"TAN.CancerCell16,183-194
54 Gong, D., et al. (2012) TGFbeta signaling plays a critical role in promoting alternative
macrophageactivation.BMCImmunol13,31
55 Pickup, M., et al. (2013) The roles of TGFbeta in the tumour microenvironment. Nat Rev
Cancer13,788-799
56 Thomas, D.A. and Massague, J. (2005) TGF-beta directly targets cytotoxic T cell functions
duringtumorevasionofimmunesurveillance.CancerCell8,369-380
57Yang,L.,etal.(2010)TGF-betaandimmunecells:animportantregulatoryaxisinthetumor
microenvironmentandprogression.TrendsImmunol31,220-227
58 Chen, M.L., et al. (2005) Regulatory T cells suppress tumor-specific CD8 T cell cytotoxicity
throughTGF-betasignalsinvivo.ProcNatlAcadSciUSA102,419-424
59 Gorelik, L. and Flavell, R.A. (2001) Immune-mediated eradication of tumors through the
blockadeoftransforminggrowthfactor-betasignalinginTcells.NatMed7,1118-1122
60 Kraman, M., et al. (2010) Suppression of antitumor immunity by stromal cells expressing
fibroblastactivationprotein-alpha.Science330,827-830
61 Feig, C., et al. (2013) Targeting CXCL12 from FAP-expressing carcinoma-associated
fibroblastssynergizeswithanti-PD-L1immunotherapyinpancreaticcancer.ProcNatlAcadSci
USA110,20212-20217
62 Akhurst, R.J. and Hata, A. (2012) Targeting the TGFbeta signalling pathway in disease. Nat
RevDrugDiscov11,790-811
24
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
63 Neuzillet, C., et al. (2015) Targeting the TGFbeta pathway for cancer therapy. Pharmacol
Ther147,22-31
64 Smith, A.L., et al. (2012) Molecular pathways: targeting the TGF-beta pathway for cancer
therapy.ClinCancerRes18,4514-4521
65 Munoz, N.M., et al. (2006) Transforming growth factor beta receptor type II inactivation
inducesthemalignanttransformationofintestinalneoplasmsinitiatedbyApcmutation.Cancer
Res66,9837-9844
66Fujii,M.,etal.(2016)AColorectalTumorOrganoidLibraryDemonstratesProgressiveLossof
NicheFactorRequirementsduringTumorigenesis.CellStemCell18,827-838
67Yan,P.,etal.(2016)ReducedExpressionofSMAD4IsAssociatedwithPoorSurvivalinColon
Cancer.ClinCancerRes22,3037-3047
68Alazzouzi,H.,etal.(2005)SMAD4asaprognosticmarkerincolorectalcancer.ClinCancer
Res11,2606-2611
69 Fessler, E., et al. (2016) TGFbeta signaling directs serrated adenomas to the mesenchymal
colorectalcancersubtype.EMBOMolMed8,745-760
70Jung,P.,etal.(2011)Isolationandinvitroexpansionofhumancolonicstemcells.NatMed
17,1225-1227
71 Sato, T., et al. (2009) Single Lgr5 stem cells build crypt-villus structures in vitro without a
mesenchymalniche.Nature459,262-265
72 Siolas, D. and Hannon, G.J. (2013) Patient-derived tumor xenografts: transforming clinical
samplesintomousemodels.CancerRes73,5315-5319
73 Chambers, A.F., et al. (2002) Dissemination and growth of cancer cells in metastatic sites.
NatRevCancer2,563-572
74 Massague, J. and Obenauf, A.C. (2016) Metastatic colonization by circulating tumour cells.
Nature529,298-306
75Luzzi,K.J.,etal.(1998)Multistepnatureofmetastaticinefficiency:dormancyofsolitarycells
aftersuccessfulextravasationandlimitedsurvivalofearlymicrometastases.AmJPathol153,
865-873
76Chaffer,C.L.andWeinberg,R.A.(2011)Aperspectiveoncancercellmetastasis.Science331,
1559-1564
77Jones,S.,etal.(2008)Comparativelesionsequencingprovidesinsightsintotumorevolution.
ProcNatlAcadSciUSA105,4283-4288
78 Mlecnik, B., et al. (2016) The tumor microenvironment and Immunoscore are critical
determinantsofdisseminationtodistantmetastasis.SciTranslMed8,327ra326
79Steeg,P.S.(2016)Targetingmetastasis.NatRevCancer16,201-218
80 Terzic, J., et al. (2010) Inflammation and colon cancer. Gastroenterology 138, 2101-2114
e2105
81Grivennikov,S.I.,etal.(2010)Immunity,inflammation,andcancer.Cell140,883-899
82Rooney,M.S.,etal.(2015)Molecularandgeneticpropertiesoftumorsassociatedwithlocal
immunecytolyticactivity.Cell160,48-61
83Schumacher,T.N.andSchreiber,R.D.(2015)Neoantigensincancerimmunotherapy.Science
348,69-74
84Galluzzi,L.,etal.(2014)Classificationofcurrentanticancerimmunotherapies.Oncotarget5,
12472-12508
25
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
85 Finn, O.J. (2012) Immuno-oncology: understanding the function and dysfunction of the
immunesystemincancer.AnnOncol23Suppl8,viii6-9
86Palucka,A.K.andCoussens,L.M.(2016)TheBasisofOncoimmunology.Cell164,1233-1247
87 Joyce, J.A. and Fearon, D.T. (2015) T cell exclusion, immune privilege, and the tumor
microenvironment.Science348,74-80
88 Pardoll, D.M. (2012) The blockade of immune checkpoints in cancer immunotherapy. Nat
RevCancer12,252-264
89Topalian,S.L.,etal.(2015)Immunecheckpointblockade:acommondenominatorapproach
tocancertherapy.CancerCell27,450-461
90 Le, D.T., et al. (2015) PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N Engl J
Med372,2509-2520
91 Llosa, N.J., et al. (2015) The vigorous immune microenvironment of microsatellite instable
coloncancerisbalancedbymultiplecounter-inhibitorycheckpoints.CancerDiscov5,43-51
92 Kloor, M., et al. (2010) Immune evasion of microsatellite unstable colorectal cancers. Int J
Cancer127,1001-1010
93Sharma,P.andAllison,J.P.(2015)Immunecheckpointtargetingincancertherapy:toward
combinationstrategieswithcurativepotential.Cell161,205-214
94Vanneman,M.andDranoff,G.(2012)Combiningimmunotherapyandtargetedtherapiesin
cancertreatment.NatRevCancer12,237-251
95Massague,J.(2008)TGFbetainCancer.Cell134,215-230
96Markowitz,S.,etal.(1995)InactivationofthetypeIITGF-betareceptorincoloncancercells
withmicrosatelliteinstability.Science268,1336-1338
97 Takaku, K., et al. (1998) Intestinal tumorigenesis in compound mutant mice of both Dpc4
(Smad4)andApcgenes.Cell92,645-656
98 The Cancer Genome Atlas Network (2012) Comprehensive molecular characterization of
humancolonandrectalcancer.Nature487,330-337
99 Tsushima, H., et al. (2001) Circulating transforming growth factor beta 1 as a predictor of
livermetastasisafterresectionincolorectalcancer.ClinCancerRes7,1258-1262
26
Cancer activates Stroma
Immunotherapy
TGF-β inhibition
Stroma secretes
Protumorigenic factors
TME drives Immune
Evasion & Drug Resistance
Stromal therapy
Epithelial
Cancer cell
CAF
MDSC
Dendritic Cell
Monocyte
Macrophage
Endothelial cell
Pericyte
MSC
Lymphoctes
Granulocyte
Extracellular
Matrix
Téléchargement