ArticlePDF Available

Genomic Evolution of Breast Cancer Metastasis and Relapse

Authors:

Abstract and Figures

Patterns of genomic evolution between primary and metastatic breast cancer have not been studied in large numbers, despite patients with metastatic breast cancer having dismal survival. We sequenced whole genomes or a panel of 365 genes on 299 samples from 170 patients with locally relapsed or metastatic breast cancer. Several lines of analysis indicate that clones seeding metastasis or relapse disseminate late from primary tumors, but continue to acquire mutations, mostly accessing the same mutational processes active in the primary tumor. Most distant metastases acquired driver mutations not seen in the primary tumor, drawing from a wider repertoire of cancer genes than early drivers. These include a number of clinically actionable alterations and mutations inactivating SWI-SNF and JAK2-STAT3 pathways.
Content may be subject to copyright.
Article
Genomic Evolution of Breast Cancer Metastasis and
Relapse
Graphical Abstract
Highlights
dMetastases mostly disseminate late from primary breast
tumors, keeping most drivers
dDrivers at relapse sample from a wider range of cancer genes
than in primary tumors
dMutations in SWI-SNF complex and inactivated JAK-STAT
signaling enriched at relapse
dMutational processes similar in primary and relapse;
radiotherapy can damage genome
Authors
Lucy R. Yates, Stian Knappskog,
David Wedge, ..., Andrew Tutt,
Per Eystein Lønning, Peter J. Campbell
Correspondence
per.eystein.lonning@helse-bergen.no
(P.E.L.),
pc8@sanger.ac.uk (P.J.C.)
In Brief
By sequencing primary, locally relapsed,
and metastatic breast cancers, Yates
et al. show that clones seeding
metastasis or relapse disseminate late
from primary tumors but continue to
acquire mutations, including clinically
actionable alterations and mutations
inactivating the SWI/SNF and JAK2-
STAT3 pathways.
Molecular time
Cancer burden
Distant
metastasis
Loco-
regional
relapse
Synchronous
lymph node
metastasis
Canonical cancer genes -
TP53, GATA3, PIK3CA, AKT1, ERBB2
MRCA
Normal
cell
Genome-
doubling
Rarer cancer genes -
SWI/SNF, JAK-STAT
Drug resistance mutations -
ESR1
Driver
mutations
Radical treatment
of primary breast cancer
Yates et al., 2017, Cancer Cell 32, 169–184
August 14, 2017 ª2017 The Authors. Published by Elsevier Inc.
http://dx.doi.org/10.1016/j.ccell.2017.07.005
Cancer Cell
Article
Genomic Evolution of Breast Cancer
Metastasis and Relapse
Lucy R. Yates,
1,2,23
Stian Knappskog,
3,4,23
David Wedge,
1,5
James H.R. Farmery,
6
Santiago Gonzalez,
1,7
Inigo Martincorena,
1
Ludmil B. Alexandrov,
8,9,10
Peter Van Loo,
11,12
Hans Kristian Haugland,
13,14
Peer Kaare Lilleng,
13,14
Gunes Gundem,
1,15
Moritz Gerstung,
1,7
Elli Pappaemmanuil,
1,15
Patrycja Gazinska,
16
Shriram G. Bhosle,
1
(Author list continued on next page)
SUMMARY
Patterns of genomic evolution between primary and metastatic breast cancer have not been studied in large
numbers, despite patients with metastatic breast cancer having dismal survival. We sequenced whole ge-
nomes or a panel of 365 genes on 299 samples from 170 patients with locally relapsed or metastatic breast
cancer. Several lines of analysis indicate that clones seeding metastasis or relapse disseminate late from pri-
mary tumors, but continue to acquire mutations, mostly accessing the same mutational processes active in
the primary tumor. Most distant metastases acquired driver mutations not seen in the primary tumor, drawing
from a wider repertoire of cancer genes than early drivers. These include a number of clinically actionable
alterations and mutations inactivating SWI-SNF and JAK2-STAT3 pathways.
INTRODUCTION
Metastatic breast cancer is almost universally fatal within 5–10
years, a dismal statistic that has not changed much in the past
20–30 years (Tevaarwerk et al., 2013). Breast cancer recurrence
can take two forms: distant metastasis (commonly bone, brain,
liver, lung, and distant lymph nodes) and locoregional relapse
(recurrence in breast, chest wall, or regional lymph nodes). Lo-
coregional relapse occurs in about 10% of patients despite
optimal management of the primary tumor and is associated
1
Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
2
Department of Clinical Oncology, Guys and St Thomas’ NHS Trust, London SE1 9RT, UK
3
Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway
4
Department of Oncology, Haukeland University Hospital, Bergen, Norway
5
Big Data Institute, University of Oxford, Oxford OX3 7BN, UK
6
Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
7
European Bioinformatics Institute EMBL-EBI, Wellcome Genome Campus, Hinxton CB10 1SD, UK
8
Theoretical Biology and Biophysics (T-6), Los Alamos National Laboratory, Los Alamos, NM 87545, USA
9
Center for Nonlinear Studies, Los Alamos National Laboratory, Los Alamos, NM 87545, USA
10
University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87102, USA
11
The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
12
Department of Human Genetics, University of Leuven, 3000 Leuven, Belgium
13
Department of Pathology, Haukeland University Hospital, Bergen, Norway
14
The Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
15
Computational Oncology, Epidemiology and Biostatistics Memorial Sloan Kettering Cancer Institute, New York, NY 10065 USA
16
Division of Cancer Studies, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9RT, UK
(Affiliations continued on next page)
Significance
These findings have implications for personalized therapy of breast cancer. The late dissemination of cells that seed metas-
tasis or local relapse suggests that the primary tumor genome can proxy for the genome of disseminated cells at the time of
first diagnosis, supporting the use of genome sequencing to aid decisions about adjuvant therapy for primary breast cancer.
Biopsy and sequencing of metastases may be helpful in some patients because most distant metastases have acquired
additional driver mutations not seen in the primary; these often involve potentially actionable genes and cellular pathways.
Sequencing local recurrences can distinguish a genuine relapse from a second primary cancer, two scenarios with very
different care pathways.
Cancer Cell 32, 169–184, August 14, 2017 ª2017 The Authors. Published by Elsevier Inc. 169
This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
with concomitant or future distant metastatic disease in 30%
and 60% of cases, respectively. In contrast, regional lymph
node metastasis found at the time of primary diagnosis is often
cured with surgery and radiotherapy but is a well-established
poor prognostic factor, associated with a higher risk of subse-
quent cancer recurrence.
Molecular profiling of breast cancer has typically focused on
the primary breast lesion. Gene expression profiles classify
breast cancers into different subtypes, with clinical trials
showing that these transcriptional signatures can be used to
support therapeutic decisions in primary breast cancer (Harris
et al., 2016). Large-scale genomics analyses have now been per-
formed in thousands of primary breast cancers, revealing the
complex mutational landscape of the disease (Banerji et al.,
2012; Cancer Genome Atlas Network, 2012; Ciriello et al.,
2015; Ellis et al., 2012; Nik-Zainal et al., 2016; Shah
et al., 2012; Stephens et al., 2012). General patterns to emerge
from these studies include that estrogen receptor (ER)-positive
primary breast cancer has a characteristic ‘‘luminal’’ transcrip-
tional profile with frequent somatic mutations activating PI3K-
AKT signaling and inactivating GATA3 and the JUN kinase
pathway. Breast cancers with amplification and/or overexpres-
sion of ERBB2 (also known as HER2) have a distinct transcrip-
tional and genomic profile, confirming the central role that
ERBB2 plays in the pathogenesis of this subtype of breast can-
cer. Breast cancers negative for ER, the progesterone receptor
(PR), and HER2, so-called triple-negative breast cancers, are
characterized by a ‘‘basal-like’’ transcriptional profile, frequent
TP53 mutation, and extensive copy number variation. A number
of studies have revealed extensive genomic heterogeneity within
primary breast tumors and changes in subclonal structure during
systemic therapy (Balko et al., 2014; Gellert et al., 2016; Miller
et al., 2016; Ng et al., 2015; Shah et al., 2012; Wang et al.,
2014; Yates et al., 2015).
While the genome of primary breast cancer has been well
characterized, there has been considerably less analysis of
relapsed or metastatic breast cancer. Those studies that have
been performed have revealed that metastases are clonally
related to the primary tumor, sharing many of the driver muta-
tions, but nonetheless have typically acquired additional variants
not detectable in the primary lesion (Brastianos et al., 2015; De
Mattos-Arruda et al., 2014; Ding et al., 2010; Hoadley et al.,
2016; Juric et al., 2015; Savas et al., 2016; Shah et al., 2009;
Yates et al., 2015). Due to small sample sizes, however, it has
proved difficult to extract general patterns of evolution between
primary and recurrence, leaving a number of unanswered ques-
tions with important biological and clinical implications. We con-
ducted this study to address some of these questions, including
how closely related a metastasis is to its primary lesion; whether
there are differences in evolution across locoregional relapse,
axillary metastases seeded by lymphatic spread, and distant
metastases seeded by hematogenous spread; whether the
driver landscape of metastases differs from primary cancers;
and whether there are cancer genes specific to metastases.
Since the survival of patients with metastatic breast cancer is
so poor, it is particularly important to establish whether newly
emerging driver mutations in the metastasis might offer opportu-
nities for personalized therapy.
RESULTS
Patient Cohort
The study comprises two major aims. In the first, to define pat-
terns of genomic evolution between the primary cancer and dis-
ease progression, we performed whole-genome sequencing of
40 tumor samples from 17 patients to an average coverage of
423, together with matched germline DNA samples (Tables S1
and S2). These 17 patients encompassed three clinical sce-
narios: synchronous axillary lymph node metastasis; distant
metastasis and local relapse subsequent to definitive treatment
for the primary tumor. In all but one case (PD11458), primary tu-
mor samples were treatment naive and sampled at diagnosis.
Metachronous recurrence samples were obtained 8–158 months
after the primary tumor diagnosis. Distant metastatic samples
were obtained from tumor deposits in lung (n = 1), liver (n = 1),
distant skin regions (n = 2), contralateral breast (n = 1), and
distant lymph nodes (n = 2) (Figure 1). All patients underwent
standard management, including curative surgery with local
radiotherapy, adjuvant anthracycline-containing chemotherapy,
and/or endocrine therapies where appropriate (Table S1).
The second aim was to study the distribution of driver muta-
tions in distant metastatic or locoregionally relapsed breast can-
cer. To achieve this, we analyzed 227 recurrence samples from
163 patients for point mutations and copy number changes in
365 known cancer genes to an average coverage of 4673
(Tables S1 and S2). For 46 patients, 2–5 recurrence samples
David Jones,
1
Keiran Raine,
1
Laura Mudie,
1
Calli Latimer,
1
Elinor Sawyer,
2,16
Christine Desmedt,
17
Christos Sotiriou,
17
Michael R. Stratton,
1
Anieta M. Sieuwerts,
18
Andy G. Lynch,
6
John W. Martens,
18
Andrea L. Richardson,
19,20
Andrew Tutt,
16,21,22
Per Eystein Lønning,
3,4,23,
*and Peter J. Campbell
1,23,24,
*
17
Breast Cancer Translational Research Laboratory, Universite
´Libre de Bruxelles, Institut Jules Bordet, Bd de Waterloo 121, 1000 Brussels,
Belgium
18
Erasmus MC Cancer Institute and Cancer Genomics Netherlands, Erasmus University Medical Center, Department of Medical Oncology,
Rotterdam, the Netherlands
19
Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115, USA
20
Dana-Farber Cancer Institute, Boston, MA 02215, USA
21
Breast Cancer Now Research Unit, King’s College London, London SE1 9RT, UK
22
The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
23
These authors contributed equally
24
Lead Contact
*Correspondence: per.eystein.lonning@helse-bergen.no (P.E.L.), pc8@sanger.ac.uk (P.J.C.)
http://dx.doi.org/10.1016/j.ccell.2017.07.005
170 Cancer Cell 32, 169–184, August 14, 2017
were sequenced, allowing heterogeneity of the recurrence land-
scape to be explored. For 51 individuals, the matched primary
tumor was available for sequencing.
Samples were obtained in clinically relevant scenarios
including first relapse or metastasis and following systemic treat-
ment interventions. All samples therefore represent clinically
progressing disease. Progression during a documented sys-
temic therapy exposure occurred for 126 samples, including
endocrine therapy (n = 43), anthracyclines (n = 45), taxanes
(n = 12), and other chemotherapeutic regimens. Samples were
obtained following a median of 2 systemic treatment exposures
(range, 0–5) and after 39 months (range, 0–196) from primary
cancer diagnosis (Table S1). Tumors were classified according
to the primary tumor TNM stage, histological type, grade, and
presence of estrogen receptor (ER), progesterone receptor
(PR), and ERBB2 (HER2) amplification.
Evolution between Primary Breast Cancer and
Metastasis/Relapse
Using whole-genome sequencing, we explored the patterns of
genomic evolution in three clinical scenarios across 17 patients:
local lymph node involvement at the time of primary tumor diag-
nosis (8 patients); locoregional relapse after apparently definitive
primary tumor treatment (n = 4); and subsequent development of
distant metastasis (n = 7) (Figure 1). We identified an average of
9,594 substitutions (range, 1,792–25,471), 1,098 indels (range,
60–12,786), and 245 structural variants (range, 6–786) within
each individual’s cancer genome (Table S3). We performed vali-
dation on 1,480 somatic substitutions and indels by custom cap-
ture pull-down or capillary sequencing (Table S4), confirming
1,436 (97%) were truly present and somatically acquired. We en-
riched our validation experiment with mutations that were private
to one of the samples to enhance our ability to identify subclonal
populations. Rearrangements were validated by the visual
confirmation of breakpoint-associated copy number changes.
To reconstruct the phylogenetic structure underlying disease
progression, we applied bioinformatic and deductive reasoning
approaches, as described previously (Nik-Zainal et al., 2012;
Yates et al., 2015)(Figures 1,S1, and S2A; Table S5). We used
multi-dimensional Bayesian Dirichlet processes to cluster so-
matic substitutions from multiple related samples according to
their respective mutation burden, corrected for tumor cellularity,
allele-specific copy number, and regions of differential chromo-
somal deletion between samples. We identified an average of 2.8
distinct clusters per patient (48 in 17 patients), with 94% of these
reproduced by independent clustering of high-coverage tar-
geted validation data (Figure S1 and Table S5). Individual clus-
ters inform on the structure of the phylogenetic tree, typically
Figure 1. Phylogenetic Trees Describe Evolution of 17 Primary Breast Cancers to Metastasis or Local Relapse
Each tree represents an individual patient’s breast cancer inferred from the analysis of a matched normal sample and 2–4 tumor samples per case (total of 40
tumor samples). Trees are derived from genome-wide substitutions. Trees are grouped according to scenario: distant metastasis (red panel), locoregional relapse
(blue panel), or synchronous axillary lymph node metastasis (green panel). Branches private to the metastasis or relapse follow the same color theme, while
branches representing clones that are specific to the primary tumor are gray. The black trunk represents clonal mutations that are present in 100% of cells in every
sample. Purple branches represent mutations within the metastasis or relapse that are subclonal within the primary tumor. Branch lengths reflect the proportio n of
clustered somatic mutations attributed to that subclone. The whole tree is scaled to the maximum length of a tree that would be inferred from mutations identified
in the primary tumor. Red circles identify the point of divergence between the metastasis/relapse-seeding clone and the primary tumor. The estimated whole-
genome doubling (WGD) time is indicated by 95% confidence intervals. Numbers in brackets reflect the months elapsed between primary tumo r and metastasis
sample acquisition.
See also Figures S1 and S2 and Tables S1,S2,S3,S4, and S5.
Cancer Cell 32, 169–184, August 14, 2017 171
ABC
D
E
(legend on next page)
172 Cancer Cell 32, 169–184, August 14, 2017
enabling a single ‘‘tree solution’’ to be derived for each case. In
16 of the 17 cases, all samples studied were clonally related,
as demonstrated by thousands of shared somatic mutations,
with the trunk of the phylogenetic tree representing 12%–
98% of all clustered somatic substitutions (Figures 1 and 2A,
Table S5).
One critical observation emerges: the genomic landscape of
the primary breast cancer at diagnosis is a good surrogate for
the somatic mutations present in disseminated cells at that
moment in time. This conclusion derives from several aspects
of the data. First, the metastatic or relapsing clones branch
late from the phylogenetic lineage of the primary breast lesion,
with relatively few mutations private to the primary tumor. On
average, metastatic divergence occurs at 87% of molecular
time within the primary tumor, estimated from phylogenetic anal-
ysis of base substitutions in regions of the genome with the same
copy number across all lesions (Figure 2A and Table S5). Sec-
ond, as expounded in more detail in the next section, the excess
mutational burden of metachronous metastasis or relapse
clones exceeded that of synchronous axillary lymph node me-
tastases (p = 0.02, F test) and the one synchronous distant
metastasis (PD11458) (Figure 2B). Indeed, synchronous lymph
node metastases are typically very similar to the primary breast
lesion (green branches, Figure 1). Third, driver mutations tend to
be concentrated on the trunk of the phylogenetic tree, notwith-
standing the 1 or 2 additional driver events acquired by relapse
or metastasis clones (explored in considerable detail in later
sections) (Figure 1). Furthermore, whole-genome duplication,
when present, precedes the branching of the recurrence clone
(Figures 1 and S2B). Finally, as we shall see, the mutational pro-
cesses active on the trunk of the phylogenetic tree tend to persist
in the metastasis, suggesting that inferences (and therapeutic
decisions) based on mutational signatures in the primary will
extend to the unseen disseminated cells.
One patient (PD8948), a germline BRCA1 mutation carrier,
was diagnosed with a triple-negative cancer of the left breast,
and over the next 10 years, treated for two apparent local re-
lapses of this lesion and a distant metastasis to the contralateral
breast. In fact, our genomic analyses revealed that the three le-
sions affecting the left breast were clonally unrelated, completely
independent primary cancers, with the second of them seeding
the contralateral breast metastasis (Figure S3). This is important
clinically as the management and prognosis of a second primary
cancer and a local relapse are distinct. This case demonstrates
that genome sequencing can clarify the nature of presumed local
‘‘relapses,’’ especially important in individuals with a genetic pre-
disposition to breast cancer.
Taken together, then, these patterns of disease evolution
strongly support the use of genome sequencing of the primary
breast cancer lesion to underpin decisions about systemic ther-
apy in the adjuvant setting. In modern breast cancer treatment,
the major aim of chemotherapy or estrogen suppression is to
kill those cells that have already spread from the primary lesion,
since surgery and local radiotherapy are usually sufficient to cure
the primary. If it were the case that relapsing or metastatic clones
disseminated early from the primary breast cancer with exten-
sive parallel evolution, as has been suggested previously (Klein,
2009), then targeting somatic mutations found in the primary
would not necessarily have much relevance to disseminated
cells without those changes.
Additional Burden of Mutations in Relapse Samples
For patients with synchronous axillary lymph node metastases,
the number of mutations private to the metastasis was broadly
equivalent to the number private to the primary cancer (Figures
1and 2B). This is perhaps not surprising since, by virtue of being
synchronous lesions, the major lineages in the primary and the
metastasis had the same time period in which to accrue muta-
tions after divergence. In contrast, for the local relapse and
metachronous distant metastasis samples, the relapse carried,
on average, 63% more mutations than the primary tumor,
albeit with considerable variability among patients (range,
24%–244% extra). The number of additional mutations in the
relapse only loosely correlated with the time elapsed between
diagnosis of the primary cancer and relapse (Pearson’s correla-
tion R = 0.29) (Figure 2C).
The additional mutation burden in the later relapse sample was
substantially greater than the chronological time elapsed be-
tween primary and metastasis would suggest, implying that the
rate at which mutations accumulate has typically increased dur-
ing breast cancer evolution. Strikingly, we find that the fraction of
additional substitutions, indels, and structural variants in the
Figure 2. Genome-wide Somatic Mutation Timing in 16 Breast Cancers
(A) For each of 17 primary tumor samples, the bar height reflects the point in molecular time that the recurrence seeding clone is estimated to diverge from the
primary tumor (relates to phylogenetic trees in Figure 1). Molecular time is determined from the number of base substitutions.
(B) The recurrence-specific mutation excess is reported in a barplot for each of 18 recurrence samples and in a boxplot split by synchronous (S) and meta-
chronous (M) cases, where the box represents the interquartile range (IQR) bisected by the median, whiskers represent the maximum and minimum range of the
data that do not exceed 1.53the IQR while outlier data points extend beyond this. The recurrence -specific mutation excess indicates the base substitution load
in branches private to the recurrence minus those in branches private to the primary tumor, presented as a percentage of all substitutions identified in the primary
tumor. The p value is generated by an F test.
(C) The recurrence-specific mutation excess as presented in (B) according to the time from primary tumor diagnosis and acquisition of the relapse sample, each
dot represents a patient. R = Pearson’s correlation coefficient.
(D) Scatterplots compare the proportion of each the major mutation types, indels (insertions and deletion), substitutions (Subs), and structural variants (SVs),
localized to the recurrence. Unlike (A) and (B), these figures include variants in regions that were variable in copy number across samples.
(E) Radiation mutation signature at relapse following external beam radiation. The mutation spectrum of an outlier sample (PD11461) highlighted by a dashed gray
circle in (D) is shown in detail. The overall contribution of indels and structural variants (SVs) outweighs that of substitutions at relapse (top left barplot). Wi thin this
sample, indels of greater lengths (bottom left barplot) and inversions and translocations (bottom, middle bar plot) are relatively more common after relapse.
Cohort-wide, the relative contribution of deletions as opposed to insertions (top right barplot) and of deletions of 5 base pairs (bp) or longer (bottom right barplot)
are reported.*p < 0.0001 (Fisher’s exact test) for enrichment in the relapse sample. Cases exposed to prior external beam radiotherapy are indicated by a star
symbol indicating that other samples do not seem to carry the same signature.
See also Figure S3.
Cancer Cell 32, 169–184, August 14, 2017 173
relapse sample compared with the primary tumor are broadly in
concert with one another (Pearson’s correlation R = 0.7–0.8; Fig-
ure 2D). One consequence of the continued structural variation is
that deletions of genomic regions add to the diversity of point
mutations between subclones.
One patient (dotted circle, Figure 2D), however, had distinctly
more indels in the relapse sample than would be suggested for
the number of additional base substitutions. This sample was
from a local relapse, occurring 2 years after a small, node-nega-
tive primary cancer treated with wide local excision and adjuvant
radiotherapy. More than 90% of the indels at relapse were dele-
tions rather than insertions, compared with <50% of indels on
the trunk of the phylogenetic tree for that patient (odds ratio
[OR] = 11.5; p = 1 310
21
; Fisher’s test) or 67% in all other pa-
tients’ cancers (OR = 3.4; p = 1 310
14
; Fisher’s test; Figure 2E).
The deletions occurring at relapse were typically longer than
those in the primary tumor, with 33% being 5–100 bp in size
versus 14% in the primary (OR = 3.1, p = 0.03; Fisher’s test; Fig-
ure 2E). We recently described the signature of small to medium-
sized deletions as a characteristic feature of radiation-induced
secondary cancers (Behjati et al., 2016). This suggests that in
this patient, the relapsing clone was exposed to adjuvant radio-
therapy and survived, albeit with genomic damage from the
ionizing radiation. In contrast, in other relapse samples from pa-
tients treated with adjuvant radiotherapy, this signature was not
evident (Figure 2E), perhaps suggesting that the cells that ulti-
mately seeded these relapses had already disseminated outside
the radiation field.
To assess which mutational signatures are most significant at
different stages of disease evolution, we examined their relative
contributions to each branch of the phylogenetic tree (Figure 3).
Perhaps the most striking feature is that the heterogeneity in
mutational signatures across patients is considerably greater
than the heterogeneity across different evolutionary stages
within a given tumor. This suggests that a given breast tumor ac-
cesses only a subset of the mutational processes potentially
available to it, but those mutational processes contribute
genomic variation on an ongoing basis. Nonetheless, there are
some shifts in the relative contributions of mutational processes
over time. The universal signature of C > T transitions at CpG
dinucleotides (signature 1) contributes a relatively higher propor-
tion of mutations early in disease evolution, likely because
this signature is relatively constant throughout life and gets
swamped by processes emerging later in disease evolution.
Mutations attributed to the activity of APOBEC enzymes, char-
acterized by C > T and C > G variants in a TpC context (signa-
tures 2 and 13), were rather variable in their timing, being
predominantly early in some patients (such as PD11461), more
prominent in late stages in others (PD9195), and relatively steady
in many (PD4243) (Figure 3). These patients had a range of
systemic cytotoxic treatments following their primary cancer
diagnosis, including anthracyclines, cyclophosphamide, and
5-fluorouracil; the lack of new signatures in relapsing lesions
suggests that these chemotherapeutic agents are not major
drivers of mutation accumulation.
Telomere Integrity during Cancer Evolution
We estimated telomere lengths from whole-genome data for
germline and tumor samples. Telomere lengths showed variation
among individuals and across samples from the same individual
(Figure S4 and Table S5). Greater variability of telomere lengths
was seen among tumors (mean = 7,703 bp; range, 2,409–
27,621 bp) compared with germline samples (mean =
6,627 bp, range, 4,351–11,077 bp) (Figure S4B). There was no
simple relationship between telomere length and the number of
somatic substitutions, indels, or structural variation within tumor
samples (Figure S4C).
In six of the eight cases where we sequenced breast tumors
and adjacent normal breast epithelium, the telomere was shorter
in the primary tumor, suggestive of telomere attrition during
cancer development. Between primary tumor and recurrence
samples within a patient, there was no consistent pattern, with
telomeres sometimes lengthening, sometimes shortening.
A few samples had especially long telomeres; one was associ-
ated with amplification of TERT (telomerase reverse transcrip-
tase) and another with amplification of TERC (telomerase RNA
template component).
Driver Mutations Are Acquired during Cancer
Progression
For each tumor, we manually curated the driver mutations
among the set of breast cancer genes known to be recurrently
targeted by point mutations (Kandoth et al., 2013; Lawrence
et al., 2014), structural variants, and copy number changes (Ber-
oukhim et al., 2010). We found that most driver mutations
occurred in the primary tumor and were located on the trunk of
the phylogenetic tree (Figure 1). Among the nine cancers that un-
derwent whole-genome duplication, all driver mutations arose
prior to this event, indicating that they are usually relatively early
events in cancer evolution.
Among the synchronous lymph node metastases, only one
patient had a driver mutation (in PTEN) seen in the metastasis
that was not present in the primary tumor, confirming that there
is generally little genomic divergence between primary and syn-
chronous local lymphatic metastases. In one case (PD11460),
we analyzed both a distant metastasis and a synchronous local
lymph node metastasis, finding that the lymph node deposit
was more closely related to the primary tumor than the subse-
quent distant metastasis and did not contain any private driver
mutations (Figure 1). This observation is consistent with the
highly divergent pattern recently reported between regional
lymph node metastases and brain metastases (Brastianos
et al., 2015).
Five of seven WGS-analysed patients with distant metasta-
ses, however, had one or two additional driver mutations
specific to the metastasis sample, suggesting that growth of
the metastatic clone in its new niche is abetted by further
genomic evolution. We observed several instances of com-
plex clusters of structural variants that were acquired late in
the major metastasis lineage. These included an event that
generated a complex amplification of CCND1 coupled with
loss of one copy of TP53 (Figure 4A) and a chromothripsis
(Stephens et al., 2011) event that resulted in FGFR1 amplifi-
cation (Figure 4B). Interestingly, these data showing complex,
catastrophic events during metastasis development echo
recent single-cell sequencing studies showing punctuated
copy number evolution in primary breast cancer lesions (Gao
et al., 2016).
174 Cancer Cell 32, 169–184, August 14, 2017
Figure 3. Genome-wide Mutation Signatures in Ten Metastatic or Locally Relapsed Breast Cancers Annotated to Phylogenetic Trees
The mutational signature composition of each phylogenetic tree branch is reported for the ten multi-sample, whole-genome cases with a local relapse or distant
metastatic sample. HRD, homologous recombination deficiency; MMR, mismatch-repair deficie ncy. See also Figure S4.
Cancer Cell 32, 169–184, August 14, 2017 175
A
BC
Figure 4. Structural Variant Driver Mutations at Relapse in Three Breast Cancers
(A) Case PD9193: De novo amplification of CCND1 in a distant lymph node metastasis. Structural variant breakpoints are represented by colored vertical lines:
interchromosomal translocations (gray arrows), tail-to-tail inversions (green), head-to-head inversions (blue), tandem duplications (orange), deletions (purple).
Rainfall plots report the inter-mutational distance of individual consecutive mutations where each dot reflects a mutation and the color represents the base
change.
(B) Case PD11460: de novo amplification of FGFR1 in a metastatic deposit.
(C) Case PD11461: a subclone containing a homozygous deletion in CDKN2A in the primary tumor seeds a local relapse.
176 Cancer Cell 32, 169–184, August 14, 2017
Of the three locoregional relapse cases, one relapse that
branched from the primary tumor particularly early in molecular
time acquired a new driver mutation in NCOR1. Another arose
from a subclone in the primary tumor that carried a homozygous
deletion of CDKN2A; this event became fully clonal in the relapse
(Figure 4C).
Thus, these data show that distant metastasis and locore-
gional relapses are typically associated with acquisition of addi-
tional driver mutations compared with the primary tumor,
whereas driver mutations in synchronous lymph node metasta-
ses are typically also present in the primary.
The Driver Landscape of Relapse and Metastasis
To provide more complete statements about the landscape of
driver mutations at breast cancer recurrence, we performed
sequencing of all coding exons of 365 known cancer genes in
227 samples from distant metastases or locoregional relapses
across 163 patients. The primary tumor was available for 51 of
these patients and germline DNA for 81. For comparison, we
also interrogated these genes from sequenced exomes of 705
primary breast cancers published by the TCGA, which we rean-
alyzed using the same pipeline as for our cohort (Table S3).
Samples that were from local relapses or metastases
harbored a higher number of driver point mutations on average
than those in the primary tumor cohort (2.0 versus 1.6;
p = 0.0008; F test). In 25 (49%) of the 51 patients from whom
we analyzed the matched primary tumor, a driver mutation was
found that was private to the relapse sample. This was more pro-
nounced for distant metastases; a driver mutation not found in
the primary lesion was seen in 74% of distant metastases
compared with 29% of locoregional relapses (p = 0.002, Fisher’s
test).
We compared the rate of non-synonymous mutations with
synonymous mutations across the 365 genes. This technique,
well established for inferring selection in comparative genetics,
was adapted for somatic mutations (Martincorena et al., 2015),
taking account of the trinucleotide composition of the genes,
gene size, mutation spectrum, and local variation in mutation
rates across the genome. A total of 21 and 20 cancer genes
were identified as significantly mutated (false discovery rate,
q < 0.1) in the primary and relapse cohorts, respectively, of which
15 genes were significant in both cohorts (Figure 5A). We note
that BRCA1 and NF1 were not significant in the primary cancer
cohort after correction for multiple hypothesis testing, something
we believe to be due to the play of chance given the wealth of
data implicating these two genes in primary breast cancer.
When split by whether tumors were ER-positive or ER-negative,
we found that most breast cancer genes showed higher rates of
driver mutation in the relapse/metastasis samples than in pri-
mary tumors (Figure 5B). The exception to this was PIK3CA
and MAP3K1 in ER-positive tumors, in keeping with reported
better relapse-free survival rates in primary breast cancers car-
rying PIK3CA mutations.
We formally tested whether each gene was significantly more
frequently mutated in relapsed or metastatic breast cancer than
primary breast cancer (Figure 5C). In general, ORs were skewed
toward greater enrichment in relapse or metastatic samples,
reflecting the greater number of driver mutations and the
wider repertoire of genes mutated. Significant differences for
individual genes were not detected among locoregional relapses
compared with distant metastases.
Driver Mutations Acquired Late Encompass a Wider
Range of Cancer Genes
There are two possible explanations for the enrichment of driver
mutations in relapse/metastasis samples compared with the
cohort of primary breast cancers. It might be that those primary
breast cancers with a more disordered genome are more likely to
subsequently relapse; or it might be that the relapsing clone con-
tinues to acquire new driver mutations after dissemination from
the primary lesion. We therefore compared the driver mutation
profile of the 51 patients in whom both the primary and a
relapse/metastasis sample were sequenced (Figures 6A–6C).
Mutations in well-known, relatively frequent breast cancer
genes, such as TP53,PIK3CA, and GATA3, when present,
were typically found in both the primary and the recurrence sam-
ples. In contrast, mutations in less frequent cancer genes were
often found only in the recurrence. This pattern was particularly
striking for genes involved in SWI/SNF signaling, such as
ARID1A,ARID1B, and ARID2, which were commonly wild-type
in the primary lesion but inactivated in the recurrence (Figures
6A, S5, and S6). This echoes recent data from metastatic endo-
metrial cancer (Gibson et al., 2016), locally progressive hepato-
cellular carcinoma (He et al., 2015), and a pan-cancer metastasis
study (Zehir et al., 2017), where mutations in these same genes
are also acquired late in disease evolution.
In primary breast cancer, ER-positive and triple-negative tu-
mors show rather distinct combinations of driver mutations,
with PIK3CA,GATA3, and MAPK-pathway mutations character-
izing the former and TP53 and copy number alterations the latter.
When studying relapse and metastasis samples, however, we
found that the genomic differences between triple-negative
and ER-positive cancers became more blurred: TP53 mutations
were seen in 40%–50% of relapsed ER-positive cases; and
PIK3CA,GATA3,CDH1, and MAP3K1 all increased several-
fold in relapsed ER-negative cancers. We identified ER and
PgR expression loss in 17% and 41% of cases, respectively,
across the relapsed breast cancer cohort (Figure 6C). Loss of
ER expression at relapse was frequently associated with driver
mutations in TP53 (90% of cases) and ARID1A (30% of cases).
While TP53 mutations were usually early events, detected in
the primary tumor, ARID1A was more often private to the relapse
sample in association with hormone receptor loss (Figures 6A,
6C, and S5).
Late JAK-STAT Pathway Inactivation
Interestingly, JAK2 and STAT3 were identified as significantly
mutated in the metastasis/relapse screen even though they
had not been discovered in the earlier (and larger) exome studies
of primary breast cancers (Banerji et al., 2012; Cancer Genome
Atlas Network, 2012; Ellis et al., 2012; Shah et al., 2012; Ste-
phens et al., 2012). Both showed an excess of protein-truncating
mutations, such as nonsense base substitutions, frameshift in-
dels, and essential splice site mutations (Figure 7A), suggesting
that they are operating as tumor suppressor genes in breast can-
cer. All such mutations in this cohort arose in ER-positive can-
cers in contrast to JAK2 amplifications that have been identified
in triple-negative cancers (Balko et al., 2016). One patient
Cancer Cell 32, 169–184, August 14, 2017 177
A
B
C
(legend on next page)
178 Cancer Cell 32, 169–184, August 14, 2017
showed an especially remarkable example of parallel evolution
of inactivating JAK2 mutations (Figure 7B). During this tumor’s
evolution, four different JAK2 inactivating mutations occurred,
all on subclonal branches of the phylogenetic tree, with several
of the lesions apparently having compound heterozygous inacti-
vation of the gene. This is reminiscent of the frequency of parallel
evolution of resistance to PARP inhibitors in ovarian cancers
through BRCA1/2 reversion mutations (Patch et al., 2015).
JAK2 and STAT3 mutations both showed a trend toward being
more frequent in distant metastasis samples (Figure 5C), which
may explain why these were detected as significant in our study
but not in previous studies of primary breast cancer. For
example, in one patient who had a local relapse followed by a
liver metastasis, the liver metastasis carried a STAT3 inactivating
mutation that was absent from both the primary cancer and the
local relapse, despite the latter being closely related to the
metastasis (Figure 7C).
Thus, inactivation of JAK-STAT signaling appears to con-
tribute to disease progression and metastasis in some patients
with breast cancer. We note that in another study of metastatic
breast cancer, a JAK2 nonsense mutation was also discovered
(Zehir et al., 2017). Interestingly, homozygous loss of JAK2 has
recently been described as a mechanism of resistance to check-
point inhibitor immunotherapies (Zaretsky et al., 2016), probably
acting through blocking the interferon-gamma pathway.
Although none of the patients here received such therapies, it
is feasible that these mutations help advanced tumors evade
the native immune response mounted against them. Cancers
with JAK2 or STAT3 truncating mutations contained a higher
number of point mutations on average than other cancers
(p = 3 310
9
; F test, Figure 7D). Although other explanations
are possible, this finding would be consistent with the notion
that these cancers may contain more neoantigens, stimulating
a more exuberant native immune response, and driving selection
of JAK-STAT pathway inactivation.
Treatment Exposures Influence Breast Cancer
Evolution
The broadening of the repertoire of cancer genes sampled by
late driver mutations likely reflects the diverse selective forces
operating during evolution of advanced breast cancer. These
include selective pressures exerted by therapeutic interventions,
by the immune system responding to the expansion of a clone
carrying many neoantigens, and by the very different microenvi-
ronment in a metastatic site compared with breast epithelium.
A total of 139 samples of recurrent disease were taken shortly
before a systemic treatment was commenced (Figure S6A), of
which 59 displayed progressive disease, indicating treatment
resistance, and 80 cancers stabilized or responded to treatment
(Table S1). Across all treatments, TP53 and ESR1 driver muta-
tions were more frequent in cases that progressed (63% in
progression cases versus 45% in stable disease, p = 0.04; and
7% versus 0%, p = 0.03 respectively; Fisher’s exact test), as
seen in a recently published series of metastatic breast cancers
(Zehir et al., 2017). Mutant TP53 has previously been associated
with endocrine and anthracycline resistance (Aas et al., 1996;
Berns et al., 1998). Gain-of-function mutations in TP53 have
been associated with metastasis and drug resistance in cell-
line and xenograft models (Petitjean et al., 2007; Turner et al.,
2017), but in our cohort loss-of-function and gain-of-function
mutations were equally enriched in patients with progressive dis-
ease compared with stable disease (p = 0.5; Fisher’s exact test)
(Figure S6B), and in recurrences compared with primary tumors
(p = 0.7; Fisher’s exact test) (Figure S6C). As previously reported,
ESR1 resistance mutations were found in five patients previously
treated with endocrine therapies (Chandarlapaty et al., 2016;
Robinson et al., 2013; Toy et al., 2013) and predicted progressive
disease upon switching treatment (Figure S5).
Truncating mutations in SWI-SNF cancer genes, including
ARID1A and ARID2, emerged in three of five cancers relapsing
after taxane chemotherapy (Figure S6D). Interestingly, an asso-
ciation between loss of ARID1A expression and chemoresist-
ance has also been observed in clear-cell ovarian cancers
(Katagiri et al., 2012).
A handful of potentially actionable driver mutations emerged
during endocrine therapy (Figures S5 and S6D). These included
amplifications of the MDM4, FGFR1, and CCND1 oncogenes
in two patients each, with an additional patient acquiring a ca-
nonical BRAF V600E mutation. FGFR1 activation and TP53
pathway inactivation (including MDM2/4 activation) have previ-
ously been associated with endocrine resistance (Ellis et al.,
2012; Turner et al., 2010). The implication here is that oncogene
amplification or activation may represent a common mode of
breast cancer evolution in the face of endocrine therapy. Since
oncogenes are more natural therapeutic targets than tumor sup-
pressor genes, this raises the interesting possibility of new
personalized interventions for some patients relapsing after
endocrine therapy.
DISCUSSION
The concept of precision oncology is founded on the presump-
tion that knowing the genomic basis of a patient’s cancer will
guide choice of targeted therapies likely to be efficacious. This
rests on the key assumption that we can obtain a sample repre-
sentative of the tumor cells that we are targeting with that ther-
apy. In patients diagnosed with primary breast cancer, systemic
therapy is aimed at killing the microscopic deposits of cells that
have disseminated from the breast, as surgery and radiotherapy
will generally cure the primary lesion. In the samples studied
Figure 5. Comparison of the Driver Landscapes of 163 Recurrent and 705 Primary Breast Cancers
(A) Cancer genes identified as significantly mutated with a false discovery rate (q) < 0.1, applied to the TCGA 705 primary breast cancer exomes or the 163
recurrent breast cancers independently.
(B) Barplots compare the prevalence of each significantly mutated cancer gene and ESR1 in the primary and recurrent breast cancer cohorts (662 and 151 cases,
respectively, where the estrogen receptor status of the primary tumor was documented).
(C) Forest plot comparing the frequency with which cancer genes are mutated in the relapse cancer cohort (163 cases) compared with the primary tumor cohort
(705 cases). Enrichment for each gene was determined using two-sided Fisher’s exact tests and Benjamini and Hochberg correction. Box size is scaled to the
number of cases and whiskers, and numbers inside brackets represent the 95% confidence interval for the odds ratio (the upper limit is clipped at 1,000).
Cancer Cell 32, 169–184, August 14, 2017 179
here, we found that at the time of initial diagnosis, the genome of
the primary would have been a good proxy for that of the cells
that ultimately seeded the relapse, whether the spread be local
or via a hematogenous or lymphatic route. In particular, the
vast majority of driver mutations found in the primary cancer
would also be present in the relapsing clone. Our observation
of late dissemination is consistent with the findings of a recent
study that combined bulk sequencing of primary tumors with sin-
gle-cell sequencing of bone marrow-derived disseminated tu-
mor cells, the presumed precursor of clinically overt metastatic
A
C
B
Figure 6. Temporal Distribution of Mutated Cancer Genes in 51 Paired Primary Tumor and Relapse Samples
(A) The heatmap indicates if the driver mutation is early (blue), defined as present in both the primary tumor and recurrence, or late, being detected in the
recurrence deposit(s) only (orange), or different mutations in the same gene seen in both the primary and recurrence (purple). Asterisks (*) indicate cancer genes
mutated in >5% of 705 primary tumor samples. The pie charts compare the proportion of mutations that are private to recurrence samples within most commonly
mutated genes and within comparatively rare cancer genes (mutated in <5% of primary tumors). Stacked barplot above the heatmap relates cumulative incidence
of point mutations and amplifications in (C) for each individual patient.
(B) Temporal ordering of amplified oncogenes derived from analysis of next-generation sequencing data. Tile colors follow the format stated in (A).
(C) Blue and pink tiles indicate the immunohistochemical (IHC) classification by estrogen receptor (ER) and progesterone receptor (PgR) of primary and relapse
samples, where a split tile indicates multiple relapse samples with different ER/PgR statuses.
See also Figures S5 and S6.
180 Cancer Cell 32, 169–184, August 14, 2017
B
C D
A
(legend on next page)
Cancer Cell 32, 169–184, August 14, 2017 181
disease (Demeulemeester et al., 2016). Although the genome of
a metastatic clone may be similar to the primary tumor at first
diagnosis, by the time it has expanded to be clinically detectable,
extensive further genomic changes have occurred.
Whether patients presenting with distant metastatic disease
should have that metastasis biopsied or not to decide on thera-
peutic interventions is a controversial question (Arnedos et al.,
2015). Many sites of metastatic disease are challenging and
invasive to sample, demonstrated by the bias seen in our cohort
toward sites of disease that are easy to access. Our data indicate
that metastases seeded by hematogenous spread do continue
to evolve after dissemination, acquiring many new somatic mu-
tations and key driver mutations. A recent study from a large ter-
tiary referral unit has shown that many patients with metastatic
breast cancer are willing to undergo biopsy of recurrent lesions
for molecular profiling (Zehir et al., 2017).
In its restless search for a genome ideally suited to autono-
mous life in far-flung regions of the body, a breast cancer can ac-
cess many different mutational processes and a wide repertoire
of cancer genes. The result is considerable patient-to-patient
variability in genomic profiles, even more pronounced than the
already daunting levels seen in primary breast cancer. Mapping
this complexity will require recruitment of large, prospective co-
horts of patients with metastatic disease and integration with
transcriptional, epigenomic, and clinical readouts. Our data
show that such an endeavor would have potential clinical
impact, providing insights into patterns of clonal evolution,
mechanisms of therapy failure, and pathways that could repre-
sent new therapeutic targets.
STAR+METHODS
Detailed methods are provided in the online version of this paper
and include the following:
dKEY RESOURCES TABLE
dCONTACT FOR REAGENT AND RESOURCE SHARING
dEXPERIMENTAL MODEL AND SUBJECT DETAILS
BSubjects, Samples and Consent
BWhole Genome ‘Triplet’ Cohort
BRelapsed Breast Cancer Cohort
BPrimary Breast Cancer (Comparison) Cohort
dMETHOD DETAILS
BSample Size
BTumor Specimen Processing
BDNA Extraction
BMulti-Sample Whole Genome Sequencing
BMulti-Region Targeted Gene Screen
BMulti-Sample Mutation Calling
BMutation Validation
BCancer Gene Discovery
BDriver Mutation Annotation
BGenome-Wide Subclonal Copy Number Analyses
BGenome-Wide Multi-Sample Clonality Analyses
BMutation Timing in Multi-Sample Analyses
BWhole Genome Duplication Timing Analysis
BDriver Mutation Enrichment Analyses
BMutational Signature Analysis
BTelomere Length Estimates
BCase PD8948 and Whole Genome Sequencing and
Analysis of an FFPE Sample
dQUANTIFICATION AND STATISTICAL ANALYSIS
dDATA AND SOFTWARE AVAILABILITY
SUPPLEMENTAL INFORMATION
Supplemental Information includes six figures and five tables and can be found
with this article online at http://dx.doi.org/10.1016/j.ccell.2017.07.005.
AUTHOR CONTRIBUTIONS
Conceptualization, L.R.Y., P.J.C., P.E.L., S.K., and A.T.; Methodology, L.R.Y.,
P.J.C., P.E.L., S.K., D.W., I.M., L.B.A., P.V.L., M.G., S.G., M.R.S., E.S., E.P.,
A.G.L., and J.H.R.F.; Software, D.W., I.M., L.B.A., P.V.L., L.R.Y., G.G., M.G.,
S.G.B., K.R., D.J., A.G.L., and J.H.R.F.; Histopathological analysis, H.K.H.,
P.K.L.; Validation, L.R.Y., S.K., L.M., C.L.; Resources, A.M.S., J.W.M.,
A.L.R., P.E.L., C.S., C.D., P.G., L.R.Y., and A.T.; Data curation, L.R.Y., S.K.,
P.E.L.; Visualization, L.R.Y.; Writing – Original Draft, L.R.Y. and P.J.C. Writing –
Review and Editing, all authors; Supervision, P.J.C. and P.E.L.
ACKNOWLEDGMENTS
This work was supported by the Wellcome Trust. Work within the project was
also supported by the Bergen Research Foundation, the Norwegian Cancer
Society, the Norwegian Research Council, Belgian Cancer Plan-Ministry of
Health, the Breast Cancer Research Foundation and the Brussels Region,
The University of Cambridge, Cancer Research UK, and Hutchison Whampoa
Limited. L.R.Y. is funded by a Wellcome Trust research fellowship. P.J.C. is a
Wellcome Trust Senior Clinical Fellow. Dr. Papaemmanuil is a Josi e Robertson
Investigator and the recipient of a European Hematology Association early
career fellowship. A.M.S was supported by Cancer Genomics Netherlands
(CGC.nl) through a grant from the Netherlands Organisation for Scientific
Research (NWO). A.G.L. and J.H.R.F. were supported by a Cancer Research
UK Program Grant to Simon Tavare
´(C14303/A17197) We thank Beryl Leirvaag
and Dagfinn Ekse of Haukeland University Hospital, Bergen, Norway, for tech-
nical assistance. We also acknowledge and thank all members of the labora-
tory and IT teams within the Cancer Genome Project at the Wellcome Trust
Sanger Institute. The laboratory work performed in Bergen was conducted
in the Mohn Cancer Research Laboratory. This research used resources pro-
vided by the Los Alamos National Laboratory Institutional Computing Pro-
gram, which is supported by the U.S. Department of Energy National Nuclear
Security Administration under contract no. DE-AC52-06NA25396. Research
performed at Los Alamos National Laboratory was carried out under the aus-
pices of the National Nuclear Security Administration of the United States
Department of Energy. This work was supported by the Francis Crick Institute,
Figure 7. JAK-STAT Inactivating Mutations Are Enriched at Relapse
(A) Pencil plots of JAK2 and STAT3 genes annotated with non-synonymous mutations identified in the relapse cohort (n = 163) and the primary cohort (n = 705).
(B) A case (PD8709) of parallel evolution involving four truncating mutations in JAK2. Response to treatment exposures are documented. SD, stable disea se; PR,
partial response; PD, progressive disease.
(C) A case (PD8727) of STAT3 truncating mutation arising in a liver metastasis.
(D) Scatter and boxplot of the number of mutations identified in samples (within the relapse cohort, 163 cases) that do or do not contain a JAK2 or STAT3
truncating mutation. The box represents the interquartile range (IQR) bisected by the median, whiskers represent the maximum and minimum range of the data
that do not exceed 1.53the IQR. p value generated using an F test.
182 Cancer Cell 32, 169–184, August 14, 2017
which receives its core funding from Cancer Research UK (FC001202), the UK
Medical Research Council (FC001202), and the Wellcome Trust (FC001202).
The results published here use data generated by the TCGA Research
Network: http://cancergenome.nih.gov/.
Received: December 21, 2016
Revised: May 13, 2017
Accepted: July 14, 2017
Published: August 14, 2017
REFERENCES
Aas, T., Borresen, A.L., Geisler, S., Smith-Sorensen, B., Johnsen, H., Varhaug,
J.E., Akslen, L.A., and Lonning, P.E. (1996). Specific P53 mutations are asso-
ciated with de novo resistance to doxorubicin in breast cancer patients. Nat.
Med. 2, 811–814.
Alexandrov, L.B., Nik-Zainal, S., Wedge, D.C., Aparicio, S.A., Behjati, S.,
Biankin, A.V., Bignell, G.R., Bolli, N., Borg, A., Borresen-Dale, A.L., et al.
(2013a). Signatures of mutational processes in human cancer. Nature 500,
415–421.
Alexandrov, L.B., Nik-Zainal, S., Wedge, D.C., Campbell, P.J., and Stratton,
M.R. (2013b). Deciphering signatures of mutational processes operative in hu-
man cancer. Cell Rep. 3, 246–259.
Arnedos, M., Vicier, C., Loi, S., Lefebvre, C., Michiels, S., Bonnefoi, H., and
Andre, F. (2015). Precision medicine for metastatic breast cancer–limitations
and solutions. Nat. Rev. Clin. Oncol. 12, 693–704.
Balko, J.M., Giltnane, J.M., Wang, K., Schwarz, L.J., Young, C.D., Cook, R.S.,
Owens, P., Sanders, M.E., Kuba, M.G., Sanchez, V., et al. (2014). Molecular
profiling of the residual disease of triple-negative breast cancers after neoad-
juvant chemotherapy identifies actionable therapeutic targets. Cancer Discov.
4, 232–245.
Balko, J.M., Schwarz, L.J., Luo, N., Estrada, M.V., Giltnane, J.M., Davila-
Gonzalez, D., Wang, K., Sanchez, V., Dean, P.T., Combs, S.E., et al. (2016).
Triple-negative breast cancers with amplification of JAK2 at the 9p24 locus
demonstrate JAK2-specific dependence. Sci. Transl. Med. 8, 334ra53.
Banerji, S., Cibulskis, K., Rangel-Escareno, C., Brown, K.K., Carter, S.L.,
Frederick, A.M., Lawrence, M.S., Sivachenko, A.Y., Sougnez, C., Zou, L.,
et al. (2012). Sequence analysis of mutations and translocations across breast
cancer subtypes. Nature 486, 405–409.
Behjati, S., Gundem, G., Wedge, D.C., Roberts, N.D., Tarpey, P.S., Cooke,
S.L., Van Loo, P., Alexandrov, L.B., Ramakrishna, M., Davies, H., et al.
(2016). Mutational signatures of ionizing radiation in second malignancies.
Nat. Commun. 7, 12605.
Berns, E.M., Klijn, J.G., van Putten, W.L., de Witte, H.H., Look, M.P., Meijer-
van Gelder, M.E., Willman, K., Portengen, H., et al. (1998). p53 protein
accumulation predicts poor response to tamoxifen therapy of patients with
recurrent breast cancer. J. Clin. Oncol. 16, 121–127.
Beroukhim, R., Mermel, C.H., Porter, D., Wei, G., Raychaudhuri, S., Donovan,
J., Barretina, J., Boehm, J.S., Dobson, J., Urashima, M., et al. (2010). The land-
scape of somatic copy-number alteration across human cancers. Nature 463,
899–905.
Brastianos, P.K., Carter, S.L., Santagata, S., Cahill, D.P., Taylor-Weiner, A.,
Jones, R.T., Van Allen, E.M., Lawrence, M.S., Horowitz, P.M., Cibulskis, K.,
et al. (2015). Genomic characterization of brain metastases reveals branched
evolution and potential therapeutic targets. Cancer Discov. 5, 1164–1177.
Brown, A.M. (2017). Phylogenetic analysis of metastatic progression in breast
cancer using somatic mutations and copy number aberrations. Nat. Commun.
8, 14944.
Cancer Genome Atlas Network. (2012). Comprehensive molecular portraits of
human breast tumours. Nature 490, 61–70.
Chandarlapaty, S., Chen, D., He, W., Sung, P., Samoila, A., You, D., Bhatt, T.,
Patel, P., Voi, M., Gnant, M., et al. (2016). Prevalence of ESR1 mutations in cell-
free DNA and outcomes in metastatic breast cancer: a secondary analysis of
the BOLERO-2 clinical trial. JAMA Oncol. 2, 1310–1315.
Ciriello, G., Gatza, M.L., Beck, A.H., Wilkerson, M.D., Rhie, S.K., Pastore, A.,
Zhang, H., McLellan, M., Yau, C., Kandoth, C., et al. (2015). Comprehensive
molecular portraits of invasive lobular breast cancer. Cell 163, 506–519.
Costello, M., Pugh, T.J., Fennell, T.J., Stewart, C., Lichtenstein, L., Meldrim,
J.C., Fostel, J.L., Friedrich, D.C., Perrin, D., Dionne, D., et al. (2013).
Discovery and characterization of artifactual mutations in deep coverage tar-
geted capture sequencing data due to oxidative DNA damage during sample
preparation. Nucleic Acids Res. 41, e67.
De Mattos-Arruda, L., Weigelt, B., Cortes, J., Won, H.H., Ng, C.K., Nuc iforo, P.,
Bidard, F.C., Aura, C., Saura, C., Peg, V., et al. (2014). Capturing intra-tumor
genetic heterogeneity by de novo mutation profiling of circulating cell-free tu-
mor DNA: a proof-of-principle. Ann. Oncol. 25, 1729–1735.
Demeulemeester, J., Kumar, P., Moller, E.K., Nord, S., Wedge, D.C., Peterson,
A., Mathiesen, R.R., Fjelldal, R., Zamani Esteki, M., Theunis, K., et al. (2016).
Tracing the origin of disseminated tumor cells in breast cancer using single-
cell sequencing. Genome Biol. 17, 250.
Ding, L., Ellis, M.J., Li, S., Larson, D.E., Chen, K., Wallis, J.W., Harris, C.C.,
McLellan, M.D., Fulton, R.S., Fulton, L.L., et al. (2010). Genome remodelling
in a basal-like breast cancer metastasis and xenograft. Nature 464, 999–1005.
Ellis, M.J., Ding, L., Shen, D., Luo, J., Suman, V.J., Wallis, J.W., Van Tine, B.A.,
Hoog, J., Goiffon, R.J., Goldstein, T.C., et al. (2012). Whole-genome analysis
informs breast cancer response to aromatase inhibition. Nature 486, 353–360 .
Futreal, P.A., Coin, L., Marshall, M., Down, T., Hubbard, T., Wooster, R.,
Rahman, N., and Stratton, M.R. (2004). A census of human cancer genes.
Nat. Rev. 4, 177–183.
Gao, R., Davis, A., McDonald, T.O., Sei, E., Shi, X., Wang, Y., Tsai, P.C.,
Casasent, A., Waters, J., Zhang, H., et al. (2016). Punctuated copy number
evolution and clonal stasis in triple-negative breast cancer. Nat. Genet. 48,
1119–1130.
Gellert, P., Segal, C.V., Gao, Q., Lopez-Knowles, E., Martin, L.A., Dodson, A.,
Li, T., Miller, C.A., Lu, C., Mardis, E.R., et al. (2016). Impact of mutational pro-
files on response of primary oestrogen receptor-positive breast cancers to
oestrogen deprivation. Nat. Commun. 7, 13294.
Gibson, W.J., Hoivik, E.A., Halle, M.K., Taylor-Weiner, A., Cherniack, A.D.,
Berg, A., Holst, F., Zack, T.I., Werner, H.M., Staby, K.M., et al. (2016). The
genomic landscape and evolution of endometrial carcinoma progression
and abdominopelvic metastasis. Nat. Genet. 48, 848–855.
Harris, L.N., Ismaila, N., McShane, L.M., Andre, F., Collyar, D.E., Gonzalez-
Angulo, A.M., Hammond, E.H., Kuderer, N.M., Liu, M.C., Mennel, R.G., et al.
(2016). Use of biomarkers to guide decisions on adjuvant systemic therapy
for women with early-stage invasive breast cancer: American Society of
Clinical Oncology clinical practice guideline. J. Clin. Oncol. 34, 1134–1150.
He, F., Li, J., Xu, J.F., Zhang, S., Xu, Y.P., Zhao, W.X., Yin, Z.Y., and Wang,
X.M. (2015). Decreased expression of ARID1A associates with poor prognosis
and promotes metastases of hepatocellular carcinoma. J. Exp. Clin. Cancer
Res. 34, 47.
Hoadley, K.A., Siegel, M.B., Kanchi, K.L., Miller, C.A., Ding, L., Zhao, W., He,
X., Parker, J.S., Wendl, M.C., Fulton, R.S., et al. (2016). Tumor evolution in two
patients with basal-like breast cancer: a retrospective genomics study of mul-
tiple metastases. PLoS Med. 13, e1002174.
Howie, B.N., Donnelly, P., and Marchini, J. (2009). A flexible and accurate ge-
notype imputation method for the next generation of genome-wide association
studies. PLoS Genet. 5, e1000529.
Juric, D., Castel, P., Griffith, M., Griffith, O.L., Won, H.H., Ellis, H., Ebbesen,
S.H., Ainscough, B.J., Ramu, A., Iyer, G., et al. (2015). Convergent loss of
PTEN leads to clinical resistance to a PI(3)Kalpha inhibitor. Nature 518,
240–244.
Kandoth, C., McLellan, M.D., Vandin, F., Ye, K., Niu, B., Lu, C., Xie, M., Zhang,
Q., McMichael, J.F., Wyczalkowski, M.A., et al. (2013). Mutational landscape
and significance across 12 major cancer types. Nature 502, 333–339.
Katagiri, A., Nakayama, K., Rahman, M.T., Rahman, M., Katagiri, H.,
Nakayama, N., Ishikawa, M., Ishibashi, T., Iida, K., Kobayashi, H., et al.
(2012). Loss of ARID1A expression is related to shorter progression-free
Cancer Cell 32, 169–184, August 14, 2017 183
survival and chemoresistance in ovarian clear cell carcinoma. Mod. Pathol. 25,
282–288.
Klein, C.A. (2009). Parallel progression of primary tumours and metastases.
Nat. Rev. Cancer 9, 302–312.
Lawrence, M.S., Stojanov, P., Mermel, C.H., Robinson, J.T., Garraway, L.A.,
Golub, T.R., Meyerson, M., Gabriel, S.B., Lander, E.S., and Getz, G. (2014).
Discovery and saturation analysis of cancer genes across 21 tumour types.
Nature 505, 495–501.
Martincorena, I., Roshan, A., Gerstung, M., Ellis, P., Van Loo, P., McLaren, S.,
Wedge, D.C., Fullam, A., Alexandrov, L.B., Tubio, J.M., et al. (2015). Tumor
evolution. High burden and pervasive positive selection of somatic mutations
in normal human skin. Science 348, 880–886.
Miller, C.A., Gindin, Y., Lu, C., Griffith, O.L., Griffith, M., Shen, D., Hoog, J., Li,
T., Larson, D.E., Watson, M., et al. (2016). Aromatase inhibition remodels
the clonal architecture of estrogen-receptor-positive breast cancers. Nat.
Commun. 7, 12498.
Ng, C.K., Martelotto, L.G., Gauthier, A., Wen, H.C., Piscuoglio, S., Lim, R.S.,
Cowell, C.F., Wilkerson, P.M., Wai, P., Rodrigues, D.N., et al. (2015). Intra-tu-
mor genetic heterogeneity and alternative driver genetic alterations in breast
cancers with heterogeneous HER2 gene amplification. Genome Biol. 16, 107.
Nik-Zainal, S., Davies, H., Staaf, J., Ramakrishna, M., Glodzik, D., Zou, X.,
Martincorena, I., Alexandrov, L.B., Martin, S., Wedge, D.C., et al. (2016).
Landscape of somatic mutations in 560 breast cancer whole-genome se-
quences. Nature 534, 47–54.
Nik-Zainal, S., Van Loo, P., Wedge, D.C., Alexandrov, L.B., Greenman, C.D.,
Lau, K.W., Raine, K., Jones, D., Marshall, J., Ramakrishna, M., et al. (2012).
The life history of 21 breast cancers. Cell 149, 994–1007.
Patch, A.M., Christie, E.L., Etemadmoghadam, D., Garsed, D.W., George, J.,
Fereday, S., Nones, K., Cowin, P., Alsop, K., Bailey, P.J., et al. (2015). Whole-
genome characterization of chemoresistant ovarian cancer. Nature 521,
489–494.
Petitjean, A., Mathe, E., Kato, S., Ishioka, C., Tavtigian, S.V., Hainaut, P., and
Olivier, M. (2007). Impact of mutant p53 functional properties on TP53 muta-
tion patterns and tumor phenotype: lessons from recent developments in
the IARC TP53 database. Hum. Mutat. 28, 622–629.
Purdom, E., Ho, C., Grasso, C.S., Quist, M.J., Cho, R.J., and Spellman, P.
(2013). Methods and challenges in timing chromosomal abnormalities within
cancer samples. Bioinformatics 29, 3113–3120.
Robinson, D.R., Wu, Y.M., Vats, P., Su, F., Lonigro, R.J., Cao, X., Kalyana-
Sundaram, S., Wang, R., Ning, Y., Hodges, L., et al. (2013). Activating ESR1
mutations in hormone-resistant metastatic breast cancer. Nat. Genet. 45,
1446–1451.
Savas, P., Teo, Z.L., Lefevre, C., Flensburg, C., Caramia, F., Alsop, K.,
Mansour, M., Francis, P.A., Thorne, H.A., Silva, M.J., et al. (2016). The subclo-
nal architecture of metastatic breast cancer: results from a prospective com-
munity-based rapid autopsy program ‘‘CASCADE’’. PLoS Med. 13, e1002204.
Shah, S.P., Morin, R.D., Khattra, J., Prentice, L., Pugh, T., Burleigh, A.,
Delaney, A., Gelmon, K., Guliany, R., Senz, J., et al. (2009). Mutational evolu-
tion in a lobular breast tumour profiled at single nucleotide resolut ion. Nature
461, 809–813.
Shah, S.P., Roth, A., Goya, R., Oloumi, A., Ha, G., Zhao, Y., Turashvili, G., Ding,
J., Tse, K., Haffari, G., et al. (2012). The clonal and mutational evolution spec-
trum of primary triple-negative breast cancers. Nature 486, 395–399.
Stephens, P.J., Greenman, C.D., Fu, B., Yang, F., Bignell, G.R., Mudie, L.J.,
Pleasance, E.D., Lau, K.W., Beare, D., Stebbings, L.A., et al. (2011). Massive
genomic rearrangement acquired in a single catastrophic event during cancer
development. Cell 144, 27–40.
Stephens, P.J., Tarpey, P.S., Davies, H., Van Loo, P., Greenman, C., Wedge,
D.C., Nik-Zainal, S., Martin, S., Varela, I., Bignell, G.R., et al. (2012). The land-
scape of cancer genes and mutational processes in breast cancer. Nature 486,
400–404.
Tevaarwerk, A.J., Gray, R.J., Schneider, B.P., Smith, M.L., Wagner, L.I.,
Fetting, J.H., Davidson, N., Goldstein, L.J., Miller, K.D., and Sparano, J.A.
(2013). Survival in patients with metastatic recurrent breast cancer after adju-
vant chemotherapy: little evidence of improvement over the past 30 years.
Cancer 119, 1140–1148.
Toy, W., Shen, Y., Won, H., Green, B., Sakr, R.A., Will, M., Li, Z., Gala, K.,
Fanning, S., King, T.A., et al. (2013). ESR1 ligand-binding domain mutations
in hormone-resistant breast cancer. Nat. Genet. 45, 1439–1445.
Turner, K.M., Deshpande, V., Beyter, D., Koga, T., Rusert, J., Lee, C., Li, B.,
Arden, K., Ren, B., Nathanson, D.A., et al. (2017). Extrachromosomal onco-
gene amplification drives tumour evolution and genetic heterogeneity.
Nature 543, 122–125.
Turner, N., Pearson, A., Sharpe, R., Lambros, M., Geyer, F., Lopez-Garcia,
M.A., Natrajan, R., Marchio, C., Iorns, E., Mackay, A., et al. (2010). FGFR1
amplification drives endocrine therapy resistance and is a therapeutic target
in breast cancer. Cancer Res. 70, 2085–2094.
Van Loo, P., Nordgard, S.H., Lingjaerde, O.C., Russnes, H.G., Rye, I.H., Sun,
W., Weigman, V.J., Marynen, P., Zetterberg, A., Naume, B., et al. (2010). Allele-
specific copy number analysis of tumors. Proc. Natl. Acad. Sci. USA 107,
16910–16915.
Wang, Y., Waters, J., Leung, M.L., Unruh, A., Roh, W., Shi, X., Chen, K.,
Scheet, P., Vattathil, S., Liang, H., et al. (2014). Clonal evolution in breast can-
cer revealed by single nucleus genome sequencing. Nature 512, 155–160.
Yates, L.R., Gerstung, M., Knappskog, S., Desmedt, C., Gundem, G., Van Loo,
P., Aas, T., Alexandrov, L.B., Larsimont, D., Davies, H., et al. (2015). Subclonal
diversification of primary breast cancer revealed by multiregion sequencing.
Nat. Med. 21, 751–759.
Zaretsky, J.M., Garcia-Diaz, A., Shin, D.S., Escuin-Ordinas, H., Hugo, W., Hu-
Lieskovan, S., Torrejon, D.Y., Abril-Rodriguez, G., Sandoval, S., Barthly, L.,
et al. (2016). Mutations associated with acquired resistance to PD-1 blockade
in melanoma. N. Engl. J. Med. 375, 819–829.
Zehir, A., Benayed, R., Shah, R.H., Syed, A., Middha, S., Kim, H.R., Srinivasan,
P., Gao, J., Chakravarty, D., Devlin, S.M., et al. (2017). Mutational landscape of
metastatic cancer revealed from prospective clinical sequencing of 10,000 pa-
tients. Nat. Med. 23, 703–713.
184 Cancer Cell 32, 169–184, August 14, 2017
STAR+METHODS
KEY RESOURCES TABLE
CONTACT FOR REAGENT AND RESOURCE SHARING
Further information and requests for resources and reagents should be directed to and will be fulfilled by the Lead Contact, Peter J
Campbell (pc8@sanger.ac.uk).
EXPERIMENTAL MODEL AND SUBJECT DETAILS
Subjects, Samples and Consent
All samples included in this project were obtained with informed patient consent and handled in line with the wider framework and
approval for the Breast Cancer Genome Analyses for the International Cancer Genome Consortium Working Group led by the Well-
come Trust Sanger Institute, Cambridgeshire, UK, REC reference: 09/H0306/36. We performed MPS and analysis of a total of 299
tumor samples collected from 170 individual’s breast cancers and 87 matched normal, germline samples (Table S1). Three patients
were male and the remainder female, the average age at primary tumor diagnosis was 53 years (range 30-85 years). Clinical details
including tumor stage, histological features and hormone receptor status are summarized in Table S1. Clinical follow-up data was
available for 96% of patients. The cohort reflects a very poor prognostic group of patients whereby 96% of these patients were diag-
nosed during their disease course witheither distant metastatic disease (86%), very poorly controlled locoregional disease not
amenable to surgical resection (10% of cases) or both (7%).
Whole Genome ‘Triplet’ Cohort
To permit phylogenetic analysis of the progression from primary tumor to metastasis a total of 39 fresh frozen tumor samples were
collected from 17 females and subjected to whole genome sequencing. Samples were obtained from Dana-Farber Cancer Institute,
Boston, US (7 cases); Kings College Hospital, London, UK (4 cases); The Erasmus MC Cancer Center, Rotterdam, The Netherlands
(4 cases); The Institute Jules Bordet, Brussels, Belgium (2 cases) in line with local ethics committee approvals (project SHARE’’ #93-
085, approved by the Dana-Farber Harvard Cancer Center institutional review board; UK, REC reference: 10/H0804/33, approved by
Guy’s and St Thomas’ NHS Trust ethics committee; MEC 02.953, approved by the medical ethical committee of the Academisch
Ziekenhuis Rotterdam (EUR/ AZR) for ‘The retrospective assessment of cell biological factors in archival tumor tissues’; Protocol
1698 and 1634, approved by the Institut Jules Bordet local ethics committee). For each individual, in addition to a primary tumor
and a matched normal sample at least one sample from a distinct metastatic scenario was included in the experiment. For 7 individ-
uals, where the metastasis scenario sample was limited to a synchronous lymph node deposit these samples were only included in
the whole genome analysis where they form a comparison cohort. For one patient (PD8948) where the apparent relapse samples
were identified as distinct primary tumors we collected two additional tumor samples from formalin fixed paraffin embedded
(FFPE) tissue blocks and performed targeted capture on both samples and whole genome sequencing on one (Figure S3).
Relapsed Breast Cancer Cohort
The number of cases of locally relapsed and distant metastatic breast cancer was extended from 10 to 163 by including a second
cohort of patients for whom 365 cancer related genes were sequenced using a targeted capture pulldown approach. Samples from
these individuals form the ‘relapsed breast cancer cohort’. The additional 153 patients were drawn from a single centre study at the
Department of Oncology, Haukeland University Hospital, performed with the aim of identifying genetic alterations in advanced and
REAGENT or RESOURCE SOURCE IDENTIFIER
Deposited Data
Targeted and whole genome
sequence data
https://www.ebi.ac.uk/ega/ Accession numbers: Targeted (2939stdy)
EGAD00001002698
Exome (492stdy): EGAD00001002697
Whole genome (2040stdy): EGAD00001002696
Somatic Mutation Calls Mendeley Data http://dx.doi.org/10.17632/g7kpzkhz8c.1
Software and Algorithms
The Sanger’s Cancer Genome Project core
somatic calling workflow from
the ICGC PanCancer Analysis of Whole
Genomes (PCAWG) project
https://dockstore.org/containers/quay.io/
pancancer/pcawg-sanger-cgp-workflow
Cancer Cell 32, 169–184.e1–e7, August 14, 2017 e1
metastatic breast cancer deposits. Between March 1996 and October 2004, a total of 206 patients with non-operable primary breast
cancers, local relapse and / or metastatic deposits suitable for biopsy were recruited to the study. All samples were snap-frozen in
liquid nitrogen in the operating room immediately upon removal from the patient. We analysed a total of 259 tumor samples from 153
patients. Patients included in the analysis had at least one sample from a distant metastatic or locoregional relapse deposit that con-
tained sufficient material for DNA extraction, allowing MPS. For 41 patients included in the study we were able to identify a primary
tumor sample and extract sufficient DNA for MPS (FFPE, n = 29; fresh frozen, n = 12). In addition, DNA was retrieved from FFPE-
blocks from four metastatic deposits undergoing routine biopsy in the time period between primary and fresh frozen metastatic tissue
collection, 2 primary tumors after neo-adjuvant chemotherapy and a synchronous lymph node deposit. The study was approved by
the regional ethics committee of the Norwegian Health Region West (218/97 – 77.97; REK Vest), and all patients provided written
informed consent.
In total, 163 individuals were therefore included within the relapsed breast cancer cohort and for each patient at least one sample
(total number of relapse samples = 227) was obtained from a distant metastatic deposit (n = 79) or a metachronous loco-regional
relapse (n = 148) (Table S1). Multiple relapse samples were collected for 46 individuals (range 2-5 samples per individual). A matched
primary tumor sample was collected in 51 cases and a matched germline sample was collected for 80 cases (adjacent normal breast
tissue, n = 6; blood, n = 74). The distribution of relapse sample sites is presented in Table S1. Most (177/227) relapse and metastasis
samples were pre-treated, being exposed to an average of 1.7 (range 0-5) lines of systemic therapy Table S1. A total of 80 samples
from 57 individuals were also obtained after exposure to external beam radiotherapy.
Primary Breast Cancer (Comparison) Cohort
The primary tumor comparison cohort consisted of previously published exome data from 705 individual’s primary breast cancers,
freely available from The Cancer Genome Atlas (TCGA). We included properly matched samples that were available for download
from CGHub on December 2015. We excluded variants where the matched normal coverage was lower than 10-fold and samples
for which less than 50% of the mutations detected by our calling pipeline were present in the somatic mutation calls released by
TCGA. To minimize bias in our comparisons we applied the same mutation calling algorithms, post-processing filters and driver
annotation processes as were used for in in-house generated data for the relapse cohort. Annotated mutation data for these samples,
within the scope of the cancer gene panel is available in Table S3. Clinical information for the 705 patients in the primary cohort was
downloaded from https://tcga-data.nci.nih.gov/docs/publications/brca_2012/ (file = BRCA_Clinical.tar.gz ). A comparison of the
clinical characteristics of the primary cohort and relapse cohort at the point of diagnosis is provided in Table S1. Cancer staging in-
formation for each dataset was determined using the American Joint Committee on Cancer (AJCC) Staging Manual, 7
th
edition. When
nodal status was recorded as ‘Nx’ within clinical information this is assumed to reflect node negative disease (‘N0’).
METHOD DETAILS
Sample Size
The sample size of 163 recurrent breast cancers has 99% power that a cancer gene mutated in 5% of breast cancer recurrences
would be seen in at least 3 patients in the cohort.
Tumor Specimen Processing
All samples within the whole genome and relapse breast cancer cohorts were histopathologically assessed to ensure adequate tu-
mor cellularity (>=70%) and if necessary macrodissection was performed. Where possible for both primary tumor and relapse
samples ER and PgR expression was determined by local pathologists as Allred scores of 4 or above. Where available, HER2
over-expression was determined by IHC scores of 3+ or 2+ confirmed by fluorescent in-situ hybridisation. Due to the historical nature
of the Haukeland University Hospital sample set, HER2 expression data however, is scarce and HER2 amplification was determined
from sequence data using the criteria for identifying amplifications in targeted capture data as described below. We have previously
shown our approach to yield results that are highly consistent with clinical HER2 status results (Yates et al., 2015).
DNA Extraction
DNA from fresh frozen tumor tissue specimens and blood samples was isolated, using spin columns from the QIAamp DNA mini kit
(Qiagen). The procedure was performed according to the manufacturer’s instructions with the exception that 400ul sample (instead of
200ul) was used as input in the cases where full blood on EDTA were used instead of leukocyte concentrates (Haukeland University
Hospital cases). DNA from formalin fixed paraffin embedded tissue (FFPE) was isolated, using spin columns from the QIAamp DNA
FFPE Tissue Kit (Qiagen). The procedure was performed according to the manufacturer’s instructions, with the following exceptions:
The de-paraffinization step with xylene was repeated three times and the subsequent washing step with ethanol was repeated twice.
Lysis of tissue was performed using 540 ml buffer ATL and 60 ml proteinase K per samples, for 2-4 hours at 56C, before addition of a
further 180 ml buffer ATL and 20 ml proteinase K and an over-night incubation at 56C.
Multi-Sample Whole Genome Sequencing
Genomic libraries with insert sizes of 300bp-600bp were derived from native DNA from 39 tumor and 17 matched normal fresh frozen
samples using Illuminapaired end sample preparation kits according to manufacturers instructions. Following cluster generation,
e2 Cancer Cell 32, 169–184.e1–e7, August 14, 2017
100bp paired-end sequence data was generated using Illumina HiSeqs and was subsequently aligned to the reference human
genome (NCBI build37) using BWA. Whole genome libraries from a single FFPE tumor (PD8948c) and matched fresh frozen normal
sample (PD8948b) were prepared using Agilent Technologies Sure Select library preparation kit (Custom library kit (cat no. 930075)
http://www.agilent.com/search/?Ntt=930075 following manufacturers instructions. 150bp paired end sequence data (with average
insert sizes of 319bp and 481bp respectively) was generated using Illumina X10. The average genome wide sequence coverage of
tumors and matched normal samples was 42 and 31 fold respectively (Table S2).
Multi-Region Targeted Gene Screen
For targeted capture pulldown experiments we used a bait design that consisted of over 8,000 targets of which almost 6,000 covered
the exons of 365 genes. To facilitate copy number analyses baits were also included to target over 2,000 SNPs outside of exonic
locations. Genomic DNA from tumor and matched normal samples, was fragmented using Covaris(average insert size 150bp)
and subjected to IlluminaDNA sequencing library preparation using Agilent’sBravo Automated liquid handling platform. Tumor
and normal samples were indexed with unique barcodes using PCR. Libraries were then hybridised to custom ribonucleic (RNA) baits
according to the AgilentSureSelectprotocol. Samples were multiplexed on average 16 samples per lane and flow-cell clusters
created. Paired-end, 75bp sequence reads were generated using Illumina HiSeq 2000. Sequence data was re-aligned to the human
genome (NCBI build 37) using BWA. Unmapped reads, PCR duplicates and those outside of the target region were excluded from
analysis. The average sequence coverage of tumors and matched normal samples was 467 and 505 fold respectively (Table S2).
Multi-Sample Mutation Calling
Substitutions, indels and structural variant breakpoints were called independently in each tumor sample using mutation calling algo-
rithms (CaVEMan, Pindel and BRASS) and post-processing filters as previously described (Yates et al., 2015). Mutation calling algo-
rithms used in the analysis are freely available at https://github.com/cancerit/. Where an individual had more than one tumor sample
we performed a comparative analysis of SNP and indel variant data for union of sites from all related samples in an unbiased manner
using in-house software – vafCorrect, that is freely available at https://github.com/cancerit/vafCorrect. For substitutions unbiased
pileup results were obtained using Bio::DB::HTS (https://github.com/Ensembl/Bio-DB-HTS). For indels the approach includes un-
mapped reads whose pair is mapped within the vicinity (defined by library insert size) of the indel site and resulting reads were aligned
using exonerate to original reference sequence and alternate reference sequence (created by inserting the indel variant at the given
reference location). Exonerate output was then parsed to count the fraction of reads aligned to original reference and alternate refer-
ence sequence. Reads that were mapped with equal identity scores to reference and alternate sequence were reported as
ambiguous reads while reads that were present at the variant location but did not map to either of the reference sequences were
categorized as unknown reads. Data quality was ensured and the impact of germline SNP contamination minimized by filtering
against an extended unmatched normal panel of over 200 samples, cross-referencing with available germline SNP databases, using
a matched normal sample where available and visually inspecting local alignments for all reported coding mutations.
Comprehensive lists of all somatic substitutions, indels and structural variants from whole genome analysis are available for down-
load at review@sftpsrv.sanger.ac.uk. All high confidence mutation calls within the scope of the cancer gene panel are presented in
Table S3.
Mutation Validation
For the whole genome experiment, native DNA where available (25 tumor and 14 normal samples) or whole genome amplified (WGA)
DNA when necessary (samples PD13596a, PD13596b, PD13596c, PD4243a, PD4252c, PD48102a, PD8948d, PD8948e) was sub-
jected to custom capture pulldown and high depth re-sequencing to a target depth of 1000-fold. Probes were designed for 6,534
genome-wide substitutions and indels using Agilent Technologies SureSelect Standard DNA Design Wizard. High-stringency repeat
masking, a tiling density of 2X and balanced boosting were applied to the design. DNA capture (paired-end, average insert size
150bp) libraries were multiplexed and sequenced using Illumina MiSeqto an average coverage of 1,076-fold. We have previously
published validation data for case PD9771 (Yates et al., 2015). To determine an experimental validation rate, all coding indels (n=144)
and substitutions (n=1,498) were included in the experiment. A true positive validation of 94% was identified for both coding indels
and substitutions independently. Amongst substitutions the most common reason for failure to validate was low coverage (4%) and
this was usually associated with the use of WGA material. Excluding WGA validation experiments was associated with a validation
rate of 97% and this is believed to be a more reliable reflection of the true positive rate (Table S4).
The remaining variants included in the high-depth pulldown design were selected to enable validation and refinement of phyloge-
netic tree structures. The approach was biased towards subclonal events and mutations that contradicted the consensus tree.
Mutations close to each other or within 200bp of germline snps were also enriched to permit reconstruction of the subclonal structure
through phasing approaches. Using this approach, across 13 cases (cases PD13596, PD4252, PD4820, PD114780 excluded for rea-
sons stated above), 45 out of 48 tree branches were identified through an independent clustering experiment, re-capitulating and
therefore validating the basic tree structure in each case. A total of 3 small branches failed to validate and consisted of 2-3% of
the overall mutation burden in each case. See Figure S1 and Table S5 for details of phylogenetic tree construction and validation
in each case.
Regarding multi-sample targeted capture experiments we have previously demonstrated a 99% consistency rate in reporting
mutation presence and absence (Yates et al., 2015) using custom pull down duplicate experiments. Furthermore, we validated
Cancer Cell 32, 169–184.e1–e7, August 14, 2017 e3
non-synonymous mutations in ESR1, JAK2 and PIK3R1 using capillary sequencing (Table S4). One ESR1 mutation was validated in
an independent exome experiment (Brown, 2017).
Cancer Gene Discovery
To identify recurrently mutated driver genes we used dNdScv, as previously described in detail (Nik-Zainal et al., 2016). This method
uses dN/dS and covariates to detect genes with higher density of non-synonymous coding mutations than expected by chance. The
method considers the trinucleotide mutation spectrum, the sequence of each gene, the impact of coding mutations (synonymous,
missense, nonsense, splice sites substitutions and indels) and the variation of the mutation rate across genes. Multiple testing correc-
tion (Benjamini-Hochberg FDR) was applied across analyzed genes and a q value < 0.1 was used to determine statistical significance.
For the relapse cohort significance was tested across the 311 genes for which at least one mutation was called. The approach was
performed across all relapse samples and across the subset of samples with a matched germline sample. STAT3 was significantly
mutated in the matched sample analysis only. Within the exome analysis over 20,000 genes are analyzed.
Driver Mutation Annotation
Each coding variant was manually curated with a likely driver status following a systematic approach. Firstly, likely cancer genes were
identified as either those found within the dNdS cancer gene discovery approach described above, from published reference mate-
rials consisting of the Cancer Gene Census (Futreal et al., 2004), the Cancer5000 series (Lawrence et al., 2014) or from literature re-
view of breast cancer sequencing studies (Banerji et al., 2012; Cancer Genome Atlas Network, 2012; Ellis et al., 2012; Shah et al.,
2012; Stephens et al., 2012). Subsequently, oncogenic mutations were annotated within these cancer genes. Oncogenic mutations
were defined as those falling into one of the following categories: 1) A canonical oncogenic mutation in a recurrent mutation hotspot;
2) A lower frequency recurrent mutation in a known oncogene with 3 or more confirmed somatic non-synonymous substitutions or in-
frame deletions previously reported at this locus in COSMIC or confirmed through experimental models or special cohorts (i.e. ESR1
resistance mutations); 3) Likely damaging events in a known tumour suppressor that include truncating (nonsense), frame-shift,
essential splice variants or those within a mutation hotspot (>=2 somatic mutations); 4) Silent mutations in a known recurrent splice
site hotspot.
Genome-Wide Subclonal Copy Number Analyses
Segmental copy number information was derived from all targeted capture and whole genome data using the Allele Specific Copy
Number Analysis of Tumors (ASCAT) algorithm (Van Loo et al., 2010). The Battenberg algorithm was used to identify clonal and sub-
clonal copy number changes in whole genome sequence data as previously described (Nik-Zainal et al., 2012; Yates et al., 2015) and
was also used to challenge and confirm copy number and ploidy estimates derived from ASCAT. The approach phases germline
SNPs within MPS data using Impute2 (Howie et al., 2009) that uses a well characterized panel of polymorphic SNPs.
Within whole genome data, copy number segments are reported as amplified when present at more than twice the estimated
average ploidy across the whole genome. Homozygous deletions are identified as segments where total copy number equals
zero or equivalent in an area of subclonal copy number. Within targeted capture data the mean logR and 95% confidence interval
was calculated across known cancer driver genes. Potential amplifications in common breast cancer genes were identified based
on a mean logR of > 1, equating to 6 alleles in a diploid genome and tumor cellularity of 50%. For related samples where heterogeneity
of amplification events was called logR and BAFs across all genes and point mutation data were reviewed manually in each related
sample to determine if heterogeneity is likely a consequence of low aberrant cell fraction as opposed to true driver heterogeneity. This
conservative approach was adopted to minimize the risk of over-calling heterogeneity.
Genome-Wide Multi-Sample Clonality Analyses
For the 17 patients with multi-sample whole genome sequencing data, to model the subclonal structure across multiple related sam-
ples previously described bioinformatics and deductive reasoning approaches were adopted. The approach follows 3 main steps
including the identification of large-scale subclonal copy number changes using the Battenberg algorithm (Nik-Zainal et al., 2012),
clustering of subclonal somatic substitutions using a Bayesian Dirichlet process in multiple dimensions across related samples
and hierarchical ordering across multiple samples using the ‘pigeon hole principle’. Strict quality control is applied to the mutations
included in clustering analysis to avoid the generation of false positive clusters of mutations:
dDuring evolution, copy number losses may result in the loss of mutations in the affected regions, resulting in clusters of muta-
tions found uniquely in the unaffected sample(s). In order to avoid falsely calling such mutations as arising from a clonal expan-
sion in the unaffected samples, such mutations are excluded from Dirichlet process clustering.
dSome mutations may be present in multiple samples, but only called in a subset of samples, due to low allele frequency in the
other sample(s). To avoid false negatives, allele frequencies of all mutations found in any sample from a patient are therefore re-
called, with a minimum mapping quality and base quality of 10.
dThe allele frequencies of all mutations are adjusted to cancer cell fraction using purity and copy number information. Copy num-
ber segments have start and end points defined by heterozygous SNP locations, so somatic variants that fall between these
boundaries have undefined copy number and are excluded from clustering.
e4 Cancer Cell 32, 169–184.e1–e7, August 14, 2017
A median of 95% (range 77 – 99%) of mutations are included in clustering. Using this approach each substitution that passed qual-
ity control was assigned to a specific cluster (Tables S5). For each individual case, data including cluster size (equating to phyloge-
netic tree branch length), cluster ‘position’ (reflecting the proportion of cells containing the mutation cluster in each related sample)
and posterior confidence intervals are presented for both discovery and validation experiments in Table S5 and Figure S1.
Mutation Timing in Multi-Sample Analyses
The relative contribution of the different mutation types during evolution (Figure 2) was estimated by comparing the proportion of
mutations that were shared, private to the primary tumor sample or private to the metastasis/ relapse sample. Each individual point
mutation was assigned to one of these categories by calculating for each mutation, in all related samples independently, R’s pbino-
mial statistic based upon a conservative, expected error rate of 1 in 200. A mutation was deemed to be present or absent from an
individual sample based upon a p value of <=0.05 or > 0.05 respectively. All structural variants reconstructed in silico were deter-
mined to be shared or private to the primary/ metastasis samples based upon either reconstruction in related samples or the pres-
ence of 4 or more split reads supporting the breakpoint using BRASS1. Substitution branch timing (Figures 1 and 2) was calculated
using mutation clustering where the cluster size dictates the branch length.
Whole Genome Duplication Timing Analysis
In this study we have estimated the prevalence of 3 different developmental stages for 22 of the breast cancer samples. The first one
corresponds to the diploid stage previous to whole genome duplication. The second one is the tetraploid cell stage after the whole
genome duplication was acquired and previous to the subclonal diversification. Lastly, the timing between the last selective sweep
and the emergence of the detected subclones. The duration of each of the stages in molecular time is estimated via the fraction of
mutations having arisen in each of the phases. To estimate the proportions of mutations in each stage we employ a strategy similar to
that of Purdom et al. (Purdom et al., 2013) and extend it to subclonal mutations.
Let rdenote the purity of the sample. The expected variant allele frequency f
i
for a mutation arising in state I depends on the number
mutated alleles m
i
, the total copy number c (4 in our case) and the prevalence of the subclone p
i
. For early clonal mutations we have
p
i
= 1 and m
i
= 2, for late clonal mutations we have m
i
= 1. For subclonal mutations we have p
i
< 1 and m
i
=1.
f=rmipi
4r+2ð1rÞ
We model the number of reads X arising from a mutation in stage I as a binomial with coverage n.
XjiBinom ðn;fiÞ
The probability that a mutation occurs in stage I is p
i
. This gives rise to a binomial mixture model.
PðX;iÞ=PðXjiÞ3pi
Using Bayes’ formula we can compute the probability of being in state I given X as
PðijXÞ=PðX;iÞ
PðXÞ=PðXjiÞ3pi
PiPðXjiÞ3pi
For a series of k observed mutations with variant reads x
1
,.x
k
, we can estimate the mixture proportions p
i
using and EM
algorithm.
Knowing the probabilities p
i
, for early (p
e
) and late (p
l
) stages we can calculate an estimate the relative time of WGD as:
t=2pe
2pe+pl
To assess the robustness of the above estimator and to calculate confidence intervals we use bootstrapping, subsampling 100
times from the number of observed mutations with replacement and calculating t for each of the subsamples.
Presented analyses were first applied to all mutations within individual samples with results being consistent with duplication
arising prior to primary-relapse divergence. A more accurate estimate of the timing of whole genome duplication was then deter-
mined by restricting the analysis to shared, clonal mutations allocated to the trunk of the phylogenetic tree.
Driver Mutation Enrichment Analyses
The frequency with which each cancer gene (ESR1 or genes significantly mutated in the driver discovery experiment) was altered by a
driver mutation was compared between the relapsed and primary breast cancer cohorts using a two-sided Fishers’s exact test. A
Benjamini-Hochberg correction for multiple testing was applied to generate false discovery rates (q). A total of 7 genes were signif-
icantly enriched in the relapsed compared to the primary cohort (defined by q < 0.1) while no genes were enriched in the primary
cohort.
Cancer Cell 32, 169–184.e1–e7, August 14, 2017 e5
Mutational Signature Analysis
We assessed the relative activity of mutational processes over time by allocating somatic mutations to their specific branch of the
phylogenetic tree and subjecting individual branches (composed of more than 20 mutations) to mutational signature analysis (Fig-
ure 3). Mutational signatures were detected in two independent ways: (i) de novo extraction based on somatic substitutions and their
immediate sequence context and (ii) refitting of previously identified consensus signatures of mutational processes. The de novo
extraction was performed using a previously developed theoretical model and its corresponding computational framework (Alexan-
drov et al., 2013b). Briefly, the algorithm deciphers the minimal set of mutational signatures that optimally explains the proportion of
each mutation type in each mutational catalogue and then estimates the contribution of each signature to each sample. Within this
dataset the computational framework identified five reproducible mutational signatures that closely resembled previously identified
breast cancer signatures.
In the second stage, 27 distinct consensus mutational signatures previously identified from examining 7,042 samples across 30
different cancer types were ‘refitted’ (Alexandrov et al., 2013a). All possible combinations of up to seven mutational signatures
were evaluated for each sample. This resulted in 1,285,623 solutions per sample and a model selection was applied to select the
optimal solution. The model selection framework excludes any solution in which a mutational signature contributes less that 2%
of the somatic mutations or less than 50 somatic mutations. Exceptions were made for Signatures 1 and 5 as these are believed
to reflect on-going endogenous mutational processes that continuously contribute very low numbers of somatic mutations (Alexan-
drov et al., 2013a). Further, the model selection framework selects the solution that optimizes the Pearson correlation between the
original pattern of somatic mutations and the one based on refitting the sample with consensus mutational signatures such that each
additional signature should improve the Pearson correlation with at least 0.02. The final solution for each sample contained between
3-6 mutational signatures and these signatures were consistent with the ones previously identified by the de novo analysis: Signature
1, Signature 2, Signature 3, Signature 5, Signature 8, and Signature 13.
Telomere Length Estimates
Telomerecat is a de novo method for the estimation of telomere length from whole genome sequencing samples. The algorithm works
by comparing the ratio of complete telomere reads to reads on the boundary between telomere and subtelomere. The ratio is trans-
formed to a measure of length using a simulation approach that takes into account the fragment length distribution of the sample. By
considering the ratio of complete telomere reads to boundary reads, Telomerecat estimates coverage over the telomere without
interface from the affects of aneuploidy, a common occurrence in cancer. Telomerecat also corrects for error in sequencing reads
by modeling the observed distribution of phred scores associated with mismatches to the telomere sequence.
Case PD8948 and Whole Genome Sequencing and Analysis of an FFPE Sample
For all but one cancer in the dataset we found that thousands or tens of thousands of somatic substitutions were shared by the pri-
mary and metastasis sample. In one case (PD8948) however, we determined from the clonal mapping of over 16,000 somatic sub-
stitutions, indels and structural variants that the two fresh frozen DNA samples from tumors in the left and right breast (samples
PD8948d and PD8948e respectively) sampled 1 year apart were clonally unrelated cancers. Only 95 (0.6%) point mutations were
detected in both samples, none of which fell within coding regions, and validation through visual inspection and/ or targeted capture
pulldown failed to identify any mutation as a true positive in both samples. Copy number profiles and structural variant profiles from
the two cancers were also distinct. In the absence of shared somatic events we conclude that these samples are derived from 2 in-
dependent primary tumors. The samples however, shared thousands of germline SNPs and a BRCA1 frame-shift mutation confirm-
ing that they are derived from the same individual who was a known germline BRCA1 mutation carrier.
To further explore the clonal evolution of this patient’s cancers we identified 2 additional FFPE samples from earlier tumor deposits
within the left breast (PD8948a and PD8948c). These samples were subjected to targeted gene panel sequencing and the likely
phylogenetic relationships between the four samples were then inferred from coding non-synonymous mutations as demonstrated
in Figure S3A. The findings were consistent with the patient having developed 3 separate primary tumors during her lifetime, each
containing a distinct TP53 mutation. Two samples (PD8948c and PD8948e) harbored identical TP53 (p.Y220C) and KDM6A muta-
tions suggesting that the later sample represented distant relapse. Genome-wide analysis could provide conclusive evidence to
confirm the relatedness of two such samples, however one sample (PD8948c) was derived from a 7 year old FFPE sample and there
is little experience of whole genome sequencing of FFPE derived tumors. Weand others have previously shown low error rates for
gene capture and whole exome sequencing of FFPE samples but to date we are only aware of the results from a single tumor sample
sequenced to whole genome level and the widespread applicability of this single case is unclear. We predicted that the process of
fixation and storage of such material could result in the introduction of technical artifacts and could compromise mutation-calling
sensitivity. However, for the purposes of this experiment identifying a significant overlap of mutations called in the later sample
(PD8948e) would confirm relatedness of the samples.
Library preparation of the FFPE sample was performed following our standard protocol and the tumor sample and a matched blood
derived normal sample were sequenced to 31X and 38X respectively using Illumina X10. Mutations were called using the same
algorithms as previously described. Confirming the clonal relationship of the two samples, a significant proportion of somatic muta-
tions of all classes – substitutions (25%), indels (18%) and structural variants (14%), were shared. This equates to almost 2,000 com-
mon somatic mutations, genome-wide (Figure S3B).
e6 Cancer Cell 32, 169–184.e1–e7, August 14, 2017
Analysis of other whole genome triplet cases within the cohort identified that all metachronous samples contain a significant
excess point mutation burden compared to the primary tumor. However, this was inconsistent in this case, where the presumed pri-
mary tumor (PD8948c, FFPE) and the relapse sample obtained 3 years later (PD8948e, fresh frozen) contained a similar private point
mutation burden (Figure S3B). We investigated whether the unexpected excess of FFPE specific mutations was a likely biological or
technical phenomenon by comparing the mutation spectra in the 2 samples. The substitution profile within the FFPE sample indi-
cated an excess of C>A base changes and these tended to occur in the context of one or two 5 prime cytosine nucleotides (Fig-
ure S3C). To investigate this further we applied formal mutational signature analysis to the private and shared branches from an
inferred phylogenetic tree derived from these samples (Figure S3D). The analysis confirmed that the shared mutations were drawn
from 3 mutation signatures – two clock-like signatures and a dominant signature associated with homologous recombination defi-
ciency (signature 3), consistent with the known BRCA1 mutation carrier status. Private mutations identified in the fresh frozen sample
(PD8948e) followed an almost identical signature distribution. In contrast, none of the mutations that were private to the FFPE sample
were assigned to these signatures, but rather were purely assigned to a mutation signature (R2) - a known sequencing artifact that
arises due to oxidative damage and has previously been described in relation to exome library preparation (Costello et al., 2013). In
constructing the phylogenetic tree in Figure 1 we therefore omit a private to primary tumor branch, although it is conceivable that a
small number of true private mutations were undetected due to the presence of an overwhelming artifact.
QUANTIFICATION AND STATISTICAL ANALYSIS
Statistical analysis was performed and graphics produced using R version 3.0.1: A language and environment for statistical
computing (R Foundation for Statistical Computing, Vienna, Austria. Alignment viewing was performed using Gbrowse, Jbrowse,
Samtoolstview and IGV. All hypothesis tests were 2-sided when appropriate and statistical tests used are specified in Results
and figure legends.
DATA AND SOFTWARE AVAILABILITY
Targeted and whole genome sequence data has been deposited at the European Genome-Phenome Archive (https://www.ebi.ac.
uk/ega/ at the EBI) with accession numbers:
dTargeted (2939stdy) EGAD00001002698;
dExome (492stdy): EGAD00001002697;
dWhole genome (2040stdy): EGAD00001002696.
Full somatic mutation calls (substitutions, indels and structural variants) for each individual cancer analyzed by whole genome
sequencing are available for download from Mendeley Data. The link for the dataset is: http://dx.doi.org/10.17632/g7kpzkhz8c.1
The most recent version of our whole genome sequencing mutation pipeline is available as a Docker image. This, together with
documentation, can be accessed from https://dockstore.org/containers/quay.io/pancancer/pcawg-sanger-cgp-workflow.
Cancer Cell 32, 169–184.e1–e7, August 14, 2017 e7
... This hypothesis is supported by multiple randomized trials showing similar complete response rates of synchronous regional lymph nodes and primary tumour 20 . In contrast, oligometastatic recurrences can acquire genomic alterations, either spontaneously or under selective pressure from prior treatment, that make them relatively resistant to treatment 21 . Assuming that the primary tumour and de novo metastatic sites have similar treatment sensitivities, complete remission could be achieved by adding de nitive RT doses to metastatic sites to standard of care treatment of early breast cancer with the aim of eradicating cell populations that have not responded favourably to systemic treatment. ...
Preprint
Full-text available
Introduction It is estimated that 10–25% of breast cancer patients have metastatic disease upon diagnosis with roughly 20% having limited metastatic sites. The optimal management of patients with de novo bone-only oligometastatic breast cancer (OMBC), particularly curative-intent approaches, continues to evolve given extremely limited evidence in survival outcomes with this strategy. Methods This was a single-center retrospective review of the survival outcomes and details of treatment of patients with de novo OMBC who received tri-modality treatment with systemic therapy (chemotherapy with or without endocrine treatment and/or HER2-directed therapy), surgery, and radiotherapy to local and metastatic site/s. Progression-free survival (PFS) was defined as the time from initiation of treatment (either surgery, or neoadjuvant systemic therapy) to date of clinical or radiologic progression. Results From January 2014-March 2024, we identified 10 women who fit the inclusion criteria. Seven had isolated bone metastasis, and none had more than 3 metastatic sites. The included cohort had a mean age of 44.2 years. Nine had hormone receptor-positive disease, and 5 were HER2-positive. All patients were discussed in a multidisciplinary meeting. Median PFS by Kaplan-Meier analysis was 40.2 months. Nine patients were still alive at the time of analysis, and 6 of them remain disease free with a median follow-up duration of 30.8 months. Conclusion Patients with de novo bone-only oligometastatic breast cancer seem to benefit from the standard curative-intent tri-modality approach with the addition of ablative radiation to metastatic sites. These patients have a long median PFS and can be rendered disease-free for many years.
... Approximately 45% of patients exhibit at least one instance of WGD [24,25], which is associated with the incidence of somatic CNAs [25] and tumor mutation burden [22,23]. WGD occurs frequently in certain TNBC subtypes, such as BLIS [5], and may play a critical role in tumor evolution [26]. ...
Article
Full-text available
Background Triple‐negative breast cancer (TNBC) is a heterogeneous disease characterized by high aggressiveness, high malignancy, and poor prognosis compared to other breast cancer subtypes. Objective This review aims to explore recent advances in understanding TNBC and to provide new insights and potential references for clinical treatment. Methods We examined current literature on TNBC to analyze molecular subtypes, genetic mutations, signaling pathways, mechanisms of drug resistance, and emerging therapies. Results Findings highlight key aspects of TNBC's molecular subtypes, relevant mutations, and pathways, alongside emerging treatments that target drug resistance mechanisms. Conclusion These insights into TNBC pathogenesis may help guide future therapeutic strategies and improve clinical outcomes for patients with TNBC.
... Elapsed time from sample collection to its genomic profiling can directly affect the validity of genomic parameters due to technical reasons, such as nucleic acid fragmentation yielding artifact mutations, or affect tumor genomic signatures, such as TMB [17,18]. Moreover, for outdated CGP, the natural trajectory of genomic tumor evolution may inaccurately reflect the current status of profiled tumors [19,20]. In our experience, about 5% of patients presented with CPG which were deemed uninformative to guide MTB's recommendations because of outdated material or narrow targeted NGS panels. ...
Article
Full-text available
Purpose Comprehensive genomic profiling is becoming increasingly important in the management of patients with metastatic breast cancer (mBC). Real-world clinical outcomes from applying molecular tumor boards (MTBs) recommendations in this context remain limited. Accordingly, we conducted a retrospective, single-institution analysis to evaluate the clinical impact of discussing patients affected by mBC at the MTB. Methods Clinicogenomic data of patients affected by mBCs referred to the European Institute of Oncology MTB between August 2019 and December 2023 were reviewed. Genomic alterations were classified by ESCAT framework. Clinical outcomes of patients showing actionable alterations and receiving molecular-matched therapy (MMT) were compared to those receiving standard therapy (ST). Results Ninety-six patients were included. Following MTB discussion, genetic counseling was recommended in 27% (n = 26) of patients, while additional molecular analyses were requested in 25% (n = 24) cases. Fifty-six patients (58%) displayed at least one actionable alteration. For patients with available follow-up (n = 50), 32 (64%) received MMTs and 18 (36%) ST. No differences in real-world progression-free survival (rwPFS) (4.07 months [95% CI 2.14–8.28] vs. 3.12 months [95% CI 1.51-NE], P = 0.8) and 12-month overall survival (OS) (58% [95%CI 43–78] vs. 57% [95%CI 34–97), P = 0.9) were observed between the MMT- and ST-group. Level I ESCAT alterations yielded longer rwPFS (5.82 months [95% CI 3.12–8.41]) compared to ESCAT II (2.14 months [95%CI 1.61-NE]) and ESCAT III (2.10 months [95% CI 2.04-NE]; P = 0.03). Twenty-four percent of patients showed a PFS2/PFS1 ratio > 1.3 from MMT. Conclusion Molecular tumor boards can provide additional treatment options for patients affected by mBC. Besides treatment recommendations, MTBs also have the utility to assess the validity of discussed genomic reports and to identify alterations worthy of genetic counseling.
... Recent next-generation sequencing-based studies, including The Cancer Genome Atlas (TCGA) program, have uncovered the genetic landscape of invasive breast cancers, [7][8][9][10] revealing driver mutations in TP53, PIK3CA, PTEN, BRCA2, ESR1, GATA3, KMT2C, NCOR1, AKT1, etc. The Clinical Proteomic Tumor Analysis Consortium (CPTAC) breast cancer study [11] reported that the proteogenomic landscape of 122 invasive breast cancers provides insights into clinically relevant biology, including cell cycle dysregulation, tumor immunogenicity, aberrant metabolism, and heterogeneity in therapeutic target expression. ...
Article
Full-text available
Multi‐omics studies of breast ductal carcinoma (BRDC) have advanced the understanding of the disease's biology and accelerated targeted therapies. However, the temporal order of a series of biological events in the progression of BRDC is still poorly understood. A comprehensive proteogenomic analysis of 224 samples from 168 patients with malignant and benign breast diseases is carried out. Proteogenomic analysis reveals the characteristics of linear multi‐step progression of BRDC, such as tumor protein P53 (TP53) mutation‐associated estrogen receptor 1 (ESR1) overexpression is involved in the transition from ductal hyperplasia (DH) to ductal carcinoma in situ (DCIS). 6q21 amplification‐associated nuclear receptor subfamily 3 group C member 1 (NR3C1) overexpression helps DCIS_Pure (pure DCIS, no histologic evidence of invasion) cells avoid immune destruction. The T‐cell lymphoma invasion and metastasis 1, androgen receptor, and aldo‐keto reductase family 1 member C1 (TIAM1‐AR‐AKR1C1) axis promotes cell invasion and migration in DCIS_adjIDC (DCIS regions of invasive cancers). In addition, AKR1C1 is identified as a potential therapeutic target and demonstrated the inhibitory effect of aspirin and dydrogesterone as its inhibitors on tumor cells. The integrative multi‐omics analysis helps to understand the progression of BRDC and provides an opportunity to treat BRDC in different stages.
... We analyzed the metastatic spread of the primary tumor of each mouse ex vivo in the distant organs lung, liver, brain, and spleen. Except for the spleen, these organs are predominant sites for metastasis formation of primary breast cancer in patients, also known as "organotropic metastasis" [44,45]. Additionally, we conducted a xenograft mouse experiment with a control cell line (MDA-MB 231 Tripz Ctrl Luc), which expresses a scrambled sequence after DOX administration. ...
Article
Full-text available
Advanced breast cancer, as well as ineffective treatments leading to surviving cancer cells, can result in the dissemination of these malignant cells from the primary tumor to distant organs. Recent research has shown that microRNA 200c (miR‐200c) can hamper certain steps of the invasion–metastasis cascade. However, it is still unclear whether miR‐200c expression alone is sufficient to prevent breast cancer cells from metastasis formation. Hence, we performed a xenograft mouse experiment with inducible miR‐200c expression in MDA‐MB 231 cells. The ex vivo analysis of metastatic sites in a multitude of organs, including lung, liver, brain, and spleen, revealed a dramatically reduced metastatic burden in mice with miR‐200c‐expressing tumors. A fundamental prerequisite for metastasis formation is the motility of cancer cells and, therefore, their migration. Consequently, we analyzed the effect of miR‐200c on collective‐ and single‐cell migration in vitro, utilizing MDA‐MB 231 and MCF7 cell systems with genetically modified miR‐200c expression. Analysis of collective‐cell migration revealed confluence‐dependent motility of cells with altered miR‐200c expression. Additionally, scratch assays showed an enhanced predisposition of miR‐200c‐negative cells to leave cell clusters. The in‐between stage of collective‐ and single‐cell migration was validated using transwell assays, which showed reduced migration of miR‐200c‐positive cells. Finally, to measure migration at the single‐cell level, a novel assay on dumbbell‐shaped micropatterns was performed, which revealed that miR‐200c critically determines confined cell motility. All of these results demonstrate that sole expression of miR‐200c impedes metastasis formation in vivo and migration in vitro and highlights miR‐200c as a metastasis suppressor in breast cancer.
... Aggregating the clonal status of all driver point mutations over time reveals an increased diversity of driver genes mutated at later stages of tumour development: 50% of all early clonal driver mutations occur in just 9 genes, whereas 50% of late and subclonal mutations occur in approximately 35 different genes each, a nearly fourfold increase (Fig. 2d). Consistent with previous studies of individual tumour types [31][32][33][34] , these results suggest that, in general, the very early events in cancer evolution occur in a constrained set of common drivers, and a more diverse array of drivers is involved in late tumour development. ...
Article
Full-text available
Cancer progression is a process of somatic evolution1,2. Sequencing data from a single biopsy represent a snapshot of this process, revealing the timing of specific genomic aberrations and the changing impact of mutational processes3. Here, by analyzing whole-genome sequencing of 2,658 cancers as part of the Pan-Cancer Analysis of Whole Genomes (PCAWG) consortium of the International Cancer Genome Consortium (ICGC) and The Cancer Genome Atlas (TCGA)4, we reconstruct the life history and mutational processes of 38 cancers and the sequence of driver mutations. Early tumorigenesis is characterized by mutations in a limited set of driver genes and specific copy number gains, such as trisomy 7 in glioblastoma and chromosome 17q in medulloblastoma. In 40% of samples, the mutational spectrum changes dramatically throughout tumor evolution. Advanced stages are characterized by a nearly fourfold increase in driver gene diversification and increased genomic instability. Copy number variations often occur during mitotic crisis and result in concurrent gains in chromosomal fragments. Temporal analysis shows that driver mutations often precede diagnosis by many years, even decades. Together, these results determine the evolutionary trajectory of cancer and highlight opportunities for early cancer detection.
Article
Full-text available
Although metastatic disease is the leading cause of cancer-related deaths, its tumor microenvironment remains poorly characterized due to technical and biospecimen limitations. In this study, we assembled a multi-modal spatial and cellular map of 67 tumor biopsies from 60 patients with metastatic breast cancer across diverse clinicopathological features and nine anatomic sites with detailed clinical annotations. We combined single-cell or single-nucleus RNA sequencing for all biopsies with a panel of four spatial expression assays (Slide-seq, MERFISH, ExSeq and CODEX) and H&E staining of consecutive serial sections from up to 15 of these biopsies. We leveraged the coupled measurements to provide reference points for the utility and integration of different experimental techniques and used them to assess variability in cell type composition and expression as well as emerging spatial expression characteristics across clinicopathological and methodological diversity. Finally, we assessed spatial expression and co-localization features of macrophage populations, characterized three distinct spatial phenotypes of epithelial-to-mesenchymal transition and identified expression programs associated with local T cell infiltration versus exclusion, showcasing the potential of clinically relevant discovery in such maps.
Conference Paper
Full-text available
The type and genomic context of cancer mutations depend on their causes. These causes have been characterized using signatures that represent mutation types that co-occur in the same tumours. However, it remains unclear how mutation processes change during cancer evolution due to the lack of reliable methods to reconstruct evolutionary trajectories of mutational signature activity. Here, as part of the ICGC/TCGA Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium, which aggregated whole-genome sequencing data from 2658 cancers across 38 tumour types, we present TrackSig, a new method that reconstructs these trajectories using optimal, joint segmentation and deconvolution of mutation type and allele frequencies from a single tumour sample. In simulations, we find TrackSig has a 3–5% activity reconstruction error, and 12% false detection rate. It outperforms an aggressive baseline in situations with branching evolution, CNA gain, and neutral mutations. Applied to data from 2658 tumours and 38 cancer types, TrackSig permits pan-cancer insight into evolutionary changes in mutational processes.
Conference Paper
Full-text available
The type and genomic context of cancer mutations depend on their causes. These causes have been characterized using signatures that represent mutation types that co-occur in the same tumours. However, it remains unclear how mutation processes change during cancer evolution due to the lack of reliable methods to reconstruct evolutionary trajectories of mutational signature activity. Here, as part of the ICGC/TCGA Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium, which aggregated whole-genome sequencing data from 2658 cancers across 38 tumour types, we present TrackSig, a new method that reconstructs these trajectories using optimal, joint segmentation and deconvolution of mutation type and allele frequencies from a single tumour sample. In simulations, we find TrackSig has a 3–5% activity reconstruction error, and 12% false detection rate. It outperforms an aggressive baseline in situations with branching evolution, CNA gain, and neutral mutations. Applied to data from 2658 tumours and 38 cancer types, TrackSig permits pan-cancer insight into evolutionary changes in mutational processes.
Article
Breast cancer is the most common cancer in women and one of the deadliest cancers worldwide. According to the distribution of tumor tissue, breast cancer can be divided into invasive and non-invasive forms. The cancer cells in invasive breast cancer pass through the breast and through the immune system or systemic circulation to different parts of the body, forming metastatic breast cancer. Drug resistance and distant metastasis are the main causes of death from breast cancer. Research on breast cancer has attracted extensive attention from researchers. In vitro construction of tumor models by tissue engineering methods is a common tool for studying cancer mechanisms and anticancer drug screening. The tumor microenvironment consists of cancer cells and various types of stromal cells, including fibroblasts, endothelial cells, mesenchymal cells, and immune cells embedded in the extracellular matrix. The extracellular matrix contains fibrin proteins (such as types I, II, III, IV, VI, and X collagen and elastin) and glycoproteins (such as proteoglycan, laminin, and fibronectin), which are involved in cell signaling and binding of growth factors. The current traditional two-dimensional (2D) tumor models are limited by the growth environment and often cannot accurately reproduce the heterogeneity and complexity of tumor tissues in vivo. Therefore, in recent years, research on three-dimensional (3D) tumor models has gradually increased, especially 3D bioprinting models with high precision and repeatability. Compared with a 2D model, the 3D environment can better simulate the complex extracellular matrix components and structures in the tumor microenvironment. Three-dimensional models are often used as a bridge between 2D cellular level experiments and animal experiments. Acellular matrix, gelatin, sodium alginate, and other natural materials are widely used in the construction of tumor models because of their excellent biocompatibility and non-immune rejection. Here, we review various natural scaffold materials and construction methods involved in 3D tissue-engineered tumor models, as a reference for research in the field of breast cancer.
Article
Full-text available
Several studies using genome-wide molecular techniques have reported various degrees of genetic heterogeneity between primary tumours and their distant metastases. However, it has been difficult to discern patterns of dissemination owing to the limited number of patients and available metastases. Here, we use phylogenetic techniques on data generated using whole-exome sequencing and copy number profiling of primary and multiple-matched metastatic tumours from ten autopsied patients to infer the evolutionary history of breast cancer progression. We observed two modes of disease progression. In some patients, all distant metastases cluster on a branch separate from their primary lesion. Clonal frequency analyses of somatic mutations show that the metastases have a monoclonal origin and descend from a common ‘metastatic precursor’. Alternatively, multiple metastatic lesions are seeded from different clones present within the primary tumour. We further show that a metastasis can be horizontally cross-seeded. These findings provide insights into breast cancer dissemination.
Article
Full-text available
Background Understanding the cancer genome is seen as a key step in improving outcomes for cancer patients. Genomic assays are emerging as a possible avenue to personalised medicine in breast cancer. However, evolution of the cancer genome during the natural history of breast cancer is largely unknown, as is the profile of disease at death. We sought to study in detail these aspects of advanced breast cancers that have resulted in lethal disease. Methods and Findings Three patients with oestrogen-receptor (ER)-positive, human epidermal growth factor receptor 2 (HER2)-negative breast cancer and one patient with triple negative breast cancer underwent rapid autopsy as part of an institutional prospective community-based rapid autopsy program (CASCADE). Cases represented a range of management problems in breast cancer, including late relapse after early stage disease, de novo metastatic disease, discordant disease response, and disease refractory to treatment. Between 5 and 12 metastatic sites were collected at autopsy together with available primary tumours and longitudinal metastatic biopsies taken during life. Samples underwent paired tumour-normal whole exome sequencing and single nucleotide polymorphism (SNP) arrays. Subclonal architectures were inferred by jointly analysing all samples from each patient. Mutations were validated using high depth amplicon sequencing. Between cases, there were significant differences in mutational burden, driver mutations, mutational processes, and copy number variation. Within each case, we found dramatic heterogeneity in subclonal structure from primary to metastatic disease and between metastatic sites, such that no single lesion captured the breadth of disease. Metastatic cross-seeding was found in each case, and treatment drove subclonal diversification. Subclones displayed parallel evolution of treatment resistance in some cases and apparent augmentation of key oncogenic drivers as an alternative resistance mechanism. We also observed the role of mutational processes in subclonal evolution. Limitations of this study include the potential for bias introduced by joint analysis of formalin-fixed archival specimens with fresh specimens and the difficulties in resolving subclones with whole exome sequencing. Other alterations that could define subclones such as structural variants or epigenetic modifications were not assessed. Conclusions This study highlights various mechanisms that shape the genome of metastatic breast cancer and the value of studying advanced disease in detail. Treatment drives significant genomic heterogeneity in breast cancers which has implications for disease monitoring and treatment selection in the personalised medicine paradigm.
Article
Full-text available
Background Single-cell micro-metastases of solid tumors often occur in the bone marrow. These disseminated tumor cells (DTCs) may resist therapy and lay dormant or progress to cause overt bone and visceral metastases. The molecular nature of DTCs remains elusive, as well as when and from where in the tumor they originate. Here, we apply single-cell sequencing to identify and trace the origin of DTCs in breast cancer. Results We sequence the genomes of 63 single cells isolated from six non-metastatic breast cancer patients. By comparing the cells’ DNA copy number aberration (CNA) landscapes with those of the primary tumors and lymph node metastasis, we establish that 53% of the single cells morphologically classified as tumor cells are DTCs disseminating from the observed tumor. The remaining cells represent either non-aberrant “normal” cells or “aberrant cells of unknown origin” that have CNA landscapes discordant from the tumor. Further analyses suggest that the prevalence of aberrant cells of unknown origin is age-dependent and that at least a subset is hematopoietic in origin. Evolutionary reconstruction analysis of bulk tumor and DTC genomes enables ordering of CNA events in molecular pseudo-time and traced the origin of the DTCs to either the main tumor clone, primary tumor subclones, or subclones in an axillary lymph node metastasis. Conclusions Single-cell sequencing of bone marrow epithelial-like cells, in parallel with intra-tumor genetic heterogeneity profiling from bulk DNA, is a powerful approach to identify and study DTCs, yielding insight into metastatic processes. A heterogeneous population of CNA-positive cells is present in the bone marrow of non-metastatic breast cancer patients, only part of which are derived from the observed tumor lineages. Electronic supplementary material The online version of this article (doi:10.1186/s13059-016-1109-7) contains supplementary material, which is available to authorized users.
Article
Full-text available
Background: Metastasis is the main cause of cancer patient deaths and remains a poorly characterized process. It is still unclear when in tumor progression the ability to metastasize arises and whether this ability is inherent to the primary tumor or is acquired well after primary tumor formation. Next-generation sequencing and analytical methods to define clonal heterogeneity provide a means for identifying genetic events and the temporal relationships between these events in the primary and metastatic tumors within an individual. Methods and findings: We performed DNA whole genome and mRNA sequencing on two primary tumors, each with either four or five distinct tissue site-specific metastases, from two individuals with triple-negative/basal-like breast cancers. As evidenced by their case histories, each patient had an aggressive disease course with abbreviated survival. In each patient, the overall gene expression signatures, DNA copy number patterns, and somatic mutation patterns were highly similar across each primary tumor and its associated metastases. Almost every mutation found in the primary was found in a metastasis (for the two patients, 52/54 and 75/75). Many of these mutations were found in every tumor (11/54 and 65/75, respectively). In addition, each metastasis had fewer metastatic-specific events and shared at least 50% of its somatic mutation repertoire with the primary tumor, and all samples from each patient grouped together by gene expression clustering analysis. TP53 was the only mutated gene in common between both patients and was present in every tumor in this study. Strikingly, each metastasis resulted from multiclonal seeding instead of from a single cell of origin, and few of the new mutations, present only in the metastases, were expressed in mRNAs. Because of the clinical differences between these two patients and the small sample size of our study, the generalizability of these findings will need to be further examined in larger cohorts of patients. Conclusions: Our findings suggest that multiclonal seeding may be common amongst basal-like breast cancers. In these two patients, mutations and DNA copy number changes in the primary tumors appear to have had a biologic impact on metastatic potential, whereas mutations arising in the metastases were much more likely to be passengers.
Article
Full-text available
Pre-surgical studies allow study of the relationship between mutations and response of oestrogen receptor-positive (ER þ) breast cancer to aromatase inhibitors (AIs) but have been limited to small biopsies. Here in phase I of this study, we perform exome sequencing on baseline, surgical core-cuts and blood from 60 patients (40 AI treated, 20 controls). In poor responders (based on Ki67 change), we find significantly more somatic mutations than good responders. Subclones exclusive to baseline or surgical cores occur in B30% of tumours. In phase II, we combine targeted sequencing on another 28 treated patients with phase I. We find six genes frequently mutated: PIK3CA, TP53, CDH1, MLL3, ABCA13 and FLG with 71% concordance between paired cores. TP53 mutations are associated with poor response. We conclude that multiple biopsies are essential for confident mutational profiling of ER þ breast cancer and TP53 mutations are associated with resistance to oestrogen deprivation therapy.
Article
Tumor molecular profiling is a fundamental component of precision oncology, enabling the identification of genomic alterations in genes and pathways that can be targeted therapeutically. The existence of recurrent targetable alterations across distinct histologically defined tumor types, coupled with an expanding portfolio of molecularly targeted therapies, demands flexible and comprehensive approaches to profile clinically relevant genes across the full spectrum of cancers. We established a large-scale, prospective clinical sequencing initiative using a comprehensive assay, MSK-IMPACT, through which we have compiled tumor and matched normal sequence data from a unique cohort of more than 10,000 patients with advanced cancer and available pathological and clinical annotations. Using these data, we identified clinically relevant somatic mutations, novel noncoding alterations, and mutational signatures that were shared by common and rare tumor types. Patients were enrolled on genomically matched clinical trials at a rate of 11%. To enable discovery of novel biomarkers and deeper investigation into rare alterations and tumor types, all results are publicly accessible.
Article
Human cells have twenty-three pairs of chromosomes. In cancer, however, genes can be amplified in chromosomes or in circular extrachromosomal DNA (ecDNA), although the frequency and functional importance of ecDNA are not understood. We performed whole-genome sequencing, structural modelling and cytogenetic analyses of 17 different cancer types, including analysis of the structure and function of chromosomes during metaphase of 2,572 dividing cells, and developed a software package called ECdetect to conduct unbiased, integrated ecDNA detection and analysis. Here we show that ecDNA was found in nearly half of human cancers; its frequency varied by tumour type, but it was almost never found in normal cells. Driver oncogenes were amplified most commonly in ecDNA, thereby increasing transcript level. Mathematical modelling predicted that ecDNA amplification would increase oncogene copy number and intratumoural heterogeneity more effectively than chromosomal amplification. We validated these predictions by quantitative analyses of cancer samples. The results presented here suggest that ecDNA contributes to accelerated evolution in cancer.
Article
Pre-surgical studies allow study of the relationship between mutations and response of oestrogen receptor-positive (ER+) breast cancer to aromatase inhibitors (AIs) but have been limited to small biopsies. Here in phase I of this study, we perform exome sequencing on baseline, surgical core-cuts and blood from 60 patients (40 AI treated, 20 controls). In poor responders (based on Ki67 change), we find significantly more somatic mutations than good responders. Subclones exclusive to baseline or surgical cores occur in ∼30% of tumours. In phase II, we combine targeted sequencing on another 28 treated patients with phase I. We find six genes frequently mutated: PIK3CA, TP53, CDH1, MLL3, ABCA13 and FLG with 71% concordance between paired cores. TP53 mutations are associated with poor response. We conclude that multiple biopsies are essential for confident mutational profiling of ER+ breast cancer and TP53 mutations are associated with resistance to oestrogen deprivation therapy.