divergent modes of clonal spread and intraperitoneal mixing in … · 2019-08-06 · high-grade...

11
758 VOLUME 48 | NUMBER 7 | JULY 2016 NATURE GENETICS High-grade serous ovarian cancer (HGSOvCa) is the most lethal histotype of ovarian cancer, where up to 80% of patients suffer relapse after initial response to treatment, with many succumbing to treatment-resistant disease. HGSOvCa is characterized by widespread and early intraperitoneal spread to organs, such as the omentum and contralateral ovary. The genomes of HGSOvCa exhibit perva- sive instability due to TP53 loss in nearly all cases 1 , accompanied by deficiencies in homologous recombination and other DNA repair pathways in the majority of cases 2 . Thus, combined dysregulation of apoptosis and DNA repair processes provides a mechanism for generating a substrate of genomically diverse clones 3–5 from a monoclonal etiology 6,7 . The peritoneal cavity lacks distinct physical barriers to ovarian cancer metastasis, resulting in early and widespread disease at dis- tal peritoneal sites. The majority of disease spread in HGSOvCa is thought to be driven by cell mixing and invasion within a physical anatomical cavity 8 . Such ‘local’ spreading properties contrast with those for other solid malignancies, where hematological or lymphatic processes govern metastasis 9–12 . It is unknown what proportion of local spread in the peritoneum arises from monoclonal seeding and expansion or from extensive cellular mixing of diverse polyclonal populations. As such, the interaction of differential migration and invasion capacities for divergent clones with the intraperitoneal fit- ness landscape is poorly understood. One model for local spread would suggest that microenvironmental niches induced by peritoneal organs introduce variable selective topologies for genomically diverse clones. Alternatively, if the composition of the peritoneal cavity were uniformly selective, this could yield a pattern consistent with multiple migrations and extensive clonal mixing. Studying the properties of intraperitoneal spread for genomically defined clones in HGSOvCa requires a multiregion whole-genome sequencing approach. Whole-genome sequencing offers a robust experimental design to study evolution in cancer, primarily because orders of magnitude more mutations are accessible to phylogenetic modeling, with ascertainment of structural copy number alterations and rearrangement breakpoints as clonal marks. Single-nucleus sequencing in turn complements bulk whole-genome sequencing approaches for direct measurement of extant clonal genotypes in a sample of interest, facilitating tracking of clones in temporal or spatial dimensions 13–15 . Divergent modes of clonal spread and intraperitoneal mixing in high-grade serous ovarian cancer Andrew McPherson 1–4,13 , Andrew Roth 1,3,13 , Emma Laks 1 , Tehmina Masud 1,4 , Ali Bashashati 1 , Allen W Zhang 1,3,5 , Gavin Ha 1,3,6,7 , Justina Biele 1 , Damian Yap 1 , Adrian Wan 1 , Leah M Prentice 8 , Jaswinder Khattra 1 , Maia A Smith 1,3 , Cydney B Nielsen 4 , Sarah C Mullaly 1 , Steve Kalloger 1 , Anthony Karnezis 8 , Karey Shumansky 1 , Celia Siu 1 , Jamie Rosner 1 , Hector Li Chan 8 , Julie Ho 8 , Nataliya Melnyk 8 , Janine Senz 8 , Winnie Yang 8 , Richard Moore 9 , Andrew J Mungall 9 , Marco A Marra 9 , Alexandre Bouchard-Côté 10 , C Blake Gilks 11 , David G Huntsman 1,4,8 , Jessica N McAlpine 12 , Samuel Aparicio 1,4 & Sohrab P Shah 1,4,9 We performed phylogenetic analysis of high-grade serous ovarian cancers (68 samples from seven patients), identifying constituent clones and quantifying their relative abundances at multiple intraperitoneal sites.Through whole-genome and single- nucleus sequencing, we identified evolutionary features including mutation loss, convergence of the structural genome and temporal activation of mutational processes that patterned clonal progression. We then determined the precise clonal mixtures comprising each tumor sample. The majority of sites were clonally pure or composed of clones from a single phylogenetic clade. However, each patient contained at least one site composed of polyphyletic clones. Five patients exhibited monoclonal and unidirectional seeding from the ovary to intraperitoneal sites, and two patients demonstrated polyclonal spread and reseeding. Our findings indicate that at least two distinct modes of intraperitoneal spread operate in clonal dissemination and highlight the distribution of migratory potential over clonal populations comprising high-grade serous ovarian cancers. 1 Department of Molecular Oncology, BC Cancer Agency, Vancouver, British Columbia, Canada. 2 School of Computing Science, Simon Fraser University, Burnaby, British Columbia, Canada. 3 Graduate Bioinformatics Training Program, University of British Columbia, Vancouver, British Columbia, Canada. 4 Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada. 5 Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Vancouver, British Columbia, Canada. 6 Dana-Farber Cancer Institute, Boston, Massachusetts, USA. 7 Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA. 8 Centre for Translational and Applied Genomics, BC Cancer Agency, Vancouver, British Columbia, Canada. 9 Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada. 10 Department of Statistics, University of British Columbia, Vancouver, British Columbia, Canada. 11 Department of Pathology, Vancouver General Hospital, Vancouver, British Columbia, Canada. 12 Department of Gynecology and Obstetrics, University of British Columbia, Vancouver, British Columbia, Canada. 13 These authors contributed equally to this work. Correspondence should be addressed to S.P.S. ([email protected]) or S.A. ([email protected]). Received 15 October 2015; accepted 25 April 2016; published online 16 May 2016; doi:10.1038/ng.3573 ARTICLES npg © 2016 Nature America, Inc. All rights reserved. npg © 2016 Nature America, Inc. All rights reserved.

Upload: others

Post on 06-Jul-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Divergent modes of clonal spread and intraperitoneal mixing in … · 2019-08-06 · High-grade serous ovarian cancer (HGSOvCa) is the most lethal . histotype of ovarian cancer, where

758 VOLUME 48 | NUMBER 7 | JULY 2016 Nature GeNetics

High-grade serous ovarian cancer (HGSOvCa) is the most lethal histotype of ovarian cancer, where up to 80% of patients suffer relapse after initial response to treatment, with many succumbing to treatment-resistant disease. HGSOvCa is characterized by widespread and early intraperitoneal spread to organs, such as the omentum and contralateral ovary. The genomes of HGSOvCa exhibit perva-sive instability due to TP53 loss in nearly all cases1, accompanied by deficiencies in homologous recombination and other DNA repair pathways in the majority of cases2. Thus, combined dysregulation of apoptosis and DNA repair processes provides a mechanism for generating a substrate of genomically diverse clones3–5 from a monoclonal etiology6,7.

The peritoneal cavity lacks distinct physical barriers to ovarian cancer metastasis, resulting in early and widespread disease at dis-tal peritoneal sites. The majority of disease spread in HGSOvCa is thought to be driven by cell mixing and invasion within a physical anatomical cavity8. Such ‘local’ spreading properties contrast with those for other solid malignancies, where hematological or lymphatic processes govern metastasis9–12. It is unknown what proportion of local spread in the peritoneum arises from monoclonal seeding and

expansion or from extensive cellular mixing of diverse polyclonal populations. As such, the interaction of differential migration and invasion capacities for divergent clones with the intraperitoneal fit-ness landscape is poorly understood. One model for local spread would suggest that microenvironmental niches induced by peritoneal organs introduce variable selective topologies for genomically diverse clones. Alternatively, if the composition of the peritoneal cavity were uniformly selective, this could yield a pattern consistent with multiple migrations and extensive clonal mixing.

Studying the properties of intraperitoneal spread for genomically defined clones in HGSOvCa requires a multiregion whole-genome sequencing approach. Whole-genome sequencing offers a robust experimental design to study evolution in cancer, primarily because orders of magnitude more mutations are accessible to phylogenetic modeling, with ascertainment of structural copy number alterations and rearrangement breakpoints as clonal marks. Single-nucleus sequencing in turn complements bulk whole-genome sequencing approaches for direct measurement of extant clonal genotypes in a sample of interest, facilitating tracking of clones in temporal or spatial dimensions13–15.

Divergent modes of clonal spread and intraperitoneal mixing in high-grade serous ovarian cancerAndrew McPherson1–4,13, Andrew Roth1,3,13, Emma Laks1, Tehmina Masud1,4, Ali Bashashati1, Allen W Zhang1,3,5, Gavin Ha1,3,6,7, Justina Biele1, Damian Yap1, Adrian Wan1, Leah M Prentice8, Jaswinder Khattra1, Maia A Smith1,3, Cydney B Nielsen4, Sarah C Mullaly1, Steve Kalloger1, Anthony Karnezis8, Karey Shumansky1, Celia Siu1, Jamie Rosner1, Hector Li Chan8, Julie Ho8, Nataliya Melnyk8, Janine Senz8, Winnie Yang8, Richard Moore9, Andrew J Mungall9, Marco A Marra9, Alexandre Bouchard-Côté10, C Blake Gilks11, David G Huntsman1,4,8, Jessica N McAlpine12, Samuel Aparicio1,4 & Sohrab P Shah1,4,9

Weperformedphylogeneticanalysisofhigh-gradeserousovariancancers(68samplesfromsevenpatients),identifyingconstituentclonesandquantifyingtheirrelativeabundancesatmultipleintraperitonealsites.Throughwhole-genomeandsingle-nucleussequencing,weidentifiedevolutionaryfeaturesincludingmutationloss,convergenceofthestructuralgenomeandtemporalactivationofmutationalprocessesthatpatternedclonalprogression.Wethendeterminedthepreciseclonalmixturescomprisingeachtumorsample.Themajorityofsiteswereclonallypureorcomposedofclonesfromasinglephylogeneticclade.However,eachpatientcontainedatleastonesitecomposedofpolyphyleticclones.Fivepatientsexhibitedmonoclonalandunidirectionalseedingfromtheovarytointraperitonealsites,andtwopatientsdemonstratedpolyclonalspreadandreseeding.Ourfindingsindicatethatatleasttwodistinctmodesofintraperitonealspreadoperateinclonaldisseminationandhighlightthedistributionofmigratorypotentialoverclonalpopulationscomprisinghigh-gradeserousovariancancers.

1Department of Molecular Oncology, BC Cancer Agency, Vancouver, British Columbia, Canada. 2School of Computing Science, Simon Fraser University, Burnaby, British Columbia, Canada. 3Graduate Bioinformatics Training Program, University of British Columbia, Vancouver, British Columbia, Canada. 4Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada. 5Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Vancouver, British Columbia, Canada. 6Dana-Farber Cancer Institute, Boston, Massachusetts, USA. 7Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA. 8Centre for Translational and Applied Genomics, BC Cancer Agency, Vancouver, British Columbia, Canada. 9Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada. 10Department of Statistics, University of British Columbia, Vancouver, British Columbia, Canada. 11Department of Pathology, Vancouver General Hospital, Vancouver, British Columbia, Canada. 12Department of Gynecology and Obstetrics, University of British Columbia, Vancouver, British Columbia, Canada. 13These authors contributed equally to this work. Correspondence should be addressed to S.P.S. ([email protected]) or S.A. ([email protected]).

Received 15 October 2015; accepted 25 April 2016; published online 16 May 2016; doi:10.1038/ng.3573

A rt i c l e s

npg

© 2

016

Nat

ure

Am

eric

a, In

c. A

ll rig

hts

rese

rved

.np

2016

Nat

ure

Am

eric

a, In

c. A

ll rig

hts

rese

rved

.

Page 2: Divergent modes of clonal spread and intraperitoneal mixing in … · 2019-08-06 · High-grade serous ovarian cancer (HGSOvCa) is the most lethal . histotype of ovarian cancer, where

Nature GeNetics VOLUME 48 | NUMBER 7 | JULY 2016 759

We set out to resolve the modes of clonal spread to gain an improved understanding of pretreatment HGSOvCa disease progression. Clonal population profiling of spatially distinct intraperitoneal samples was performed, combining the experimental approaches of whole-genome and deep, targeted sequencing in all samples and direct sequencing of >1,500 individual nuclei across multiple samples from three patients. We established robust clonal genotypes and their phylogenetic rela-tionships, gaining new insights into the mechanisms of tumor evolu-tion, including mutation loss over evolutionary history and convergent evolution of the structural genome. We measured the abundance of clones across all sites for each patient, identifying properties of intra-peritoneal spread across two dimensions: the clonal composition of individual samples and the migration capacity of individual clones. A minority of patients exhibited a high degree of polyphyletic clonal mixing and reseeding of clones at distal foci, whereas in the majority of cases clonal diversity emerged at a primary site followed by unidi-rectional, monoclonal seeding to distal intraperitoneal sites.

RESULTSWe collected 68 tumor samples (Supplementary Table 1) from seven patients (between 5 and 13 samples per patient) (Table 1), including samples from the ovary, omentum, fallopian tube, peritoneal sites, and other distant metastatic sites (Fig. 1a). For one patient (patient 7), samples were obtained at initial diagnosis, at first relapse after 13 months, and at second relapse after an additional 19 months. For the remaining patients, all samples were obtained during initial debulking surgery. Additional detail on sample acquisition, including the limi-tations of surgical sampling, is provided in the Supplementary Note.

SamplephylogeniesandclonaldivergencebymutationlossWhole-genome sequencing (~30× coverage) was applied to 31 cryop-reserved tissues that yielded high-quality DNA extractions (discovery samples), in addition to matched normal blood from each patient (Supplementary Table 2). The remaining 37 formalin-fixed, paraffin- embedded specimens (archival samples) were used in targeted sequencing as described below. Analysis of the discovery samples resulted in a catalog of somatic genomic aberrations, including single- nucleotide variants (SNVs), allele-specific absolute copy number changes, and rearrangement breakpoints, for each patient (see the Supplementary Note for details on the analytical approaches). Between 3,577 and 16,987 SNVs and between 132 and 1,092 rearrangement breakpoints were identified per patient. A high pro-portion of events were specific to a subset of samples, ranging from

0.28 to 0.87 for SNVs and from 0.22 to 0.48 for rearrangement break-points (Supplementary Table 3), providing an initial quantification of the degree of clonal divergence between sites.

We inferred the evolutionary relationships between dominant clones in each sample (sample trees) as defined by both SNVs and rearrange-ment breakpoints. We implemented a probabilistic phylogenetic model based on the stochastic Dollo process16,17 (Supplementary Note) accounting for genomic instability and associated deletion of chromo-some segments containing point mutations. We computed maximum-likelihood sample tree topologies and rate of mutation loss parameters for SNVs (Fig. 1a and Supplementary Figs. 1–3) and rearrangement breakpoints (Supplementary Figs. 4–6). The SNV- and breakpoint-derived trees were consistent for all patients except patient 3, providing corroborating topologies from completely orthogonal features of the genome. The number of rearrangement breakpoints and the number of SNVs originating on each branch in each tree were correlated (Spearman ρ = 0.88, P = 2.2 × 10−16, patient 3 excluded; Supplementary Fig. 1b and Supplementary Table 4). However, numbers of mutation losses were only weakly correlated (Spearman ρ = 0.37, P = 0.01, patient 3 excluded; Supplementary Fig. 1c and Supplementary Table 4). In all patients, sample tree topologies indicated the presence of multiple ancestrally related clones with substantial divergences between samples.

The inferred probability of losing a mutation on a branch ranged from 0.00094 (patient 10) to 0.012 (patient 9) for SNVs and from 0 (patients 2, 3, and 10) to 0.044 (patient 9) for rearrangement breakpoints. The pro-portion of SNV losses attributed to copy number changes ranged from 0.28 to 0.89 (Fig. 1b, Supplementary Figs. 7–13, and Supplementary Table 3), with patients 1 and 7 exhibiting the highest proportions at 0.87 and 0.89, respectively (Supplementary Table 3). The majority of lost SNVs were found in regions of divergent copy number across samples (see the example in Fig. 1c), suggesting that SNVs were likely removed by sample-specific or clade-specific segmental deletions.

TemporalattributesofmutationsignaturesinHGSOvCaWe next sought to use the sample trees to determine the mutational signatures (as defined by Alexandrov et al.18) operating at different points in the evolutionary histories of the tumors. We used a latent Dirichlet allocation (LDA) model19 to learn the relative propor-tion of sample-specific SNVs attributed to each of a curated list of 30 signatures20 (Supplementary Fig. 14, Supplementary Table 5, and Supplementary Note). Hierarchical clustering of the signature pro-portions indicated that sample-specific signatures were similar within each patient, with the brain metastasis in patient 7 being the lone

table 1 Patients and samples included in the study

PatientSamples (index)

Anatomical samples (index)

Samples (archival) Anatomical samples (archival)

Age at diagnosis (years) Stage Recurrence

RFS (months) Statusa

OS (months)

1 6 ROv1–ROv4; Om1; SBwl 6 ApC1; LFTB4; LOvB2; RFTA16; ROvA4; SBwlE4

71.7 IIIC No NED 55

2 4 Om1, Om2; ROv1, ROv2 6 LOvD3; OmA2; OmB1; RFTC10; ROvC2; ROvC4

75.5 IIIC Yes 11 DOD 45

3 4 ROv1, ROv2; Om1; LOv2 7 CDSB1; ClnE1; LFTC1; LOvC5; OmF2; RFTA2; ROvA7

69.2 IIIC Yes 27 AWD 53

4 5 ROv1–ROv4; LPvS 4 LOvB2; LPvSC1; ROvA5; RPvSD1 52.9 IIIB Yes 46 AWD 52

7b 3 LOv1; BrnM; RPvM 10 LOvA10; LOvA4; ROvC4; ROvC5; ROvC6; RUtD1; RUtD2; RUtD3; BrnMA1; BwlA6

47.2 IIIC Yes 9 DOD 52

9 5 ROv1 LOv1, LOv2; Om1, Om2 0 NA 52.5 IIIC Yes 6 DOD 31

10 4 ROv1–ROv4 4 LFTB2; OmC1; ROvA4; ROvA9 73.9 IIA No NED 49

All cases are of high-grade (grade III) serous histology, having undergone secondary pathology review. Stage refers to surgical staging. RFS, recurrence-free survival; OS, overall survival. The abbreviations for anatomical samples are defined in supplementary table 1. aCurrent disease status: AWD, alive with disease; NED, no evidence of disease; DOD, dead of disease. bSamples were acquired temporally: BrnM was acquired 14 months after the LOv, ROv, and RUt samples, and the RPvM and Bwl samples were acquired after an additional 19 months.

A rt i c l e s

npg

© 2

016

Nat

ure

Am

eric

a, In

c. A

ll rig

hts

rese

rved

.np

2016

Nat

ure

Am

eric

a, In

c. A

ll rig

hts

rese

rved

.

Page 3: Divergent modes of clonal spread and intraperitoneal mixing in … · 2019-08-06 · High-grade serous ovarian cancer (HGSOvCa) is the most lethal . histotype of ovarian cancer, where

760 VOLUME 48 | NUMBER 7 | JULY 2016 Nature GeNetics

A rt i c l e s

Ancestraln = 7

Descendantn = 33

00.10.20.30.40.50.60.70.80.9

Signature 3(P = 5.2 × 10–3)

Ancestraln = 7

Descendantn = 33

00.020.040.060.080.100.120.140.16

Signature 11(P = 4.7 × 10–3)

Ancestraln = 7

Descendantn = 33

00.020.040.060.080.10

Signature 15(P = 1.6 × 10–3)

Ancestraln = 7

Descendantn = 33

00.020.040.060.080.100.120.14

Signature 25(P = 2.5 × 10–3)

Ancestraln = 7

Descendantn = 33

0

0.05

0.10

0.15

0.20

Signature 26(P = 8.5 × 10–3)

Patient12347910

d

Ancestraln = 7

Descendantn = 33

0

0.05

0.10

0.15

0.20

Signature 1(P = 1.6 × 10–3)

SN

V p

ropo

rtio

n

Om1/2

LOv1

0123

Om1

0123

ROv1

0123

0 100 200Chromosome 2 position (Mb)

0123

ROv3

0 100 200Chromosome 2 position (Mb)

0123

ROv4

0 100 200Chromosome 2 position (Mb)

0123

SBwl

ROv2c

Alle

le-

spec

i�c

copy

nu

mbe

r

Alle

le-

spec

i�c

copy

nu

mbe

r

b350

SNV lossDeletedUnexplained

DeletedUnexplained

300

250

200

150

100

50

01 2 3 4 7 9 10

Patient

Cou

nta

ROv2ROv1

Adnx

Om1

0

1,000

2,000

3,000

4,000

5,000

6,000

SN

V c

ount

TP53

–25–8 –3

–41STAG2ELF3

ROv1ROv3

ROv2ROv4

LPvS

0

1,000

2,000

3,000

4,000

5,000

6,000

SN

V c

ount

BRCA2

TP53

PHF6–10–5

–19 –2 –1 –24

LOv1

RPvMBrn

M

01,0002,0003,0004,0005,0006,0007,0008,0009,000

SN

V c

ount

TP53

–21 –73USP9X

Patient 3 Patient 7Patient 4

LOv1RPvM

BrnM 321

ROv1, ROv2

Om1

LOv1

ROv1–ROv4

LPvS

ROv3ROv2

ROv1ROv4

SBwlOm

1

0

1,000

2,000

3,000

4,000

5,000

6,000

7,000

SN

V c

ount

TP53

CDK12

–46–26

–102 –11NAV3

–60 –110

–2–4

Om2

Om1

ROv1ROv2

0

1,000

2,000

3,000

4,000

5,000

6,000

7,000S

NV

cou

nt

TP53

TET2

–25

S1

Om2

ROv1Om

1LO

v2LO

v1

0500

1,0001,5002,0002,5003,0003,5004,000

SN

V c

ount TP53

–280–21–10–7

–7S1

ROv1ROv4

ROv3ROv2

02,0004,0006,0008,000

10,00012,00014,00016,000

SN

V c

ount

FAT3

TP53

SETBP1

–53–3

Patient 10

ROv1–ROv4

Patient 1

ROv1–ROv4

SBwl

Om1

Patient 2

ROv1, ROv2

Om1 Om2

Patient 9

ROv1 LOv1, LOv2

Om1 Om2

Figure 1 SNV phylogenies for seven patients with HGSOvCa. (a) Anatomical sites sampled for whole-genome sequencing in seven patients with HGSOvCa. Phylogeny was inferred from SNVs, with previously identified driver genes annotated on the branches. Branch lengths represent the number of SNVs originating on each branch. Branches are annotated with the number of SNVs lost along each branch. (b) Number of SNVs classified as deleted or unexplained by any identifiable deletion event. (c) Allele-specific copy number for the major (red) and minor (blue) alleles (y axis) are shown for chromosome 2 in all samples from patient 1. SNVs classified as deleted or unexplained by identifiable deletions are shown as orange and green ticks above the x axis. Only SNVs classified as lost are shown. A deletion event common to ROv2 and ROv3 is highlighted in yellow. (d) Box plots of the inferred proportion of SNVs attributed to each mutational signature for SNVs acquired on the ancestral versus descendant branches of each sample tree. Only signatures with a significant difference in the inferred proportion between the ancestral and descendant branches are shown (P < 0.01, Mann–Whitney U test). Branches with fewer than 100 mutations were excluded. Superimposed strip plot points representing each data point are colored by patient. Boxes extend from the third (Q3) to the first (Q1) quartile (IQR), with a line at the median; whiskers extend to Q3 + 1.5 IQR and Q1 − 1.5 IQR.

npg

© 2

016

Nat

ure

Am

eric

a, In

c. A

ll rig

hts

rese

rved

.np

2016

Nat

ure

Am

eric

a, In

c. A

ll rig

hts

rese

rved

.

Page 4: Divergent modes of clonal spread and intraperitoneal mixing in … · 2019-08-06 · High-grade serous ovarian cancer (HGSOvCa) is the most lethal . histotype of ovarian cancer, where

Nature GeNetics VOLUME 48 | NUMBER 7 | JULY 2016 761

A rt i c l e s

exception. Mutations were predominately generated by signature 1, the age-associated signature; signature 3, associated with failure of DNA double-strand break repair by homologous recombination; and signatures 5 and 8, common signatures of unknown etiology.

We characterized the relative evolutionary timing of each signature, applying LDA to SNVs partitioned by their phylogenetic origin in the sample tree (Supplementary Fig. 15 and Supplementary Table 6). Hierarchical clustering of branch-specific signature proportions resulted in two major clusters, one of which contained all ancestral branches. Two signatures were highly differentiated in ancestral and descendant SNVs. Signature 3 (homologous recombination deficiency) generated a higher proportion of descendant SNVs, and signature 1 (age associ-ated) generated a higher proportion of ancestral SNVs (Mann–Whitney U test, P < 0.01; Fig. 1d), consistent with the activation of homologous recombination deficiency after the expansion of ancestral clones. Four additional signatures, including signatures 15 and 26 (associated with defective DNA mismatch repair), were significantly associated with SNVs on descendant branches in a minority of patients. Two descendant branches in patients 2 and 9 exhibited a high proportion of signature 1 SNVs and a corresponding low proportion of signature 3 SNVs (Fig. 1d), suggesting that clones in patients 2 and 9 had either intact or only partially deficient homologous recombination.

EvidenceofclonalmixturesinspecificsamplesMultiple lines of evidence nominated samples from patients 2, 3, and 9 as mixtures of divergent tumor lineages. For all three patients, more than 50% of lost mutations could not be explained by sample- specific deletion (Fig. 1b). For patients 2 and 3, densely spaced sets of SNVs predicted to be lost coincided with clone-specific copy number changes (Fig. 2a,b). By contrast, lost SNVs in patient 9 were distributed

uniformly throughout the genome (Fig. 2c and Supplementary Fig. 12), without convincing corroborating copy number changes. Moreover, the top four samples with respect to length of the genome called as having a subclonal copy number state included a sample each from patients 2, 3, and 9 (Fig. 2d and Supplementary Table 7), supportive of multiple divergent clones being present in samples from these patients.

We performed single-nucleus sequencing on patients 2, 3, and 9 (n = 1,680 cells in total; Table 2) with the aim of determining the co-occurrence of breakpoints and PyClone SNV clusters to be used as clonal marks (Online Methods). Single-cell sequencing, in com-bination with the single-cell genotyper (SCG) model21, produced clusters of cells (clones) defined by common patterns of presence and absence for clonal marks (clonal genotypes). The coexistence of multiple clones was evident in patient 2 (right ovary 2 sample (ROv2; genotype clusters b, c, e, and f)) (Fig. 3a), patient 3 (adnexa sam-ple (Adnx; genotype clusters b and e), omentum 1 sample (Om1; genotype clusters a and f), and ROv1 (genotype clusters a and c)) (Fig. 3b), and patient 9 (ROv1 (genotype clusters c and d) and Om2 (genotype clusters c and d)) (Fig. 3c)). Clonal genotypes were used to infer clone phylogenies, ancestral genotypes, and the relative order of mutation acquisition on the basis of a stochastic Dollo-like model

ba

0123

Alle

le-s

peci

ficco

py n

umbe

r

Alle

le-s

peci

ficco

py n

umbe

rA

llele

-spe

cific

copy

num

ber

Alle

le-s

peci

ficco

py n

umbe

r

Om1

0123

Om2

0 100 200

Chromosome 2 position (Mb)

0123

ROv1

0 100 200

Chromosome 2 position (Mb)

0123

ROv2

0123

Adnx

0123

Om1

0 100 200

Chromosome 2 position (Mb)

0123

ROv1

0 100 200

Chromosome 2 position (Mb)

0123

ROv2

dc

Alle

le-s

peci

ficco

py n

umbe

rA

llele

-spe

cific

copy

num

ber

0123

LOv1

0123

LOv2

0 50 100 150 200

Chromosome 3 position (Mb)

0123

Om2

0 50 100 150 200

Chromosome 3 position (Mb)

0123

ROv1

1 2 3 4 7 9 10

RO

v2O

m1

RO

v3R

Ov4

SB

wl

RO

v1

RO

v2O

m2

Om

1R

Ov1

Om

1R

Ov1

Adn

xR

Ov2

RO

v4LP

vSR

Ov2

RO

v1R

Ov3

Brn

MR

PvM

LOv1

LOv2

RO

v1O

m2

LOv1

Om

1

RO

v4R

Ov2

RO

v1R

Ov3

Patient sample

0

200

400

600

800

1,000

1,200

1,400

Leng

th o

f sub

clon

al c

opy

num

ber

regi

on (

Mb)

Patient12347910

DeletedUnexplainedDeletedUnexplained

Figure 2 Evidence of samples composed of divergent clonal mixtures. (a–c) Predicted copy number for major (red) and minor (blue) alleles for each sample of chromosome 2 from patient 2 (a), chromosome 2 from patient 3 (b), and chromosome 3 from patient 9 (c). Ticks above the x axis show the positions of SNVs present in each sample. SNV ticks are colored orange if they can be explained by a clonally dominant deletion; otherwise, they are colored green. In a, a subclonally deleted chromosomal segment is highlighted in yellow. In c, SNVs are evenly distributed across the chromosome (in addition to other chromosomes) and are not associated with identifiable deletion events. (d) Length of the reference genome predicted as having a subclonal copy number state for each sample in each patient.

table 2 single-nucleus sequencing statistics

Patient Samplesa Cellsb Breakpointsc SNVsdMedian coverage Mean coverage

2 4 588 5 43 391 1,273

3 4 672 12 84 52 355

9 5 420 5 43 375 2,775

aNumber of samples sequenced. bNumber of nuclei sequenced. cNumber of breakpoint targets. dNumber of SNV targets.

npg

© 2

016

Nat

ure

Am

eric

a, In

c. A

ll rig

hts

rese

rved

.np

2016

Nat

ure

Am

eric

a, In

c. A

ll rig

hts

rese

rved

.

Page 5: Divergent modes of clonal spread and intraperitoneal mixing in … · 2019-08-06 · High-grade serous ovarian cancer (HGSOvCa) is the most lethal . histotype of ovarian cancer, where

762 VOLUME 48 | NUMBER 7 | JULY 2016 Nature GeNetics

A rt i c l e s

(Supplementary Fig. 16, Supplementary Table 8, and Supplementary Note).

IntraperitonealsamplediversityandmigrationcapacityIn total, we inferred 44 clonal genotypes across the seven patients, ranging in number from 3 (patient 9) to 9 (patient 1) per patient (Supplementary Fig. 16 and Supplementary Table 8), resolved by either single-cell sequencing (patients 2, 3, and 9) or inferred from deep sequencing (patients 1, 4, 7, and 10; Online Methods). We applied targeted deep sequencing of mutations comprising the clonal genotypes on both discovery and archival samples to determine their prevalence conditioned on the PyClone clustering of the discovery samples (Supplementary Note). For each patient, we elucidated the pattern of spread for each clone and a robust estimate of the clonal composition of each sample (Supplementary Table 9).

We categorized samples as clonally pure or polyclonal; polyclonal samples were further classified into samples where multiple clones were from within the same phylogenetic clade (monophyletic) or were from different clades (polyphyletic) (Fig. 4 and Supplementary Fig. 17). The majority of samples were monophyletic or clonally pure; how-ever, each patient exhibited at least one polyphyletic sample (Fig. 5a), including LOvB2 in patient 1, ROv2 in patient 2, ROv2 and ROv4 in patient 4, ROvA7 in patient 3, and ROvA9 in patient 10. Polyphyletic samples are indicative of an ancestral population having undergone branched clonal evolution or of the site having been seeded either serially or simultaneously (polyclonal migration) by multiple clones. For patients with a single polyphyletic sample (patients 2, 7, and 9), the ancestral population model is more parsimonious with respect to the number of migration events needed to explain the data.

For four of the six patients with primary ovary and non-ovarian intraperitoneal discovery samples (patients 1, 2, 4, and 9), one of the two major clades of the clone phylogeny dominated the putative primary ovary site, with the divergent clade dominating the perito-neal and contralateral ovary sites (Fig. 5b). For patients 1 and 2, the primary ovary samples were pure for the primary clade, whereas, for patients 4 and 9, the primary ovary samples contained minor populations of the peritoneal clones. For patients 4 and 9, analysis of competing migration scenarios (Supplementary Note) was most parsimonious with migrating clones originating within and then disseminating away from the primary ovary site (Supplementary Figs. 18–24). Thus, for four of the six patients, we did not observe clonal reseeding from peritoneal to primary sites. For patient 7, the

minimum-event migration scenario nominated RUtD3 as the pre-treatment site in which the observed tumor clones originated, with subsequent metastasis to the brain and other peritoneal sites.

To characterize the distribution of clonal composition over all samples, we calculated two parameters for each sample: the sam-ple mixture entropy (the relative balance of the clonal proportions of the clones in a sample) and the intraperitoneal and between-clade divergence of the clones comprising the sample (Fig. 5a and Supplementary Note). The distribution across samples was patterned by three broad categories: samples that were clonally pure (Fig. 5a, circles), samples with multiple clones present but with relatively low entropy, consistent with monophyletic topologies (Fig. 5a, trian-gles), and samples with balanced mixtures from polyphyletic clones (Fig. 5a, stars). The spectrum of samples over the space of this distri-bution can be interpreted as follows: samples in the upper right region are clonally diverse and composed of clones from distinct clades in the clone phylogeny, whereas samples in the lower left region are clonally pure or are formed from closely related clones. Notably, every patient exhibited at least one sample with high entropy and high clone divergence (Fig. 5a). However, the shape of the sample distribution indicates that intraperitoneal spread in the cohort is not characterized by repeat reseeding of multiple clones. Most of the density in Figure 5a is found in the lower left region, where clones are characterized by either clonally pure or monophyletic topologies. Thus, only a minority of specific sites (often primary ovary sites) are permissive to coexisting clones (whether from multiple migrations or branched evolution), which seed the remainder of the intraperitoneal sites composed of clonally pure or chain topologies. Exceptions to this trend were found in patients 3 and 10, for whom samples comprised multiple polyphyletic samples in the upper right region of the distri-bution (Fig. 5a, purple and brown stars). As such, for patients 3 and 10, the existence of multiple samples composed of divergent lineages is concordant with polyclonal reseeding or polyclonal migration, in contrast to the scenarios in patients 1, 2, 4, and 9. Furthermore, these two patients ranked highest in the number of predicted migration events per site (Fig. 5d).

We next calculated the spreading and relative within-sample preva-lence characteristics of individual clones. We plotted the number of

Mutation

Cell

HetSNPCluster 5Cluster 8

Cluster 11Cluster 1Cluster 9

Cluster 4Breakpoints 1Absent

Site

Om1Om2

ROv1ROv2

Normal

a b c

a

bc

d

e

f

Patient 2

Mutation

Cell

HetSNPCluster 7Cluster 10Cluster 11Cluster 15

Cluster 16Cluster 17Cluster 8Cluster 9

Cluster 6Cluster 1Cluster 4Absent

Site

AdnxOm1

ROv1ROv2

Normal

a

b

c

d

e

f

Patient 3

Mutation

Cell

HetSNPCluster 1Cluster 4

Cluster 5ERBB2Cluster 6

Breakpoints 1Absent

Site

LOv1LOv2Om1

Om2ROv1

Normal

a

b

c

d

ef

Patient 9Figure 3 Single-nucleus sequencing of patients with samples containing mixtures of divergent lineages. (a–c) Nucleus × target matrices for patients 2 (a), 3 (b), and 9 (c) with nuclei clustered by genotype using the SCG model. Matrix entries are colored by variant cluster (PyClone cluster, breakpoint, or heterozygous SNP). Nuclei are annotated with the sample from which they originated (colored “site” indicators), in addition to their genotype clusters (labels to the left of the site indicators). HetSNPs represent loci predicted in the whole-genome sequencing data to be heterozygous in the normal sample that were predicted to be homozygous reference in tumors; these SNPs act as indicators of normal cells. For patient 3, HetSNPs also include SNPs predicted to be encompassed by subclonal LOH events. Cluster labels correspond to those in supplementary table 16.

npg

© 2

016

Nat

ure

Am

eric

a, In

c. A

ll rig

hts

rese

rved

.np

2016

Nat

ure

Am

eric

a, In

c. A

ll rig

hts

rese

rved

.

Page 6: Divergent modes of clonal spread and intraperitoneal mixing in … · 2019-08-06 · High-grade serous ovarian cancer (HGSOvCa) is the most lethal . histotype of ovarian cancer, where

Nature GeNetics VOLUME 48 | NUMBER 7 | JULY 2016 763

A rt i c l e s

samples harboring each clone as a function of the maximum prevalence observed across all samples and identified clones possessing three phe-notypes with respect to spread in the peritoneal cavity: clones with site

specificity, clones with the capacity to colonize and become dominant, and high-mobility clones that never achieve dominance (Fig. 5c). Clones in these three groupings were distributed non-randomly

ROv1

ROv2

Om1

Om2

Clone phylogeny

Patient 2

A

B

C

D

E F

Om1

Om2

LOv1

LOv2

ROv1

ROv3

ROv4

ROvA4

ROvA9 OmC1

LFTB2

ROv1

ROv2

Clone phylogeny

Patient 9

A

B

C

ROv3

ROv4

ROvA5

RPvSD1

LPvS

LPvSC1

LOvB2

ROv1

ROv2

AB

CD

E

F

G

H

Clone phylogeny

Patient 4

Clone phylogeny

Patient 10

A

B

C

D

E F

ROvA4

ROv4

ROv3

ROv2

ROv1

RFTA16 Om1

SBwl

SBwlE4

LFTB4

LOvB2

ApC1

Clone phylogeny

Patient 1

A

B C

DE

FG

H I

ROv1

ROv2

ROvA7

RFTA2

Om1

OmF2

CDSB1

ClnE1

Adnx

LFTC1

LOvC5

Clone phylogeny

Patient 3

A

B

C D

E

F G

RUtD2

RUtD3

ROvC4

ROvC5

ROvC6

LOv1

LOvA10

LOvA4

BrnM

BrnMA1

BwlA6

RPvM

RUtD1

PolyphyleticMonophyleticPure

Mixture classification

A

B

C

D

E

Clone phylogeny

Patient 7

1 2

3

Figure 4 Clone phylogenies and sample clone mixtures. A clone phylogeny is shown for each patient, with distinct clonal genotypes color-coded. The clonal composition of each sample is depicted as a schematic tumor, with the coloring of ‘cells’ proportional to the prevalence of each clone. Samples are arrayed in a circle around the anatomical diagram, with constituent clones from the overall clone phylogeny represented in an outer circle. Index samples sequenced by whole-genome sequencing are underlined. Pure, monophyletic, and polyphyletic samples are indicated by circles, triangles, and stars, respectively, and correspond to data points in Figure 5a. Samples obtained at the first, second, and third time points from patient 7 are indicated by number. Patients with prevalence patterns consistent with multiple reseeding events and/or extensive mixing (patients 3 and 10) are underlined. Fully quantitative prevalences are given in supplementary table 9.

npg

© 2

016

Nat

ure

Am

eric

a, In

c. A

ll rig

hts

rese

rved

.np

2016

Nat

ure

Am

eric

a, In

c. A

ll rig

hts

rese

rved

.

Page 7: Divergent modes of clonal spread and intraperitoneal mixing in … · 2019-08-06 · High-grade serous ovarian cancer (HGSOvCa) is the most lethal . histotype of ovarian cancer, where

764 VOLUME 48 | NUMBER 7 | JULY 2016 Nature GeNetics

A rt i c l e s

in patients. Patients 3 and 10 were enriched for subdominant clones that were present at multiple intraperitoneal sites—ROvA9 and LFTB2 for clone B of patient 10 and CDSB1, Om1, ClnE2, and ROvA7 for clone B of patient 3 (Fig. 4)—consistent with multiple reseeding events and extensive intraperitoneal mixing. Furthermore, the preva-lence of clone B in patient 3 was found to be correlated across sam-ples with the prevalence of clone F, suggesting that these polyphyletic clones migrated together or reseeded multiple sites (Fig. 5e). By contrast, all pairwise correlations of clones for patients 1, 7, and 9 were negative (Fig. 5e), suggesting that the majority of sites were populated by a single clone or a monophyletic mixture. The spreading and invasion capacity of clones was decidedly non-uniform, indi-cating that there is widespread variation in the propensity of clones to mix and invade. Together, the variation in sample diversity and clonal migration patterns define two distinct modes of intraperitoneal mixing across patients, corresponding to patients with a single site permissive of clonal diversity and monoclonal seeding and patients exhibiting extensive polyclonal mixing and reseeding.

MutationofovariancancergenesindivergentclonesWe next analyzed the distribution of mutations affecting the known driver genes TP53, BRCA1 and BRCA2, RB1, NF1, PTEN, and RAD51B across the clonal phylogenies to determine the timing of acquisition and clone specificity of these events. TP53 mutation was

detected in all clones (Supplementary Table 10), accompanied by loss of heterozygosity (LOH) of chromosome 17 in all whole-genome sequencing discovery samples (Supplementary Figs. 7–13), indicat-ing an ancestral role for TP53 mutation, consistent with previous stud-ies1–3,22. Additional ancestral changes included mutations of CDK12 in patient 1, BRCA2 (stop codon) in patient 4, and FAT3 in patient 10 (Fig. 1a and Supplementary Table 10). We observed many clade-specific (and, therefore, evolutionarily ‘late’) driver events, including clone-specific homozygous segmental deletions affecting CDKN2A and WWOX in the right ovary clones of patient 1, MAP2K4 in the ROv2 clone of patient 10, and LRP1B in the non-ROv2 clones of patient 10 (Supplementary Figs. 25–29). In patient 4, homozygous deletion of ANKRD11 was detected exclusively in the LPvS sample. In patient 7, homozygous deletion of NF1 was specific to right utero-sacral samples from time point 1 and brain metastasis samples from time point 2 (Supplementary Fig. 30). Clone-specific focal high-level amplification of ERBB2 was identified in patient 9 (Supplementary Figs. 31–33), with single-nucleus sequencing confirming that the clone harboring this amplification was present in the primary ovary site and ubiquitous in all peritoneal metastases (Fig. 3c).

ConvergentevolutionofthestructuralgenomeFinally, we determined the presence of convergent evolution resulting from copy number alterations in specific clones. Five patients showed

12347910

CladePrimary ovaryPeritoneal

0 0.2 0.4 0.6 0.8 1.0Clone divergence

0

0.5

1.0

1.5

2.0

Sam

ple

mix

ture

ent

ropy

1 LOvB2

2 ROv2

7 RUtD39 LOv2

10 ROvA9

n = 13

Patient

Mixtureclassification

PolyphyleticMonophyleticPure

a

10 3 1 7 2 4 9Patient

0

0.5

1.0

1.5

2.0

2.5

Mig

ratio

ns p

er m

etas

tatic

site

d

0 0.2 0.4 0.6 0.8 1.0Maximum clade prevalence

0

2

4

6

8

10

12

14

Num

ber

of s

ampl

es w

ith th

ecl

one

pres

ent

3 B

10 F

0

0.2

0.4

0.6

0.8

1.0

Mea

n pr

eval

ence

Primary ovary samples

1 2 3 4 7 9 10Patient

0

0.20.4

0.6

0.8

1.0

Mea

n pr

eval

ence

Peritoneal samples

cb

A B C D E FClone

A

B

C

D

E

F

Clo

ne

Patient 10

A B C D E FClone

A

B

C

D

E

F

Clo

ne

Patient 2

A B C D E F GClone

A

B

C

D

E

F

G

Clo

ne

Patient 3

A B C D E F G H

Clone

A

B

C

D

E

F

G

H

Clo

ne

Patient 4

A B C D EClone

A

B

C

D

E

Clo

ne

Patient 7

A B CClone

A

B

C

Clo

ne

Patient 9

A B C D E F G H IClone

A

B

C

D

E

F

G

H

I

Clo

ne

Patient 1

−1.0−0.8−0.6−0.4−0.2

Correlation

00.20.40.60.81.0

eFigure 5 Analysis of clonal phylogenies and clone mixtures. (a) The relationship between clone divergence and sample mixture entropy for clones present in each sample of each patient. Divergence was calculated as the Jaccard index of clonal genotypes. Only clones with a prevalence greater than 0.01 were included in the calculation. Scatter points are colored by patient, and grayscale contours show a kernel density estimate of the data points. (b) Each major clade was labeled as primary or non-primary on the basis of which clade had higher mean prevalence in putative primary samples. Bars show the prevalence of the primary (blue) and non-primary (green) clades, with putative primary samples shown in the upper plot and non-primary samples shown in the bottom plot. Bar height corresponds to the mean prevalence; error bars show the 95% confidence interval for 1,000 bootstrap samples. (c) Relationship between the pervasiveness (y axis) and dominance (x axis) of each clade in the clone phylogeny. Pervasiveness is measured as the number of samples in which the clade is present; dominance corresponds to maximum clade prevalence (the sum of the prevalence for a clone and its descendants) across samples in which the clade is present. Scatter points are colored by patient. (d) Bar plot of the number of migration events per site for each patient’s most parsimonious migration scenario. (e) Correlations of sample prevalences for clone pairs. Only correlations for clone pairs from divergent lineages are shown; those for clones with an ancestral–descendant relationship were not considered (gray).

npg

© 2

016

Nat

ure

Am

eric

a, In

c. A

ll rig

hts

rese

rved

.np

2016

Nat

ure

Am

eric

a, In

c. A

ll rig

hts

rese

rved

.

Page 8: Divergent modes of clonal spread and intraperitoneal mixing in … · 2019-08-06 · High-grade serous ovarian cancer (HGSOvCa) is the most lethal . histotype of ovarian cancer, where

Nature GeNetics VOLUME 48 | NUMBER 7 | JULY 2016 765

A rt i c l e s

evidence of the independent acquisition of breakpoints leading to LOH events or amplifications affecting known cancer-related genes. The total length of the segments classified as LOH in any sample ranged from 1,100 to 1,600 Mb across patients (Supplementary Table 11). In patients for whom analysis was not confounded by divergent mix-tures, between 1.5 and 7.2% of LOH segments, by length, could not have originated on a single branch of the clone phylogeny and are thus the result of convergent LOH (Supplementary Table 11).

Clone-specific rearrangements leading to LOH of RAD51B were identified in three patients. For patient 10, multiple RAD51B

rearrangements were detected in samples composed of descendant clones but were absent from the two samples (LFTB2 and OmC1) dom-inated by the ancestral (or ancestrally divergent) clone. In patient 3, RAD51B was affected by two rearrangements, including one event common to all samples and a ROv2-specific event. Patient 7 exhibited convergent evolution toward LOH of chromosome 14, with two distinct events independently deleting the reference allele of a SNP affecting a splice site of RAD51B (Fig. 6a). Both RAD51B LOH clones coexisted in the RUtD3 sample at time point 1, with clone B eventually dominating the brain metastasis at time point 2, clone D dominating

0 20 40 60 80 100

01234

Brn

M

RAD51B

0 20 40 60 80 100 120 140 160

01234

0 20 40 60

01234

0 20 40 60 80 100

01234

0 20 40 60 80 100

01234

LOv1

RAD51B

0 20 40 60 80 100 120 140 160

01234

0 20 40 60

01234

0 20 40 60 80 100

0 20 40 60 80 100 0 20 40 60 80 100 120 140 160 0 20 40 60 0 20 40 60 80 100

01234

Chromosome 14 position (Mb)

01234

RP

vM

RAD51B

Chromosome 8 position (Mb)

01234

Chromosome 19 position (Mb)

01234

Chromosome 16 position (Mb)

01234

28.0 29.0 30.0 31.0

Chromosome 19 position (Mb)

32.0 33.0 34.0 35.0

0

5

10

15

20

25

30

Cop

y nu

mbe

r

CCNE1

Om1Om2ROv1ROv2

8000

0579

7875

7965

5579

6555

7978

4180

2593

7997

1679

9716

7978

4179

6582

8020

7179

6560

7978

7180

0024

7965

6080

0024

7978

7779

7824

7978

7180

1629

7979

0979

7909

7269

0079

7877

7965

7179

6609

7979

1779

7862

6539

5479

6609

8005

8665

4245

7965

5779

6557

4990

8379

6556

8000

6679

7862

7966

1079

6610

Breakpoint ID

Om1

Om2

ROv1

ROv2

Sam

ple

Omentum Right ovary

Anatomical site

−5

0

5

10

15

Cop

y nu

mbe

r di

ffere

nce

n = 38 n = 10

P = 0.0069

n = 15 n = 33

Mann–Whitney U

P = 0.00024

Breakpoint status

AbsentPresent

a

cb

Figure 6 Examples of convergent evolution of copy number. (a) Copy numbers for major (red) and minor (blue) alleles for chromosome 14 of patient 7, with the positions of experimentally validated breakpoints indicated (colored arcs). The brain metastasis (BrnM) and right pelvic mass (RPvM) recurrences exhibit LOH of distinct segments of chromosome 14, as corroborated by the presence of distinct sets of breakpoints. (b) The top plot shows total copy number for omentum (Om1, Om2) and right ovary (ROv1, ROv2) samples in patient 2. The bottom plot shows the presence (blue) or absence (white) of rearrangement breakpoints, as identified by analysis of discordant paired-end reads, with break ends in the CCNE1 amplified region. Green lines show correspondence between rearrangement breakpoints in the heat map and the positions of break ends, highlighting the association between breakpoints and copy number transitions. (c) Break ends are associated with significantly greater copy number changes in samples in which they are predicted to be present versus those in which they are absent. Shown is the distribution of the difference in copy number between segments on the right and left of each break end, correcting for break end orientation. A greater copy number difference indicates a greater copy number change induced by the breakpoint. For each break end in each sample, we predicted whether the break end existed in that sample. We then compared the copy number difference in each sample for break ends that were predicted to be absent (left) or present (right) in those samples.

npg

© 2

016

Nat

ure

Am

eric

a, In

c. A

ll rig

hts

rese

rved

.np

2016

Nat

ure

Am

eric

a, In

c. A

ll rig

hts

rese

rved

.

Page 9: Divergent modes of clonal spread and intraperitoneal mixing in … · 2019-08-06 · High-grade serous ovarian cancer (HGSOvCa) is the most lethal . histotype of ovarian cancer, where

766 VOLUME 48 | NUMBER 7 | JULY 2016 Nature GeNetics

A rt i c l e s

the right pelvic samples at time point 3, and RAD51B-heterozygous clones extinguished at both the later time points.

Five patients exhibited chromosome-arm-level amplification affecting known oncogenes: CCNE1 in patients 2, 9, and 10; KRAS in patient 3; and MYC in patient 7. The predicted numbers of copies were highly variable among discovery samples (between 10 and 20 copies of CCNE1 in patient 2, for example, as confirmed by FISH analysis) (Supplementary Figs. 34 and 35, and Supplementary Table 12). Unbalanced intrachromosomal rearrangements were the predomi-nant class of events (Supplementary Figs. 36–42). The distributions of rearrangement breakpoints throughout the samples were indicative of a dynamic process of progressive amplification conferring distinct sets of breakpoints to specific clones. For patient 3, the presence/absence profiles of breakpoints associated with KRAS amplification were moderately heterogeneous across samples, although shared clonal mixtures in this patient prevented unambiguous assignment of breakpoints to clones (Supplementary Fig. 43). In patient 7, several breakpoints associated with MYC amplification were specific to a sub-set of samples (Supplementary Fig. 44). In patient 2, the distribution of breakpoints found within the CCNE1-amplified region recapitu-lated the distinct clonal lineages in right ovary and omentum sites identified by maximum likelihood for both SNVs and breakpoints (Fig. 6b and Supplementary Figs. 2 and 5). For all 28 breakpoints within the CCNE1-amplified region (Supplementary Table 13 and Supplementary Note), the posterior probability of originating on the ancestral branch was <0.001, indicating that acquisitions were sam-ple specific and, therefore, occurred independently (Supplementary Figs. 45–47). Furthermore, break ends within the CCNE1-amplified region coincided with significantly greater copy number differences in samples in which they were predicted to be present, as compared to samples in which they were predicted to be absent (Fig. 6c), confirm-ing that sample-specific breakpoints induced sample-specific copy number changes (Supplementary Note).

DISCUSSIONOur findings challenge the notion that the intraperitoneal spread-ing of cell populations should result in widespread clonal mixing within a local potential space, as only a minority of patients exhib-ited patterns consistent with extensive migration and reseeding of polyclonal mixtures at multiple sites. This observation implies that the selection potential of the peritoneal cavity is likely non-uniform because of specific microenvironmental properties. Despite the lack of treatment-induced selective pressures, established clones present at diagnosis may have been selected by the intrinsic topology of the intraperitoneal fitness landscape. Intriguingly, we found that clones could be categorized by three modes of spreading behavior, providing a rationale for linking genotypic mechanisms to the differential spreading capacity of individual clones. Further study on larger series will be needed to confirm the generalizability of these find-ings. Ultimately, comparisons of primary and relapse samples over time would show whether clones with a high degree of spreading capacity could dominate in the presence of combined constraints from microenvironmental and therapeutic selective pressures. For example, in patient 7, we studied two recurrences. Notably, diagnos-tic specimens already contained clones with properties of intrinsic resistance before therapeutic intervention, leading to brain metas-tasis and right pelvic mass recurrences, respectively, at the two later time points. Whether such diverse pretreatment samples could be selectively targeted remains an open question.

Together, the properties of HGSOvCa evolution and the distinct modes of local intra-abdominal spread present an opportunity to study

how these patterns relate to treatment outcomes. We note that the properties uncovered in our analysis have anecdotal association with clinical outcomes. Patients whose divergent clones were dominated by the age-related mutational signature (and not the mutational signature corresponding to homologous recombination deficiency; patients 2 and 9) had the poorest survival outcomes, and both died of disease (Table 1). Patients 2 and 9 were also characterized by monoclonal seed-ing patterns, exhibiting the fewest migrations per site. By contrast, the patients with the most migrations per site (patients 1, 3, and 10) did not succumb to disease, with patients 1 and 10 showing no evidence of disease after treatment within the follow-up period. Our study design is constrained by the limitations of surgical sampling and is likely characterizing a subset of the clonal diversity present in each patient. Nevertheless, with a median of n = 10 samples per patient, we always detected a sample representing diversity, allowing for interpretation of spatial clonal dynamics in the other sites. Furthermore, we note that most conclusions about ovarian cancer biology are derived from study-ing single specimens. Our observations and those of others5,23 empha-size that the analysis of single samples provides a dramatically restricted view of tumor biology. The analytical and experimental approaches presented here will inform the study of larger series, including rapid autopsies2,12, where clones will have escaped numerous therapeutic interventions. In addition, preclinical model system24 and clinical trial studies assaying response to pharmacologic and biologic agents will be informed by the evolutionary properties reported here.

Our results identify several properties of HGSOvCa evolution, includ-ing loss of mutations due to copy number deletion, convergent evolu-tion of the structural genome, and temporal ‘activation’ of mutational processes. Loss of mutations and structural convergence will affect ana-lytical approaches for reconstructing evolutionary histories from mul-tiregion sequencing experiments. Many genomically unstable tumors like HGSOvCa will likely exhibit properties that violate the assumptions imposed by perfect and persistent phylogeny. Ignoring these additional complexities will inevitably lead to the erroneous inference of both sample trees and the temporal ordering of mutations if convergence and mutation loss are not taken into account. Notably, our observation of convergent amplification of driver genes such as CCNE1 raises new hypotheses that clade-specific clones harboring independently acquired amplifications would respond differently to intervention.

In summary, we have presented a detailed view of HGSOvCa clonal dynamics within the context of intraperitoneal local spread. Our results provide an impetus for dissecting the specific biological mechanisms underlying the interaction of fitness properties and the evolutionary selection governing the invasive capacity of ovarian cancers.

URLs. BC Cancer Agency Cancer Management Guidelines, http://www.bccancer.bc.ca/health-professionals/professional-resources/cancer-management-guidelines; Dollo source code, https://bitbucket.org/dranew/dollo; interactive visualization of patient-specific clonal maps, http://compbio.bccrc.ca/research/tumour-evolution/.

METHODSMethods and any associated references are available in the online version of the paper.

Accession codes. Sequencing is available for download from the European Genome-phenome Archive (EGA) under study accession EGAS00001000547.

Note: Any Supplementary Information and Source Data files are available in the online version of the paper.

npg

© 2

016

Nat

ure

Am

eric

a, In

c. A

ll rig

hts

rese

rved

.np

2016

Nat

ure

Am

eric

a, In

c. A

ll rig

hts

rese

rved

.

Page 10: Divergent modes of clonal spread and intraperitoneal mixing in … · 2019-08-06 · High-grade serous ovarian cancer (HGSOvCa) is the most lethal . histotype of ovarian cancer, where

Nature GeNetics VOLUME 48 | NUMBER 7 | JULY 2016 767

A rt i c l e s

ACKNoWLEDGMENTSWe thank V. Earle for the artwork depicting anatomical sites sampled in the study. We acknowledge generous long-term funding support provided by the BC Cancer Foundation. In addition, the groups of S.P.S. and S.A. receive operating funds from the Canadian Breast Cancer Foundation, the Canadian Cancer Society Research Institute (grant 701584), the Terry Fox Research Institute, Genome Canada/Genome BC (173-CIC and 177-EVO), the Canadian Institutes for Health Research (CIHR) (MOP-115170, MOP-126119, and FDN-143246), a new investigator grant to J.N.M. (MSH-261515), and a TFRI new investigator award to S.P.S. S.P.S. and S.A. are supported by Canada Research Chairs. S.P.S. is a Michael Smith Foundation for Health Research scholar. A.M. is supported by a NSERC CGS scholarship. A.R. is supported by a CIHR CGS scholarship. T.M. is supported by a Canadian Breast Cancer Foundation British Columbia/Yukon Postdoctoral Fellowship.

AUTHoR CoNTRIBUTIoNSA.M. and A.R. performed algorithm development and software implementation and led the data analysis. G.H., A.W.Z., K.S., C.S., J.R., and A.B. performed data analysis and bioinformatics. E.L., T.M., J.B., D.Y., A.W., and J.K. performed single-nucleus sequencing. L.M.P., S.K., J.S., and W.Y. performed sample preparation and validation experiments. M.A.S. and C.B.N. performed data visualization. A.K., H.L.C., J.H., and N.M. performed immunohistochemistry and FISH analyses. R.M., A.J.M., and M.A.M. performed library construction and genome sequencing. C.B.G. and D.G.H. analyzed molecular and histological pathology. A.B.-C. contributed to algorithm development. J.N.M. performed surgery and tumor banking and was a clinical leader. S.C.M. edited the manuscript. A.M., A.R., S.A., and S.P.S. wrote the manuscript. S.A. oversaw experimental design and single-cell sequencing. S.P.S. is the senior responsible author and conceived the project and provided oversight.

COMPETING FINANCIAL INTERESTSThe authors declare no competing financial interests.

Reprints and permissions information is available online at http://www.nature.com/reprints/index.html.

1. Ahmed, A.A. et al. Driver mutations in TP53 are ubiquitous in high grade serous carcinoma of the ovary. J. Pathol. 221, 49–56 (2010).

2. Patch, A.-M. et al. Whole-genome characterization of chemoresistant ovarian cancer. Nature 521, 489–494 (2015).

3. Bashashati, A. et al. Distinct evolutionary trajectories of primary high-grade serous ovarian cancers revealed through spatial mutational profiling. J. Pathol. 231, 21–34 (2013).

4. Ha, G. et al. TITAN: inference of copy number architectures in clonal cell populations from tumor whole-genome sequence data. Genome Res. 24, 1881–1893 (2014).

5. Schwarz, R.F. et al. Spatial and temporal heterogeneity in high-grade serous ovarian cancer: a phylogenetic analysis. PLoS Med. 12, e1001789 (2015).

6. Jacobs, I.J. et al. Clonal origin of epithelial ovarian carcinoma: analysis by loss of heterozygosity, p53 mutation, and X-chromosome inactivation. J. Natl. Cancer Inst. 84, 1793–1798 (1992).

7. Khalique, L. et al. The clonal evolution of metastases from primary serous epithelial ovarian cancers. Int. J. Cancer 124, 1579–1586 (2009).

8. Lengyel, E. Ovarian cancer development and metastasis. Am. J. Pathol. 177, 1053–1064 (2010).

9. Campbell, P.J. et al. The patterns and dynamics of genomic instability in metastatic pancreatic cancer. Nature 467, 1109–1113 (2010).

10. Gerlinger, M. et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 366, 883–892 (2012).

11. Gerlinger, M. et al. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat. Genet. 46, 225–233 (2014).

12. Gundem, G. et al. The evolutionary history of lethal metastatic prostate cancer. Nature 520, 353–357 (2015).

13. Eirew, P. et al. Dynamics of genomic clones in breast cancer patient xenografts at single-cell resolution. Nature 518, 422–426 (2015).

14. Gawad, C., Koh, W. & Quake, S.R. Dissecting the clonal origins of childhood acute lymphoblastic leukemia by single-cell genomics. Proc. Natl. Acad. Sci. USA 111, 17947–17952 (2014).

15. Wang, Y. et al. Clonal evolution in breast cancer revealed by single nucleus genome sequencing. Nature 512, 155–160 (2014).

16. Alekseyenko, A.V., Lee, C.J. & Suchard, M.A. Wagner and Dollo: a stochastic duet by composing two parsimonious solos. Syst. Biol. 57, 772–784 (2008).

17. Ryder, R.J. & Nicholls, G.K. Missing data in a stochastic Dollo model for binary trait data, and its application to the dating of Proto-Indo-European. J. R. Stat. Soc. Ser. C Appl. Stat. 60, 71–92 (2011).

18. Alexandrov, L.B. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013).

19. Blei, D.M., Ng, A.Y. & Jordan, M.I. Latent dirichlet allocation. J. Machine Learning Res. 3, 993–1022 (2003).

20. Bamford, S. et al. The COSMIC (Catalogue of Somatic Mutations in Cancer) database and website. Br. J. Cancer 91, 355–358 (2004).

21. Roth, A. et al. Clonal genotype and population structure inference from single-cell tumor sequencing. Nat. Methods http://dx.doi.org/10.1038/nmeth.3867 (2016).

22. Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature 474, 609–615 (2011).

23. Hoogstraat, M. et al. Genomic and transcriptomic plasticity in treatment-naive ovarian cancer. Genome Res. 24, 200–211 (2014).

24. Cooke, S.L. & Brenton, J.D. Evolution of platinum resistance in high-grade serous ovarian cancer. Lancet Oncol. 12, 1169–1174 (2011).

npg

© 2

016

Nat

ure

Am

eric

a, In

c. A

ll rig

hts

rese

rved

.np

2016

Nat

ure

Am

eric

a, In

c. A

ll rig

hts

rese

rved

.

Page 11: Divergent modes of clonal spread and intraperitoneal mixing in … · 2019-08-06 · High-grade serous ovarian cancer (HGSOvCa) is the most lethal . histotype of ovarian cancer, where

Nature GeNetics doi:10.1038/ng.3573

ONLINEMETHODSPatient consent and surgery. Ethical approval was obtained from the University of British Columbia (UBC) Ethics Board. Women undergoing debulking surgery (primary or recurrent) for carcinoma of ovarian, perito-neal, and/or fallopian tube origin were approached for informed consent for the banking of tumor tissue. Cases of HGSOvCa where more than one sample was collected in different anatomical locations (for example, different loca-tions within the ovary and omentum) or where material was available at dif-ferent time points (for example, at primary surgery and at recurrence) were chosen for this analysis. Clinicopathological and outcome data were collected by chart review. Consistent with practices at UBC and the British Columbia Cancer Agency, all patients with high-grade serous cancer are referred to the hereditary cancer clinic and offered genetic testing for BRCA1 and BRCA2 mutations25,26.

Specimen preservation and histological evaluation. In cases identified as having high-grade serous histology, multiple tissue samples were obtained from the primary ovarian tumor and metastatic sites where adequate tumor volume permitted. When the ovary was pathologically enlarged, samplings were taken from up to five different areas, with an effort made to equally space sampling sites while staying within the bounds of grossly apparent tumor tissue. Each sampling was cut into three pieces, yielding two end pieces for cryovials and a middle portion placed in 10% buffered formalin. All paraffin-embedded blocks, including formalin-fixed tumor samples and molecularly fixed fallopian tubes, were sectioned and stained with hematoxylin and eosin before expert histopathological review (C.B.G.) to confirm the presence of high-grade serous carcinoma.

Library construction and sequencing. Samples for a total of ten patients were submitted for library construction and sequencing. Sample size was deter-mined by the availability of resectable, cryopreserved tissue and also by DNA quality. Samples from patients 5 and 6 were excluded because of low tumor cel-lularity, as indicated by the absence of SNVs and copy number changes. Patient 8 was excluded as contaminated because of the high number of somatic vari-ants present in dbSNP. For all tumor and normal samples, DNA extraction was followed by library construction and sequencing using Illumina HiSeq 2000

whole-genome shotgun v3 chemistry with paired-end 100-bp reads. Samples were sequenced to an average of 30× coverage (Supplementary Table 2).

Targeted deep sequencing. To resolve the clonal composition of each sample, we performed targeted deep sequencing of 300 loci per patient with multiplex PCR amplicons sequenced to a median of 7,700× coverage (Supplementary Tables 10, 14, and 15). These data were used to confirm the predicted presence/absence patterns of SNVs and breakpoints from whole-genome sequencing and to infer the clonal prevalence of SNV clusters using PyClone27 (Supplementary Table 16). We defined a genotype as a set of present or absent PyClone SNV clusters and breakpoints. The set of aberrations present at a cel-lular prevalence of >50% in a sample provides an estimate of the genotype of the clone dominant in that sample. Furthermore, the sample tree provides an estimate of the phylogeny relating dominant clones. Under these assumptions, we estimated clonal genotypes and clone phylogenies.

Targeted single-cell sequencing. Individual nuclei from each sample were subjected to multiplex PCR reactions on Fluidigm Access arrays with libraries sequenced on Illumina MiSeq machines as previously reported13. We designed primers for 48 loci for patients 2 and 9 and for 96 loci for patient 3 (Supplementary Table 17). We sequenced 1,680 cells in total to a median depth of 100× per locus (Table 2 and Supplementary Tables 18 and 19). Resulting data produced read counts for each targeted locus in each sequenced nucleus. Nuclei were clustered, and clonal genotypes were inferred using the SCG model21 version 0.3.0; see the Supplementary Note for parameter settings. In each patient, heterozygous SNPs predicted to be rendered homozygous reference in tumors from whole-genome sequencing analysis were used to distinguish normal nuclei from tumor nuclei.

Additional methods are described in detail in the Supplementary Note.

25. Schrader, K.A. et al. Germline BRCA1 and BRCA2 mutations in ovarian cancer: utility of a histology-based referral strategy. Obstet. Gynecol. 120, 235–240 (2012).

26. McAlpine, J.N. et al. BRCA1 and BRCA2 mutations correlate with TP53 abnormalities and presence of immune cell infiltrates in ovarian high-grade serous carcinoma. Mod. Pathol. 25, 740–750 (2012).

27. Roth, A. et al. PyClone: statistical inference of clonal population structure in cancer. Nat. Methods 11, 396–398 (2014).

npg

© 2

016

Nat

ure

Am

eric

a, In

c. A

ll rig

hts

rese

rved

.np

2016

Nat

ure

Am

eric

a, In

c. A

ll rig

hts

rese

rved

.