8
ORGANOIDS Patient-derived organoids model treatment response of metastatic gastrointestinal cancers Georgios Vlachogiannis, 1 Somaieh Hedayat, 1 Alexandra Vatsiou, 2 Yann Jamin, 3 Javier Fernández-Mateos, 1,2 Khurum Khan, 1,4 Andrea Lampis, 1 Katherine Eason, 1 Ian Huntingford, 1 Rosemary Burke, 5 Mihaela Rata, 3 Dow-Mu Koh, 3,6 Nina Tunariu, 3,6 David Collins, 3 Sanna Hulkki-Wilson, 1 Chanthirika Ragulan, 1 Inmaculada Spiteri, 2 Sing Yu Moorcraft, 4 Ian Chau, 4 Sheela Rao, 4 David Watkins, 4 Nicos Fotiadis, 6 Maria Bali, 3,6 Mahnaz Darvish-Damavandi, 1 Hazel Lote, 1,4 Zakaria Eltahir, 1 Elizabeth C. Smyth, 4 Ruwaida Begum, 4 Paul A. Clarke, 5 Jens C. Hahne, 1 Mitchell Dowsett, 7 Johann de Bono, 8 Paul Workman, 5 Anguraj Sadanandam, 1 Matteo Fassan, 9 Owen J. Sansom, 10 Suzanne Eccles, 5 Naureen Starling, 4 Chiara Braconi, 4,5 Andrea Sottoriva, 2 Simon P. Robinson, 3 David Cunningham, 4 Nicola Valeri 1,4 * Patient-derived organoids (PDOs) have recently emerged as robust preclinical models; however, their potential to predict clinical outcomes in patients has remained unclear.We report on a living biobank of PDOs from metastatic, heavily pretreated colorectal and gastroesophageal cancer patients recruited in phase 1/2 clinical trials. Phenotypic and genotypic profiling of PDOs showed a high degree of similarity to the original patient tumors. Molecular profiling of tumor organoids was matched to drug-screening results, suggesting that PDOs could complement existing approaches in defining cancer vulnerabilities and improving treatment responses. We compared responses to anticancer agents ex vivo in organoids and PDO-based orthotopic mouse tumor xenograft models with the responses of the patients in clinical trials. Our data suggest that PDOs can recapitulate patient responses in the clinic and could be implemented in personalized medicine programs. H igh-throughput sequencing has been ex- tensively used in precision medicine to identify somatic mutations that can be exploited for cancer treatment and drug development (1). However, the limited role of genomic profiling in predicting response to targeted therapies and the limitations of pre- clinical models used for drug validation are important obstacles hampering the success of personalized medicine (2). Co-clinical trials are parallel studies in which drug responses in patients are matched to laboratory preclinical models to personalize treatment and understand mechanisms of chemosensitivity through func- tional genomics and reverse translation (3). Most co-clinical trials rely on the use of genetically engineered mouse models or patient-derived xenografts, posing logistic, ethical, and eco- nomic issues (4). LGR5 + stem cells can be isolated from a num- ber of organs and propagated as epithelial or- ganoids in vitro to study physiology and neoplastic transformation (5). Most studies on human colo- rectal cancer (CRC) organoids have been con- ducted on cultures derived from primary tumors (6). In contrast, examples of PDOs from meta- static cancer sites remain sparse (79). Further- more, very limited evidence is available on the ability of PDOs to predict response to treatment in the clinic (10). Here we present a living bio- bank of PDOs from heavily pretreated metastatic gastrointestinal cancer patients and show exam- ples of how the drug responses of these cancer organoids can be compared with those of the ac- tual patient. A total of 110 fresh biopsies from 71 patients enrolled in four prospective phase 1/2 clinical trials were processed between October 2014 and February 2017. In line with previous data (7), PDOs were grown from 70% of biopsies with a cellularity of 2+ and above, and their establish- ment rate strongly correlated with tumor cellu- larity in the parental biopsy (c 2 test, P < 0.0001). No inverse correlation was observed between PDO establishment rate and presence of necrosis (cutoff 20%). Tumor percentage is a key lim- iting factor for genomic and transcriptomic an- alyses. When the 60% threshold used in large sequencing studies of primary CRC (11) or gastro- esophageal cancer (GOC) (12) was applied in our cohort, we found no correlation between PDO take-up rate and tumor percentage, suggesting that PDOs can also be established in cases of a low tumor/stroma ratio, thus allowing the ex vivo expansion of the cancer population in samples that would have otherwise failed quality-control tests for next-generation sequencing (NGS). PDOs presented in this study were derived from ultrasound (n = 20), computed tomography (CT)guided (n = 7), or endoscopic (n = 2) biop- sies of metastatic CRC (mCRC; n = 16), metastatic GOC (mGOC; n = 4), and metastatic cholangio- carcinoma (n = 1) patients (fig. S1). Liver, pelvic, peritoneal, and nodal metastases of chemore- fractory patients were used to establish PDOs. In several cases, PDOs were established from se- quential biopsies at baseline (BL), at the time of best response [partial response (PR) or stable disease (SD)], and at the time of disease progres- sion (PD), as well as from multiregion biopsies (table S1). Histological evaluation revealed notable mor- phological similarities between PDOs and the patient biopsies from which they were originally derived (Fig. 1, A and B, and figs. S2A and S2B). Immunohistochemistry markers routinely used in the diagnosis of CRC (CDX-2 and CK7) showed that the parental tumors expression pattern was maintained in PDOs, even when derived from sequential biopsies during treatment (fig. S2, C to E). Similarly, amplification of oncogenic drivers such as ERBB2 (Fig. 1C and fig. S2F) and re- arrangements in FGFR2 (fig. S2G) were retained in PDOs from mGOC and metastatic cholangio- carcinoma, respectively. NGS was used to profile 151 cancer-related genes in both PDOs (n = 23) and their parental biopsies; archival material from primary cancer or pretreatment diagnostic biopsies was also sequenced for eight patients, and whole-genome sequencing (WGS) was performed for one PDO (tables S2 and S3). The molecular landscape of our PDOs (Fig. 1D) largely overlapped with that re- ported for mCRC and mGOC in the MSK-IMPACT study (1), with the exception of SRC and EGFR amplifications and ATM and BRCA2 mutations that were more frequent in our mCRC PDO co- hort (table S4). Overall, a 96% overlap in muta- tional spectrum was observed between PDOs and their parental biopsies (Fig. 1D), whereas intra- tumor heterogeneity was observed between ar- chival material (primary cancer) and metastatic deposits (biopsies or PDOs) (fig. S3A and table S2). PDOs were able to capture spatiotemporal intratumor heterogeneity when established from multiple biopsies at the time of disease progres- sion and when compared with PDOs established at the beginning of treatment (Fig. 1D, fig. S3A, and table S2). Similar results were observed for copy number alterations (CNAs) in PDOs and biopsies collected at different time points during treat- ment (figs. S3B and S4). WGS confirmed CNAs extrapolated from targeted NGS of PDOs or PDO- derived orthotopic tumors (PDO-xenografts) (figs. S3B and S4); CNAs detected in key oncogenic RESEARCH Vlachogiannis et al., Science 359, 920926 (2018) 23 February 2018 1 of 7 1 Division of Molecular Pathology, The Institute of Cancer Research, London, UK. 2 Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK. 3 Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and Royal Marsden Hospital, London, UK. 4 Department of Medicine, The Royal Marsden NHS Trust, London, UK. 5 Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, UK. 6 Department of Radiology, The Royal Marsden NHS Trust, London, UK. 7 Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital NHS Trust, London, UK. 8 Division of Clinical Studies, The Institute of Cancer Research, London, UK. 9 Department of Medicine, Surgical Pathology and Cytopathology Unit, University of Padua, Padua, Italy. 10 Cancer Research UK Beatson Institute, Glasgow, UK. *Corresponding author. Email: [email protected] Corrected 20 February 2018. See full text. on July 30, 2020 http://science.sciencemag.org/ Downloaded from

ORGANOIDS Patient-derived organoids model treatment response … · ORGANOIDS Patient-derived organoids model treatment response of metastatic gastrointestinal cancers Georgios Vlachogiannis,

  • Upload
    others

  • View
    4

  • Download
    0

Embed Size (px)

Citation preview

Page 1: ORGANOIDS Patient-derived organoids model treatment response … · ORGANOIDS Patient-derived organoids model treatment response of metastatic gastrointestinal cancers Georgios Vlachogiannis,

ORGANOIDS

Patient-derived organoids modeltreatment response of metastaticgastrointestinal cancersGeorgios Vlachogiannis,1 Somaieh Hedayat,1 Alexandra Vatsiou,2 Yann Jamin,3

Javier Fernández-Mateos,1,2 Khurum Khan,1,4 Andrea Lampis,1 Katherine Eason,1

Ian Huntingford,1 Rosemary Burke,5 Mihaela Rata,3 Dow-Mu Koh,3,6 Nina Tunariu,3,6

David Collins,3 Sanna Hulkki-Wilson,1 Chanthirika Ragulan,1 Inmaculada Spiteri,2

Sing Yu Moorcraft,4 Ian Chau,4 Sheela Rao,4 David Watkins,4 Nicos Fotiadis,6

Maria Bali,3,6 Mahnaz Darvish-Damavandi,1 Hazel Lote,1,4 Zakaria Eltahir,1

Elizabeth C. Smyth,4 Ruwaida Begum,4 Paul A. Clarke,5 Jens C. Hahne,1

Mitchell Dowsett,7 Johann de Bono,8 Paul Workman,5 Anguraj Sadanandam,1

Matteo Fassan,9 Owen J. Sansom,10 Suzanne Eccles,5 Naureen Starling,4

Chiara Braconi,4,5 Andrea Sottoriva,2 Simon P. Robinson,3

David Cunningham,4 Nicola Valeri1,4*

Patient-derived organoids (PDOs) have recently emerged as robust preclinical models;however, their potential to predict clinical outcomes in patients has remained unclear. Wereport on a living biobank of PDOs from metastatic, heavily pretreated colorectal andgastroesophageal cancer patients recruited in phase 1/2 clinical trials. Phenotypic andgenotypic profiling of PDOs showed a high degree of similarity to the original patient tumors.Molecular profiling of tumor organoids was matched to drug-screening results, suggestingthat PDOs could complement existing approaches in defining cancer vulnerabilitiesand improving treatment responses. We compared responses to anticancer agents ex vivo inorganoids and PDO-based orthotopic mouse tumor xenograft models with the responsesof the patients in clinical trials. Our data suggest that PDOs can recapitulate patient responsesin the clinic and could be implemented in personalized medicine programs.

High-throughput sequencing has been ex-tensively used in precision medicine toidentify somatic mutations that can beexploited for cancer treatment and drugdevelopment (1). However, the limited role

of genomic profiling in predicting response totargeted therapies and the limitations of pre-clinical models used for drug validation areimportant obstacles hampering the success ofpersonalized medicine (2). Co-clinical trials areparallel studies in which drug responses inpatients are matched to laboratory preclinicalmodels to personalize treatment and understandmechanisms of chemosensitivity through func-tional genomics and reverse translation (3). Mostco-clinical trials rely on the use of genetically

engineered mouse models or patient-derivedxenografts, posing logistic, ethical, and eco-nomic issues (4).LGR5+ stem cells can be isolated from a num-

ber of organs and propagated as epithelial or-ganoids in vitro to study physiology andneoplastictransformation (5). Most studies on human colo-rectal cancer (CRC) organoids have been con-ducted on cultures derived from primary tumors(6). In contrast, examples of PDOs from meta-static cancer sites remain sparse (7–9). Further-more, very limited evidence is available on theability of PDOs to predict response to treatmentin the clinic (10). Here we present a living bio-bank of PDOs from heavily pretreatedmetastaticgastrointestinal cancer patients and show exam-ples of how the drug responses of these cancerorganoids can be compared with those of the ac-tual patient.A total of 110 fresh biopsies from 71 patients

enrolled in four prospective phase 1/2 clinicaltrials were processed between October 2014 andFebruary 2017. In line with previous data (7),PDOs were grown from 70% of biopsies with acellularity of 2+ and above, and their establish-ment rate strongly correlated with tumor cellu-larity in the parental biopsy (c2 test, P < 0.0001).No inverse correlation was observed betweenPDOestablishment rate and presence of necrosis(cutoff ≥ 20%). Tumor percentage is a key lim-iting factor for genomic and transcriptomic an-alyses. When the 60% threshold used in large

sequencing studies of primary CRC (11) or gastro-esophageal cancer (GOC) (12) was applied in ourcohort, we found no correlation between PDOtake-up rate and tumor percentage, suggestingthat PDOs can also be established in cases of alow tumor/stroma ratio, thus allowing the ex vivoexpansion of the cancer population in samplesthat would have otherwise failed quality-controltests for next-generation sequencing (NGS).PDOs presented in this study were derived

fromultrasound (n= 20), computed tomography(CT)–guided (n = 7), or endoscopic (n = 2) biop-sies ofmetastatic CRC (mCRC; n= 16),metastaticGOC (mGOC; n = 4), and metastatic cholangio-carcinoma (n = 1) patients (fig. S1). Liver, pelvic,peritoneal, and nodal metastases of chemore-fractory patients were used to establish PDOs.In several cases, PDOs were established from se-quential biopsies at baseline (BL), at the time ofbest response [partial response (PR) or stabledisease (SD)], and at the time of disease progres-sion (PD), as well as frommultiregion biopsies(table S1).Histological evaluation revealed notable mor-

phological similarities between PDOs and thepatient biopsies from which they were originallyderived (Fig. 1, A and B, and figs. S2A and S2B).Immunohistochemistry markers routinely usedin the diagnosis of CRC (CDX-2 and CK7) showedthat the parental tumor’s expression pattern wasmaintained in PDOs, even when derived fromsequential biopsies during treatment (fig. S2, Cto E). Similarly, amplification of oncogenic driverssuch as ERBB2 (Fig. 1C and fig. S2F) and re-arrangements in FGFR2 (fig. S2G) were retainedin PDOs frommGOC andmetastatic cholangio-carcinoma, respectively.NGS was used to profile 151 cancer-related

genes in both PDOs (n = 23) and their parentalbiopsies; archival material from primary canceror pretreatment diagnostic biopsies was alsosequenced for eight patients, andwhole-genomesequencing (WGS) was performed for one PDO(tables S2 and S3). Themolecular landscape of ourPDOs (Fig. 1D) largely overlapped with that re-ported formCRCandmGOC in theMSK-IMPACTstudy (1), with the exception of SRC and EGFRamplifications and ATM and BRCA2mutationsthat were more frequent in our mCRC PDO co-hort (table S4). Overall, a 96% overlap in muta-tional spectrumwas observed between PDOs andtheir parental biopsies (Fig. 1D), whereas intra-tumor heterogeneity was observed between ar-chival material (primary cancer) and metastaticdeposits (biopsies or PDOs) (fig. S3A and tableS2). PDOs were able to capture spatiotemporalintratumor heterogeneity when established frommultiple biopsies at the time of disease progres-sion and when compared with PDOs establishedat the beginning of treatment (Fig. 1D, fig. S3A, andtable S2). Similar results were observed for copynumber alterations (CNAs) in PDOs and biopsiescollected at different time points during treat-ment (figs. S3B and S4). WGS confirmed CNAsextrapolated from targeted NGS of PDOs or PDO-derived orthotopic tumors (PDO-xenografts) (figs.S3B and S4); CNAs detected in key oncogenic

RESEARCH

Vlachogiannis et al., Science 359, 920–926 (2018) 23 February 2018 1 of 7

1Division of Molecular Pathology, The Institute of CancerResearch, London, UK. 2Centre for Evolution and Cancer, TheInstitute of Cancer Research, London, UK. 3Cancer ResearchUK Cancer Imaging Centre, Division of Radiotherapy andImaging, The Institute of Cancer Research and RoyalMarsden Hospital, London, UK. 4Department of Medicine,The Royal Marsden NHS Trust, London, UK. 5CancerResearch UK Cancer Therapeutics Unit, The Institute ofCancer Research, London, UK. 6Department of Radiology,The Royal Marsden NHS Trust, London, UK. 7Ralph LaurenCentre for Breast Cancer Research, Royal Marsden HospitalNHS Trust, London, UK. 8Division of Clinical Studies, TheInstitute of Cancer Research, London, UK. 9Department ofMedicine, Surgical Pathology and Cytopathology Unit,University of Padua, Padua, Italy. 10Cancer Research UKBeatson Institute, Glasgow, UK.*Corresponding author. Email: [email protected]

Corrected 20 February 2018. See full text. on July 30, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 2: ORGANOIDS Patient-derived organoids model treatment response … · ORGANOIDS Patient-derived organoids model treatment response of metastatic gastrointestinal cancers Georgios Vlachogiannis,

Vlachogiannis et al., Science 359, 920–926 (2018) 23 February 2018 2 of 7

H&

EE

RB

B2

CIS

HE

RB

B2

IHC

Tissue PDOsF-013

H&

E

PD

Os

Tis

sue

R-006

Colorectal

C-0

01

C-0

02

C-0

03

C-0

04

R-0

05

R-0

06

R-0

07

R-0

08

R-0

09 B

L

R-0

09 P

D A

R-0

09 P

D B

R-0

10

R-0

11 B

L

R-0

11 P

D

F-0

12

F-0

14 B

L

F-0

14 P

D

F-0

15

F-0

16

APCKRAS

SMAD4TP53SRC

PIK3CASMARCA4

BRCA2FGFR1

MYCABCB1

RB1EGFRFLT1

AKT3MAP3K1

MAPK1ATM

BRAFCDH1

CDKN2AFBXW7FGFR2FGFR3

KITMAP2K1

NF1PIK3R1

ROS1AKT2ALK

ERBB4MET

NRASAKT1HRAS

R-0

11 S

D

Colorectal Gastroesophageal

Missense/Nonsense/Indel

Missense/Nonsense/Indel & Amplification/Gain

Amplification/GainDeletion

Missense/Nonsense/Indel & Deletion

R-009

PDOs TissueBL (p

.04)

BL (p.0

6)

PD A

PD B BL

PD A

PD B

0

2

4

6

PIK

3CA

co

py

nu

mb

er

6.2

12.5

25.0

50.0

100.

00.0

0.5

1.0

GDC0980 (nM)

BL (N=3)

PD A (N=3)

PD B (N=3)

115

5 12

Tissue PDOs

Gastroesophageal

PD

Os

F-012(Diffuse)

F-015(Intestinal)

Tis

sue

Viability in response to palbociclib

RB1Log2R

Total

TotalP<0.005

426

01212

41418

+MK-2206 (1 µM):

pAKT

pERK1/2

Cleavedcaspase-3

pPRAS40

Actin

PARP

AKT

PRAS40

ERK1/2

+-R-0

06AKT1-w

t

-

F-016

AKT1 E

17K; A

KT1-am

p

Vemurafenib (1 µM):

pMEK1/2

pERK1/2

pAKT

Actin

AKT

MEK1/2

ERK1/2

+-

C-003

BRAF-wt

+-

C-004

BRAF V60

0E

pEGFR

EGFR

pC-RAF

Actin

PARP

Lapatinib (1 µM): +-

F-014

BL

ERBB2-wt; E

GFR-am

pF-0

13ERBB2-

amp

+-

pERK1/2

pAKT

EGFR

AKT

pERBB2

ERK1/2

pEGFR

ERBB2

ERBB2(Long exposure)

Rel

ativ

e vi

abili

ty

50 µm 20 µm

20 µm20 µm

50 µm

0 µm 250

100 µm 100 µm100 µm

100 µm

100 µm

50 µm50 µm

50 µm

50 µm0 100µm

0 µm 100

Fig. 1. Histopathological, molecular, and functional characterizationof patient-derived organoids (PDOs). (A) Phase-contrast image ofa mCRC PDO culture (top) and hematoxylin and eosin staining comparingorganoids to their matching patient biopsy (bottom). (B) Intestinaland diffuse growth patterns are retained in mGOC PDOs. (C) ERBB2amplification and overexpression in mGOC PDOs and parental tissuebiopsy (CISH, chromogenic in situ hybridization; IHC, immunohisto-chemistry). (D) Heatmap showing the most frequently mutated and/orcopy number–altered genes in PDOs (left) and Venn diagram demon-strating 96% mutational overlap between PDOs and parental tissuebiopsies (right). (E) Target engagement in genotype–drug phenotypecombinations: pathway analysis downstream of ERBB2 in ERBB2-amplified and nonamplified PDOs treated with lapatinib (24 hours)(right), BRAF inhibition (24 hours) (center), and AKT inhibition (4 hours)

(left). wt, wild type. (F) Concentration-dependent effect of thedual PI3K/mTOR inhibitor GDC-0980 in three PDOs from patientR-009, all carrying an acquired PIK3CA mutation (H1047R). PDOsestablished from a liver metastasis biopsied at disease progression(R-009 PD-A) that also harbored PIK3CA amplification showedconcentration-dependent response to GDC-0980. PIK3CA-mutant butnonamplified PDOs established before regorafenib treatment (R-009 BL)or from a different liver metastasis biopsied at disease progression(R-009 PD-B) did not respond to GDC-0980. Viability data shownare means ± SEM of indicated independent experiments. (G) Correlation(Fisher’s exact test) between presence of RB1 amplification in PDOs(D) and response to the CDK4/CDK6 inhibitor palbociclib in thereported drug screen (fig. S9A). BL, baseline; SD, stable disease; PD,posttreatment/progressive disease.

RESEARCH | REPORT

Corrected 20 February 2018. See full text. on July 30, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 3: ORGANOIDS Patient-derived organoids model treatment response … · ORGANOIDS Patient-derived organoids model treatment response of metastatic gastrointestinal cancers Georgios Vlachogiannis,

drivers were further validated by digital-dropletpolymerase chain reaction (fig. S5). High con-cordance was observed in mutational, CNA, andtranscriptomic profiling over successive passageswhen PDOswere tested before and after severalmonths of continuous culture [passage range,5 to 13; mutations, coefficient of determination(R2) = 0.96,P< 0.0001; CNA,R2 = 0.97,P<0.0001;gene expression (RNA sequencing), R2 = 0.7,P < 0.001] (fig. S6).Next we tested the feasibility of using PDOs

derived from metastatic cancers as drug-screeningtools and validated the robustness of our approachby identifying several genotype–drug phenotype

correlations across the PDOpanel.We ran three-dimensional (3D) screening assays over a periodof 2 weeks (figs. S7 and S8), using a library of55 drugs now in phase 1 to 3 clinical trials or inclinical practice (table S5). The heatmap shownin fig. S9A summarizes the screening data; hitvalidation at lower drug concentrations is re-ported in fig. S9B. For all 19 screens, a very highcorrelation was observed among each screen’sthree replicate assays and controls (fig. S10).F-013 was the only ERBB2-amplified PDO in

our cohort (Fig. 1C), and it exhibited the strongestresponse to lapatinib [dual ERBB2/epidermalgrowth factor receptor (EGFR) inhibitor]; lapatinib

potently inhibited themitogen-activated proteinkinase (MAPK) andphosphatidylinositol 3-kinase(PI3K)/AKT signaling downstream of EGFR/ERBB2, inducing apoptosis in the F-013 PDO (Fig.1E and fig. S9A). In a PDO (F-014) inwhichEGFRwas amplified but ERBB2was not, lapatinib hadno effect on viability and onlymodestly reducedMAPK and PI3K/AKT signaling (Fig. 1E andfig. S9A).Similarly, across all PDOs, F-016 was the only

tumor carrying an AKT1 amplification and E17Kmutation (E, glutamic acid;K, lysine) (Fig. 1D) andwas the only one to respond strongly to both AKTinhibitors present in the drug library (MK-2206

Vlachogiannis et al., Science 359, 920–926 (2018) 23 February 2018 3 of 7

F-014 BL F-014 PR F-014 PD Paclitaxel (nM): DMSO

1.25

2.5

5 10

Actin

PARP

pHistone H3

pH2A.X

DMSO

1.25

2.5

5 10

F-014 BL F-014 PD

F-014 BL F-014 PD

DM

SO

Pac

litax

el (

2.5

nM)

BL PR PD0

20

40

60

80

100

120

RE

CIS

T 1.

1 (%

)

Total tumor burdenSeg. III (used for PDOs)

0.31

0.62

1.25

2.50

5.00

10.0

00.0

0.5

1.0

Paclitaxel (nM)

Rel

ativ

e vi

abili

ty F-015 (N=3)

F-014 BL (N=4)

F-012 (N=3)

F-014 PD (N=4)

0.6

1.2

2.5 5 10 20 40

0.0

0.5

1.0

1.5

Cetuximab (ug/ml)

Rel

ativ

e vi

abili

ty

C-001

C-002

C-003

C-004

R-007

KRAS/BRAF-wt

KRAS/BRAF-mut

DPYDAPCAPC

PTPN11TP53

Arc

hiva

l FF

PE

Tum

or

p.04

Tissue

PDOs

C-002

0 1VAF

PIK3CAPIK3CAPIK3CA

APCAPCAPCAPCAPCAPC

KRASTP53

Tissue

Arc

hiva

l FF

PE

Tum

or

p.04

p.07

PDOs

C-003

DPYDERBB4

KDRLAMA2ABCB1

BRAFCHD7

CREBBPTP53TP53TP53

SMAD4

Arc

hiva

l FF

PE

Tum

or

p.06

Tissue

PDOs

C-004

ResponseAcquired resistancePrimary resistanceAcquired resistance

Patient clinical outcome at time of/following biopsyF-014

Fig. 2. PDO-based ex vivo co-clinical trials in mGOC and mCRC.(A) PDOs were generated from sequential biopsies of a liver metastasis(red circles in the bottom panel) of mGOC patient F-014 that showedinitial response to paclitaxel (F-014 BL) and subsequently progressed(F-014 PD). Violet bars indicate overall tumor volume [according toRECIST (Response Evaluation Criteria in Solid Tumors) 1.1], andred bars indicate volume of the target metastasis used to generatePDOs. (B) Cell viability upon paclitaxel treatment was compared in BLand PD PDOs from patient F-014 and PDOs from patients that exhibitedprimary (F-015) or acquired (F-012) resistance to paclitaxel in the clinic.Viability data shown are means ± SEM of indicated independentexperiments. (C) Cell cycle analysis upon paclitaxel treatment in the

F-014 BL PDO compared with the F-014 PD PDO. DMSO, dimethylsulfoxide. (D) Concentration-dependent DNA damage was observed in theF-014 BL PDO in response to paclitaxel but not in PDOs from the samepatient established at PD. (E) PDOs were established from BL (C-003and C-004) and PD (C-001 and C-002) biopsies from patients treatedwith the anti-EGFR monoclonal antibody cetuximab. PDOs weretreated with cetuximab in vitro; data shown are means ± SD fromindependent experiments performed in triplicate. (F) Molecular analysisof BL and PD PDOs, matching biopsy (tumor), and primary bowelcancer (archival); arrows indicate the presence of clonal or subclonalmutations in BRAF or KRAS, respectively, in two patients. VAF, variantallele frequency; FFPE, formalin-fixed paraffin-embedded.

RESEARCH | REPORT

Corrected 20 February 2018. See full text. on July 30, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 4: ORGANOIDS Patient-derived organoids model treatment response … · ORGANOIDS Patient-derived organoids model treatment response of metastatic gastrointestinal cancers Georgios Vlachogiannis,

Vlachogiannis et al., Science 359, 920–926 (2018) 23 February 2018 4 of 7

R-009(Primary resistance)

Day

0D

ay 1

5

R-005(Response)

DCE-MRI CD31 IHCDCE-MRI CD31 IHC

KRAS-mutmetastatic CRC

Day 15 scanPost-treatment

scanPre-treatment

scan

Core biopsy fromsuitable site (BL)

Core biopsy at response or SD

Core biopsy atprogression (PD)

Regorafenib, 160 mg daily3 weeks on/1 week off, until disease progression

0

0.4

Ktrans (min-1)

R-005PDO-xeno

Vehicl

e (n=6

)

Regora

fenib

(n=6

)

Vehicl

e (n=3

)

Regora

fenib

(n=3

)0

5

10

15

Tum

or-

asso

ciat

ed C

D31

sco

re

R-009PDO-xeno

NS

NS

p=0.03

Day 0

Day 5

Day 0

Day 5

0

5

10

15

20fB

V (%

)p=0.04

Vehicl

e

Regora

fenib

0

5

10

15

Tum

or-

asso

ciat

ed C

D31

sco

re

p=0.02

5 10 15

-10

0

10

20

30

Tumor-associated CD31 score

MR

I fB

VD

ay5

(%) R2=0.64

p=0.006

Vehicl

e (n=5

)

Regora

fenib

(n=5

)0

1

2

3

Nec

rosi

s

p=0.008

R-011 BL/PDliver orthotopic

injections

Vehicle

Regorafenib

SurvivalSu

CD31 IHC

CD31 IHC

Survival

R-011

BL

R-011

SD

R-011

PD

0

2

4

6

8

Pat

ien

t CD

31 s

core

0 20 40 60 80 1000

50

100

Days post treatment

Per

cen

t su

rviv

al

Regorafenib (n=5)Vehicle (n=5)

p=0.04

R-011 BLPDO-xenografts

0 50 100 1500

50

100

Days post treatment

Per

cen

t su

rviv

al

Regorafenib (n=5)Vehicle (n=5)

NS

R-011 PDPDO-xenografts

Veh

icle

Reg

oraf

enib

R-011 BLPDO-xenografts

R-011 PDPDO-xenografts

CD31 IHC

Vehicl

e (n=4

)

Regora

fenib

(n=4

)0

2

4

6

Tum

or-

asso

ciat

ed C

D31

sco

re

Vehicl

e (n=3

)

Regora

fenib

(n=6

)0

2

4

6

8

Tum

or-

asso

ciat

ed C

D31

sco

rep=0.001 NS

RegorafenibVehicle

100 µm 100 µm

100 µm100 µm

Fig. 3. PDO-based co-clinical trials mimic primary and acquiredresistance to regorafenib in mice. (A) mCRC patients on regorafenibtreatment underwent biopsies at BL, SD, or PD. An early reduction(15 days) in functional imaging (DCE-MRI) parameters correlated withchanges in microvasculature assessed by CD31 staining and clinical benefitfrom regorafenib (right). Arrowheads indicate CRC metastases; Ktrans,volume transfer constant. (B) Changes in microvasculature in response toregorafenib were assessed in PDO-xenografts in mice by quantificationof tumor-associated CD31-positive vessels. Data show PDO-xenograftsfrom a primary resistant patient (R-009) and a long-term responder(R-005) to regorafenib. Means ± SD from the indicated number of mice(n) in a representative experiment are shown; significance was determinedusing Student’s unpaired t test. (C) Reduction in fractional blood volume(fBV) in regorafenib-treated mice carrying long-term regorafenibresponder (R-005) PDO-xenografts. A total of 10 animals were analyzed(five in each arm); shown are the means ± SD of an individual experiment.Day 0 fBV values could not be obtained for two animals owing torespiratory movement. Significance was determined using Student’spaired t test for fBV and unpaired t test for CD31 and necrosis.

(D) Schematic representation of the animal experiment using PDOsfrom patient R-011, established pre- and posttreatment with regorafenib.Mice carrying liver orthotopic R-011 pretreatment (BL) and posttreatment(PD) PDO-xenografts were randomized to control and treatment armsand treated with vehicle or regorafenib for 10 days. After treatment,each arm was further randomized to a cohort culled for histopathologicalanalysis and a survival cohort, which was monitored over time.(E) CD31 immunostaining in the parental patient BL, SD, and PDbiopsies, demonstrating an initial reduction in tumor microvasculaturein response to regorafenib. Shown are means ± SD calculated by scoring10 high-power-field tumor areas. (F) Representative images (top) andanalysis (bottom) of CD31 immunostaining in the BL and PD R-011PDO-xenografts. Shown are means ± SD calculated by scoring at least10 high-power-field tumor areas per animal in an individual experiment;n, number of animals analyzed in each group. Significance was determinedusing Student’s unpaired t test. (G) Kaplan-Mayer curves for regorafenib-or vehicle-treated mice bearing BL and PD PDO-xenografts from patientR-011 from an individual experiment (n, number of mice analyzed).Significance was determined using the Mantel-Cox log-rank test.

RESEARCH | REPORT

Corrected 20 February 2018. See full text. on July 30, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 5: ORGANOIDS Patient-derived organoids model treatment response … · ORGANOIDS Patient-derived organoids model treatment response of metastatic gastrointestinal cancers Georgios Vlachogiannis,

andGSK690693) (Fig. 1E and fig. S9A). OnemCRCPDO (C-004) harboredaBRAFV600Emutation (V,valine) (Fig. 1D) andwas the onlyPDO that showedsignificantly decreased viability after treatmentwith the BRAF inhibitor vemurafenib (fig. S9A).Consistent with this, vemurafenib selectively in-hibitedMEK/ERK (MAPK kinase/extracellularsignal–regulated kinase) signaling in the C-004PDO (Fig. 1E) but failed to induce apoptosis, inkeeping with the lack of efficacy of single-agentBRAF inhibitors in mCRC (13).Overall,PIK3CAmutationswere not predictive

of response to GDC-0980 (a dual PI3K/mTOR in-hibitor) in the PDOs panel (Fig. 1D and fig. S9A). Inline with this observation, in a patient fromwhompre- and posttreatment PDOs were establishedfrommultiple metastases (R-009 BL, PD-A, andPD-B), a PIK3CA H1047R mutation (H, histidine;R, arginine) common to all the PDOswas not asso-ciated with any response to GDC-0980. However,PDOscarryinga synchronousPIK3CAamplification(R-009 PD-A) showed a concentration-dependentreduction in cell viability in response to GDC-0980 (Fig. 1F and fig. S3A). Last, in keeping with

published data (14), a significant correlation wasobserved between RB1 amplification and sen-sitivity of PDOs to palbociclib [cyclin-dependentkinase 4 (CDK4)/CDK6 inhibitor] (Fig. 1G).After extensive molecular and functional char-

acterization of our PDOs, we examined their clin-ical predictive value in 21 comparisons of clinicalresponses observed in patients with ex vivo re-sponse data gathered in organoids (table S6).Taxanes are a standard second-line treatmentoption for metastatic gastric cancer; however,efficacy is modest, and no predictive biomarkersare available to inform clinical decisions (15). Wecompared response to paclitaxel in sequentialPDOs established before and after treatment ina paclitaxel-sensitive patient (F-014) with thatin PDOs established from liver metastases of twopaclitaxel-resistant patients (Fig. 2, A and B).PDOs derived from the responsive metastasisshowed aGI50 (concentration that inhibits growthof cancer cells by 50%) for clinically relevantpaclitaxel concentrations (16) that was about one-fourth that for PDOs from the same patientderived at progression; these resistant PDOs dem-

onstrated an identical paclitaxel concentration-response profile to the two PDOs established frompaclitaxel-refractory patients (Fig. 2B). Cell cycleanalysis showed marked apoptosis and G2 arrestupon taxane treatment in the pretreatment F-014PDOs, whereas no significant difference wasobserved in PDOs established at progression(Fig. 2C and fig. S11A). Similarly, paclitaxel in-duced concentration-dependent DNA damage,mitotic arrest, and apoptosis in the pretreatmentF-014 PDOs but had a much weaker impact onthe progression (and thus resistant) PDOs (Fig.2D). Consistent with data observed for second-line treatment, a ~10-fold difference in GI50 wasobserved in response to the combination of5-fluorouracil and cisplatin in PDOs collectedfrom chemosensitive and chemorefractorymGOCpatients receiving first-line treatment (fig. S11B),highlighting the clinical potential of PDOs fortreatment selection in cancers of unmet need.Anti-EGFRmonoclonal antibodies, regorafenib,

andTAS-102areU.S.FoodandDrugAdministration–approvedoptions for treatment of chemorefractorymCRC;however,with the exceptionofRASpathway

Vlachogiannis et al., Science 359, 920–926 (2018) 23 February 2018 5 of 7

Seg. 2

Seg. 5

0

10

20

30

40

Tum

or

dia

met

er (m

m)

Pre-treatment

Post-treatment

0.06

0.12

0.25 0.

5 1 2 40.0

0.5

1.0

TAS-102 (uM)

Rel

ativ

e vi

abili

ty

R-019 BL Seg.2 (N=2)

R-019 PD Seg.2 (N=1)

R-019 PD Seg.5 (N=2)

Seg

. 2S

eg. 5

R-019Pre-treatment Post-treatment

TK

1 IH

C

R-019

BL Seg.2 PD Seg.2 PD Seg.5

1 20.0

0.5

1.0

TAS-102 (uM)

Rel

ativ

e vi

abili

ty

R-019 PD Seg.5 (N=2)

R-017 (N=2)

R-018 (N=1)

R-019 BL Seg.2 (N=2)

R-019 PD Seg.2 (N=1)

R-020 (N=2)

Stable disease

Resistance

Patient clinical outcome at time of/following biopsy

0.0

0.5

1.0

TK

1 m

RN

A le

vels

R-019 PD Seg.5

R-017

R-018

R-019 BL Seg.2

R-019 PD Seg.2

R-020

Fig. 4. PDOs recapitulate intra- and interpatient heterogeneityin response to TAS-102. (A) PDOs were established from a patient(R-019) with mixed response to TAS-102. Whereas the segment2 metastasis rapidly progressed, the segment 5 metastasis remainedstable upon TAS-102 treatment (white arrowheads in the CT scanindicate metastases; bars represent pre- and posttreatmentmeasurements of the indicated metastases). (B) Ex vivo

concentration-response curves in BL and PD multiregion PDOs frompatient R-019 (with mixed response to TAS-102). (C) TK1 IHC expressionin TAS-102–refractory (segment 2) and –sensitive (segment 5) PDOs.(D) Cell viability (left) and TK1 mRNA expression (right) in PDOsfrom TAS-102–responsive and –refractory patients. In (B) and (D),N indicates the number of independent experiments, and viabilityvalues are expressed as means ± SEM.

RESEARCH | REPORT

Corrected 20 February 2018. See full text. on July 30, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 6: ORGANOIDS Patient-derived organoids model treatment response … · ORGANOIDS Patient-derived organoids model treatment response of metastatic gastrointestinal cancers Georgios Vlachogiannis,

mutations for anti-EGFR therapy, there are novalidated clinical biomarkers for patient selectionin this setting. We initially tested the predictivevalue of PDOs inmCRCby comparing response toanti-EGFR treatment (cetuximab) in five PDOsand their respective patients (Fig. 2E). Two PDOsestablished from BL biopsies before anti-EGFRtreatment in the PROSPECT-C trial showed noresponse to cetuximab, in keeping with the pri-mary resistance observed in these two patientsin the clinic. Unsurprisingly (17), both PDOs andtheir respective patient biopsies harbored ei-ther KRAS G12D (subclonal) (G, glycine; D,aspartic acid) orBRAFV600E (clonal)mutations(Fig. 2F). The third cetuximab-resistant PDO(C-002) was established from the progressionbiopsy of a patient who initially responded tocetuximab, and it harbored an EGFR amplifica-tion (Fig. 1D and fig. S5), no RAS pathwaymuta-tional aberrations (Fig. 2F), andhigh amphiregulinmRNA levels. Despite these molecular markersbeing suggestive of responsiveness to cetuximab,the C-002 PDO showedno response (and, in fact,paradoxically showed enhanced proliferation)upon cetuximab treatment, in linewith the respec-tive patient’s clinical outcome, thus highlightingthe potential of PDOs to predict clinical outcomesbetter than molecular pathology alone. AnotherKRASwild-type PDOderived froma slow-growingprogressing metastasis in a patient with other-wise stable disease (C-001) (fig. S11C) showed amarginal response to cetuximab. Last, theKRASwild-type PDO established from a BL biopsy of apatient enrolled in the PROSPECT-R trial (R-007)(fig. S11C) showed response to cetuximab at con-centrations higher than 5 mg/ml; this, however,could not be compared with clinical response,because the patient did not receive anti-EGFRmonoclonal antibodies.Next we tested the ability of PDOs to reca-

pitulate response to regorafenib, amultiple tyro-sine kinase inhibitor blocking oncogenic andangiogenic signaling pathways. No response toregorafenibwas observed in our 3Dex vivo screen-ing assays (fig. S9A), an observation in keepingwith our recently reported clinical results fromthe PROSPECT-R trial (18), suggesting that re-sponse to regorafenib ismainly driven by its anti-angiogenic effect (Fig. 3A).Tomatch responses to regorafenib in the clinic

and in aligned PDOs, we established an ortho-topic human tumor xenograft model by implant-ing luciferase-expressing (Luc+) PDOs in the liverof NSG mice (PDO-xenografts) (fig. S12A). Weinitially compared response to regorafenib inPDO-xenografts from a patient with primary re-sistance (R-009; n = 11) and from a patient whoachieved a durable (10-month) response (R-005;n = 6) to regorafenib (fig. S12, B and C). In keep-ing with the clinical responses (Fig. 3A), PDO-xenografts from the regorafenib-sensitive patientdisplayed a significant (P = 0.03) reduction intheir microvasculature in response to regorafe-nib, as revealed by CD31 immunostaining; incontrast, no significant changes were observed inPDO-xenografts from the regorafenib-resistantpatient (Fig. 3B). To mimic our clinical obser-

vations, we performed functional susceptibilitycontrast magnetic resonance imaging (MRI) ofPDO-xenografts of the responding patient (R-005;n = 10) before and after treatment (fig. S12D).In line with dynamic contrast-enhanced MRI(DCE-MRI) results in patients (Fig. 3A), suscep-tibility contrastMRI revealed a significant reduc-tion in tumor fractional blood volume (fBV) inregorafenib-treated mice (Fig. 3C). These changeswere associatedwith a reduction in CD31 stainingand increased necrosis (Fig. 3C). Notably, acrossall animals, a robust correlation was observed be-tween the fBV values obtained from susceptibilitycontrast MRI and the microvasculature assess-ment (CD31) of the same samples (R2 = 0.64, P =0.006) (Fig. 3C). Consistent with our clinical data,changes in microvasculature indicative of re-sponse appeared to be independent of changesin tumor volume (fig. S12E) (18). Three differ-ent histopathological growth patterns (HGPs)—desmoplasticHGP,pushingHGP, and replacementHGP—havebeen associatedwithdifferent degreesof response to anti-angiogenic drugs, with the re-placementHGP being frequently associatedwithvessel co-option and primary resistance (19). Inour experiments, a predominance of replacementHGP, and thus vessel co-option, was observedin PDO-xenografts from the resistant patient,whereas tumors established from the PDOs ofthe sensitive patient showed a prevalence of des-moplastic and pushingHGPs (fig. S12F), suggest-ing that vessel co-optionmight be themechanismunderpinning primary resistance to regorafenib.When the responder to regorafenib (R-005) pro-gressed and received subsequent treatment, hewas enrolled in a phase 1 trial of the ATR inhib-itor VX-970. No response was observed in thispatient with VX-970monotherapy, and this wasin keeping with the lack of response to ATM/ATR inhibitors observed in his PDOs in the drugscreening reported in fig. S9A.To test the PDOs’ ability to capture tumor

evolution and acquired resistance to treatment,we generated xenografts using PDOs from thesame liver metastasis before (BL) and after (PD)treatment inmCRC patient R-011 that exhibitedinitial response to regorafenib and subsequentlyprogressed (fig. S13A). Mice were randomized totreatment and control arms, and, after treatment,each arm was further randomized for survival orfunctional analysis (Fig. 3D). In line with clinicalfindings (Fig. 3E) (18), CD31 immunostainingrevealed a ~60% reduction in microvasculaturein response to regorafenib in BL PDO-xenografts(P = 0.001), whereas no significant change wasobserved in PD PDO-xenografts (Fig. 3F). Moreimportantly, regorafenib treatment offered a se-lective survival benefit in mice carrying BL PDO-xenografts (Fig. 3G and fig. S13B), confirming thepredictive value of PDOs and their ability to re-flect cancer evolution upon treatment.TAS-102, a combination of the nucleoside an-

alog trifluridine and the thymidine phosphorylaseinhibitor tipiracil, is approved for the treatmentof chemorefractory mCRC, but no validated bio-markers are available (20). We compared clinicaland preclinical response to TAS-102 in six organ-

oids from four different patients treated withTAS-102. Initially, we tested response to TAS-102in PDOs from a patient (R-019) who had a mixedresponse, with stability of disease in one of theliver metastases (segment 5) and rapid progres-sion in another (segment 2) (Fig. 4A). Ex vivoconcentration-response data showed about aneightfold difference in GI50 between PDOs derivedfrom the TAS-102–sensitive metastasis and thosederived from pre- and posttreatment biopsy ofthe rapidly progressing metastasis (Fig. 4B),highlighting the ability of PDOs to recapitulateintrapatient heterogeneity. TK1 has been pro-posed as a potential biomarker of response toTAS-102 (21); TK1 protein expression was indeedhigher in PDOs from the responding metastasisthan in those from the nonresponding site(Fig. 4C). When we extended the TAS-102 sensitiv-ity analysis to PDOs from three other patients, weconfirmed that PDOs from patients who achieveddisease control were sensitive to low micromolarconcentrations of TAS-102, whereas no significanteffect on cell viability was observed in PDOs fromresistant (primary or acquired) patients (Fig. 4D,left); consistent with previous data, TK1 mRNAexpression was higher in PDOs from patients thatachieved stable disease in response to TAS-102(Fig. 4D, right).Overall, for the PDOs that we analyzed, we

found 100% sensitivity, 93% specificity, 88% pos-itive predictive value, and 100% negative pre-dictive value in forecasting response to targetedagents or chemotherapy in patients (Fisher’s ex-act test, P < 0.0001) (table S7). Our data suggestthat PDOs can be exploited for functional ge-nomics to simulate cancer behavior ex vivo andintegrate molecular pathology into the decision-making process of early-phase clinical trials.

REFERENCES AND NOTES

1. A. Zehir et al., Nat. Med. 23, 703–713 (2017).2. E. E. Voest, R. Bernards, Cancer Discov. 6, 130–132

(2016).3. A. T. Byrne et al., Nat. Rev. Cancer 17, 254–268 (2017).4. J. G. Clohessy, P. P. Pandolfi, Nat. Rev. Clin. Oncol. 12, 491–498

(2015).5. A. L. Bredenoord, H. Clevers, J. A. Knoblich, Science 355,

eaaf9414 (2017).6. M. van de Wetering et al., Cell 161, 933–945 (2015).7. F. Weeber et al., Proc. Natl. Acad. Sci. U.S.A. 112, 13308–13311

(2015).8. M. Fujii et al., Cell Stem Cell 18, 827–838 (2016).9. C. Pauli et al., Cancer Discov. 7, 462–477 (2017).10. J. F. Dekkers et al., Sci. Transl. Med. 8, 344ra84 (2016).11. Cancer Genome Atlas Network, Nature 487, 330–337 (2012).12. Cancer Genome Atlas Research Network, Nature 513, 202–209

(2014).13. D. M. Hyman et al., N. Engl. J. Med. 373, 726–736 (2015).14. C. J. Sherr, D. Beach, G. I. Shapiro, Cancer Discov. 6, 353–367

(2016).15. E. C. Smyth et al., Ann. Oncol. 27, v38–v49 (2016).16. L. M. Zasadil et al., Sci. Transl. Med. 6, 229ra43 (2014).17. C. S. Verissimo et al., eLife 5, e18489 (2016).18. K. Khan et al., Gut 10.1136/gutjnl-2017-314178 (2017).19. S. Frentzas et al., Nat. Med. 22, 1294–1302 (2016).20. R. J. Mayer et al., N. Engl. J. Med. 372, 1909–1919 (2015).21. Y. Kuboki et al., Lancet Oncol. 18, 1172–1181 (2017).

ACKNOWLEDGMENTS

This work was supported by Cancer Research UK (grant numberCEA A18052), the National Institute for Health Research (NIHR)Biomedical Research Centre (BRC) at The Royal Marsden NHSFoundation Trust and The Institute of Cancer Research

Vlachogiannis et al., Science 359, 920–926 (2018) 23 February 2018 6 of 7

RESEARCH | REPORT

Corrected 20 February 2018. See full text. on July 30, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 7: ORGANOIDS Patient-derived organoids model treatment response … · ORGANOIDS Patient-derived organoids model treatment response of metastatic gastrointestinal cancers Georgios Vlachogiannis,

(grant numbers A62, A100, A101, and A159), and the EuropeanUnion Seventh Framework Programme (grant number CIG 334261)to N.V.; by Cancer Research UK (C52506/A22909) to A.So.; by aWellcome Trust grant (105104/Z/14/Z) to the Centre for Evolutionand Cancer; by Cancer Research UK Cancer Imaging Centre funding(C1060/A16464) to The Institute of Cancer Research; by CancerResearch UK Programme Grants C309/A11566 and C2739/A22897 to the Cancer Therapeutics Unit of The Institute of CancerResearch; and by a Bayer Oncology Group Research Grant toD.Cu. A.So. is supported by The Chris Rokos Fellowship in Evolutionand Cancer. I.C. has had advisory roles with Merck Serono, Roche,

Sanofi Oncology, Bristol Myers Squibb, Eli Lilly, Novartis, and GileadScience; has received research funding from Merck-Serono, Novartis,Roche, and Sanofi Oncology; and has received honoraria fromRoche, Sanofi-Oncology, Eli Lilly, and Taiho. D.Cu. has receivedresearch funding from Roche, Amgen, Celgene, Sanofi, MerckSerono, Novartis, AstraZeneca, Bayer, Merrimack, and MedImmune.All other authors declare no conflicts of interest. The data presentedin this paper are tabulated in the main text and supplementarymaterials. Sharing of materials is subject to a material transferagreement with The Institute of Cancer Research, London (pleasedirect requests to N.V.).

SUPPLEMENTARY MATERIALS

www.sciencemag.org/content/359/6378/920/suppl/DC1Materials and MethodsSupplementary TextFigs. S1 to S13Tables S1 to S8References (22–34)

4 July 2017; resubmitted 26 October 2017Accepted 11 January 201810.1126/science.aao2774

Vlachogiannis et al., Science 359, 920–926 (2018) 23 February 2018 7 of 7

RESEARCH | REPORT

Corrected 20 February 2018. See full text. on July 30, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 8: ORGANOIDS Patient-derived organoids model treatment response … · ORGANOIDS Patient-derived organoids model treatment response of metastatic gastrointestinal cancers Georgios Vlachogiannis,

Patient-derived organoids model treatment response of metastatic gastrointestinal cancers

Nicola ValeriSansom, Suzanne Eccles, Naureen Starling, Chiara Braconi, Andrea Sottoriva, Simon P. Robinson, David Cunningham and Clarke, Jens C. Hahne, Mitchell Dowsett, Johann de Bono, Paul Workman, Anguraj Sadanandam, Matteo Fassan, Owen J.Fotiadis, Maria Bali, Mahnaz Darvish-Damavandi, Hazel Lote, Zakaria Eltahir, Elizabeth C. Smyth, Ruwaida Begum, Paul A. Hulkki-Wilson, Chanthirika Ragulan, Inmaculada Spiteri, Sing Yu Moorcraft, Ian Chau, Sheela Rao, David Watkins, NicosLampis, Katherine Eason, Ian Huntingford, Rosemary Burke, Mihaela Rata, Dow-Mu Koh, Nina Tunariu, David Collins, Sanna Georgios Vlachogiannis, Somaieh Hedayat, Alexandra Vatsiou, Yann Jamin, Javier Fernández-Mateos, Khurum Khan, Andrea

DOI: 10.1126/science.aao2774 (6378), 920-926.359Science 

, this issue p. 920Sciencedevelopment.had similar molecular profiles to those of the patient tumor, reinforcing their value as a platform for drug screening andcompare organoid drug responses with how the patient actually responded in the clinic. Encouragingly, the organoids gastrointestinal cancer who had previously been enrolled in phase I or II clinical trials. This allowed the authors toto predict drug responses in the clinic. They generated a live organoid biobank from patients with metastatic

asked whether such ''tumor-in-a-dish'' approaches can be usedet al.tumors and studied in the laboratory. Vlachogiannis Cancer organoids are miniature, three-dimensional cell culture models that can be made from primary patient

Cancer organoids to model therapy response

ARTICLE TOOLS http://science.sciencemag.org/content/359/6378/920

MATERIALSSUPPLEMENTARY http://science.sciencemag.org/content/suppl/2018/02/21/359.6378.920.DC1

CONTENTRELATED

http://stm.sciencemag.org/content/scitransmed/6/250/250ra115.fullhttp://stm.sciencemag.org/content/scitransmed/8/344/344ra84.full

REFERENCES

http://science.sciencemag.org/content/359/6378/920#BIBLThis article cites 34 articles, 9 of which you can access for free

PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

Terms of ServiceUse of this article is subject to the

is a registered trademark of AAAS.ScienceScience, 1200 New York Avenue NW, Washington, DC 20005. The title (print ISSN 0036-8075; online ISSN 1095-9203) is published by the American Association for the Advancement ofScience

Science. No claim to original U.S. Government WorksCopyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of

on July 30, 2020

http://science.sciencemag.org/

Dow

nloaded from