Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

ARTICLE

https://doi.org/10.1038/s41467-020-17249-7 OPEN

Comprehensive characterization of claudin-low


breast tumors reflects the impact of the cell-of-
origin on cancer evolution
Roxane M. Pommier 1,2,3, Amélien Sanlaville 1,2, Laurie Tonon3, Janice Kielbassa3, Emilie Thomas3,
Anthony Ferrari3, Anne-Sophie Sertier3, Frédéric Hollande 4, Pierre Martinez1,2, Agnès Tissier 1,2,
Anne-Pierre Morel1,2, Maria Ouzounova 1,2 & Alain Puisieux 1,2,5 ✉
1234567890():,;

Claudin-low breast cancers are aggressive tumors defined by the low expression of key
components of cellular junctions, associated with mesenchymal and stemness features.
Although they are generally considered as the most primitive breast malignancies, their
histogenesis remains elusive. Here we show that this molecular subtype of breast cancers
exhibits a significant diversity, comprising three main subgroups that emerge from unique
evolutionary processes. Genetic, gene methylation and gene expression analyses reveal that
two of the subgroups relate, respectively, to luminal breast cancers and basal-like breast
cancers through the activation of an EMT process over the course of tumor progression. The
third subgroup is closely related to normal human mammary stem cells. This unique sub-
group of breast cancers shows a paucity of genomic aberrations and a low frequency of TP53
mutations, supporting the emerging notion that the intrinsic properties of the cell-of-origin
constitute a major determinant of the genetic history of tumorigenesis.

1 Cancer Research Center of Lyon, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Equipe Labellisée
Ligue contre le Cancer, 69008 Lyon, France. 2 LabEx DEVweCAN, Université de Lyon, F-69000 Lyon, France. 3 Fondation Synergie Lyon Cancer, Plateforme
de bioinformatique Gilles Thomas, Centre Léon Bérard, Lyon, France. 4 Department of Clinical Pathology, The University of Melbourne, Victorian
Comprehensive Cancer Centre, Parkville, Victoria, Australia. 5 Institut Curie, PSL Research University, Paris, France. ✉email: alain.puisieux@curie.fr

NATURE COMMUNICATIONS | (2020)11:3431 | https://doi.org/10.1038/s41467-020-17249-7 | www.nature.com/naturecommunications 1


ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-17249-7

B
reast cancer is a highly heterogeneous group of diseases Results
with variable biological and clinical behaviors. Providing Selection of claudin-low tumors. Claudin-low tumors exhibit
early insights into this diversity, gene-expression profiling marked immune and stromal cell infiltration, when compared
analyses initially resulted in the identification of four clinically with all other breast cancer subtypes4,19. As gene expression
relevant molecular subtypes, known as intrinsic subtypes (luminal analyses do not allow to accurately discriminate mesenchymal
A, luminal B, HER2-enriched and basal-like, according to the cancer cells from normal stromal cells, we used the allele-specific
PAM50 classification) mostly corresponding to hormone receptor copy number analysis of tumors (ASCAT) copy number-based
and HER2 status, and a normal breast-like group1–4. Among tumor purity estimation method to assess tumor cell fraction20
these intrinsic subtypes, the basal-like subtype appears to be the (Supplementary Fig. 1a). To avoid any substantial bias due to
most distinct, as it is characterized by the unique expression of non-tumor cell contamination, a stringent purity threshold was
cytokeratins typically expressed by the basal layer of the skin and determined by applying Wilcoxon test, for which the degree of
a very low level of expression of luminal-related genes2,5. This contamination of claudin-low tumors was not statistically dif-
observation led to the hypothesis that breast cancers may arise ferent from that of non-claudin-low tumors, thus enabling a
from the transformation of two distinct cell types of origin or comparable distribution of tumors (Supplementary Fig. 1b).
developmental stages of mammary epithelial cell development, Using this stringent threshold, 45 out of 152 tumors classified as
one generating basal-like tumors and the other non-basal-like claudin-low from Molecular Taxonomy of Breast Cancer Inter-
malignancies5. Although appealing in its simplicity, this model national Consortium (METABRIC) were selected for further
overlooks the potential reprogramming of lineage-restricted studies. Demonstrating an unexpected degree of diversity, only
populations initiated by an oncogenic event or a microenviron- 35.6% of these tumors were classified as TNBCs (Supplementary
mental signal over the course of tumor development6–8. More- Fig. 1d). This result was not due to the purity selection, as the
over, it does not account for the intrinsic diversity of each breast percentage of TNBCs within all claudin-low tumors was similar
cancer subtype, notably basal-like tumors known for their great before applying the purity threshold (Supplementary Fig. 1c). Of
heterogeneity9–11. An additional intrinsic subtype of breast can- note, only four normal-like tumors were estimated as pure. Due
cers, known as claudin-low, has recently been identified in human to the lack of statistical power, they were excluded from sub-
and mouse tumors and in breast cancer cell lines, showing several sequent analyses.
common features with basal-like tumors and reflecting the
diversity of tumors with a low luminal differentiation status4,12.
Basal-like and claudin-low tumors form the majority of triple- Identification of CNA-devoid claudin-low tumors. We next
negative breast cancers (TNBCs), an aggressive subgroup of analyzed claudin-low tumors using the stratification based on
breast malignancies defined as tumors lacking expression of the gene expression and copy number alterations (CNAs), previously
estrogen receptor (ER), progesterone receptor (PR), and HER2. A defined by Curtis et al.21,22 (Fig. 1a). As expected for aggressive
hallmark of the claudin-low subtype is the low expression level of neoplasms, basal-like tumors mostly belonged to the integrative
critical cell–cell adhesion molecules, including claudins 3, 4, and cluster 10, characterized by multiple CNAs affecting most chro-
7, occludin, and E-cadherin. Tumors of this subtype are highly mosomes. Luminal A and B tumors were distributed across
enriched in mesenchymal traits and stem cell features and are clusters 1, 2, 3, 6, 7, 8, and 9, whereas HER2-positive tumors were
therefore considered as the most primitive breast cancers13. mainly found in cluster 5. Interestingly, claudin-low tumors were
Although in vivo preclinical data suggested that basal-like tumors stratified into three main clusters, again demonstrating a sig-
arise from the transformation of a luminal progenitor (LP)14, the nificant heterogeneity. Only 17.8% were found in cluster 10,
putative cell-of-origin of claudin-low tumors remains unknown. whereas 48.8% and 17.8% were found in integrative clusters 4 and
The prevailing hypothesis is that, over the course of tumor pro- 3, respectively, regardless of tumor purity when compared with
gression, basal cancer cells undergo an epithelial-mesenchymal non-claudin-low tumors (Supplementary Fig. 2). Both integrative
transition (EMT) in response to acquired oncogenic events and/ clusters 3 and 4 are marked by a paucity of copy number and cis-
or microenvironmental signals, thereby gaining mesenchymal acting alterations. Integrative cluster 3 displays a simplex pattern
and stemness features15–17. An alternative hypothesis is that of rearrangements, dominated by whole arm gain of chromosome
claudin-low tumors originate from an early epithelial precursor 1q and 16p and loss of 16q11,21,22. It was initially described as
with inherent stemness features4,13. To gain further insight into essentially composed of luminal breast tumors. Integrative cluster
the developmental origin of claudin-low tumors, we first sought 4 includes both ER-positive and ER-negative cases. It was defined
to analyze their genomic architecture. Indeed, we have recently as a CNA-devoid subgroup, with an expression profile dominated
demonstrated that, whereas the oncogene-driven transformation by immune-related genes, leading to the hypothesis that it may be
of mature luminal (mL) cells and progenitor luminal cells triggers essentially composed of samples with a high infiltration of normal
massive oncogene-induced DNA damage and an early onset of cells11. As the method used in the present study to select tumors
chromosomal instability (CIN), normal human mammary stem with a high index of purity does not rely on gene expression
cells (MaSCs) can withstand an aberrant mitogenic activity18. The patterns, it argues against this hypothesis. Nevertheless, to for-
endogenous expression of the ZEB1 EMT-inducing transcription mally demonstrate the existence of claudin-low tumors with a flat
factor prevents replication and oxidative stress, leading to a genomic landscape, we analyzed the genomic architecture of
process of malignant transformation in the absence of exacer- tumor samples exhibiting a very low level of aberrations (fraction
bated genomic instability18. As basal-like breast cancers generally of genome altered, FGA < 1%). After having previously removed
exhibit numerous genomic aberrations, we thus speculated that germline copy number variations (CNVs), we analyzed B allele
the extent of genomic aberrations might be used as a molecular frequency (BAF) and log R ratio (LRR) profiles of single-
indicator of the developmental origin of claudin-low breast nucleotide polymorphisms (SNPs) localized in genomic regions
cancers. of deletion (LRR segment mean cutoff value < −0.4) (Fig. 1b and
To comprehensively characterize the claudin-low breast tumor Supplementary Fig. 3a, c). This analysis led to the identification of
subtype, we used a multi-omics approach that reveal the existence regions of single-copy allelic loss displaying a clear separation
of three distinct subgroups with specific transcriptomic, epige- between BAF values (<0.8 and >0.2). As previously demonstrated,
netic, and genetic characteristics, strongly supporting the this observation is incompatible with a massive contamination of
hypothesis of different cells-of-origin. the tumor tissue by normal cells23. Consistent with these data, the

2 NATURE COMMUNICATIONS | (2020)11:3431 | https://doi.org/10.1038/s41467-020-17249-7 | www.nature.com/naturecommunications


NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-17249-7 ARTICLE

a 300 All tumors 25 Claudin-low tumors b 1.0


48.8%
250 19.3% 0.8
17.5% 20

BAF
0.6
Number of tumors

200
13.8% 15 0.4

150 0.2
9.3% 9.9%
8.1% 10
7.6% 17.8% 17.8% 0.0
100
5.9%

Chr 10

Chr 11

Chr 12

Chr 13

Chr 14

Chr 15
Chr 16
Chr 17
Chr 18
Chr 19
Chr 20
Chr 21
Chr 22
2

Chr 1

Chr 2

Chr 3

Chr 4

Chr 5

Chr 6

Chr 7

Chr 8

Chr 9
4.7%
4.1% 5
50 6.7% pos
2.2% 2.2% 2.2% 2.2% 1
0% 0%
0 0

LRR
iC1 iC2 iC3 iC4 iC5 iC6 iC7 iC8 iC9 iC10 iC1 iC2 iC3 iC4 iC5 iC6 iC7 iC8 iC9 iC10 0

125 Basal tumors 70 HER2 tumors –1


82.9% 59.8%
60
100 –2

50 0.0e + 00 5.0e + 08 1.0e + 09 1.5e + 09 2.0e + 09 2.5e + 09


Number of tumors

75
40

30 Chromosome 7 Chromosome 20 Chromosome 20


50
1.0 1.0 1.0
20
13.8% 0.8 0.8 0.8
25
10 5.9% 8.8%

BAF
BAF
9.3%

BAF
0.6 0.6 0.6
3.9% 2.3% 3.9% 2.9% 2.9% 2.0%
1.6% 0% 0%
0% 0% 0% 0% 0%
0 0 0.4 0.4 0.4
iC1 iC2 iC3 iC4 iC5 iC6 iC7 iC8 iC9 iC10 iC1 iC2 iC3 iC4 iC5 iC6 iC7 iC8 iC9 iC10
0.2 0.2 0.2

175 Luminal A tumors 125 Luminal B tumors 0.0 0.0 0.0


21.7%
33.4% 0.5
150 0.4
100 pos pos 0.5
27.3% 0.2
125
Number of tumors

14.9% 0.0
14.4% 0.0 0.0

LRR

LRR
LRR
100 20.2% 75 13.0%

10.4% –0.4 –0.5 –0.5


75 9.3%
50 8.1%
11.9%
–1.0
50
5.1% –0.8 –1.0
25
25 154,020,000 154,080,000 14,500,000 14,650,000 14,800,000 53,000,000 53,400,000 53,800,000
1.7% 1.5% 2.0% 1.3% 1.5% 1.5%
0.7% 0%
0 0
iC1 iC2 iC3 iC4 iC5 iC6 iC7 iC8 iC9 iC10 iC1 iC2 iC3 iC4 iC5 iC6 iC7 iC8 iC9 iC10 c Gene Mutation type Chromosome Exon Reference allele Altered allele vaf FGA (%) ASCAT purity
Integrative clusters (IntClust) Integrative clusters (IntClust) CHD1 Missense SNV chr5 exon34 C T 0.48
KMT2C Missense SNV chr7 exon14 G T 0.14
KMT2C Missense SNV chr7 exon43 G T 0.42 0.29% 0.75
Luminal-related clusters Claudin-low-related cluster 4
AHNAK2 Missense SNV chr14 exon7 T C 0.15
Her2-related cluster 5 Basal-related cluster 10 DNAH2 Missense SNV chr17 exon42 A T 0.49

Fig. 1 Claudin-low tumors are distributed across luminal-like IntClust3, basal-like IntClust10, and CNA-devoid IntClust4, which harbors rare focal
genomic alterations. a Repartition of breast tumors from the METABRIC cohort for each molecular subtype among integrative clusters. Number of tumors
are represented on the y axis and the corresponding percentages are indicated above each bar. All tumors (n = 1266); claudin-low tumors (n = 45); basal
tumors (n = 129); HER2 tumors (n = 102); luminal A tumors (n = 461); luminal B tumors (n = 429). b, c Identification of focal genomic alterations in a
CNA-devoid IntClust4 claudin-low sample (MB-6052/FGA < 0.3%). b BAF and LRR plots of SNPs localized in genomic regions of copy number deletion
(LRR < 0.4). c Mutation analysis from targeted sequencing data. BAF: B allele frequency; CNA: copy number alteration; FGA: fraction of genome altered;
IntClust: integrative cluster; LRR: log R ratio; SNP: single-nucleotide polymorphism.

analysis of somatic mutations highlighted the presence of several note, although these analyses were performed on tumors selected
point mutations with a variant allele frequency higher than 0.4 for their high tumor purity, similar data were obtained when
(Fig. 1c and Supplementary Fig. 3b). These two results unequi- studying the whole cohort of claudin-low tumors (Supplementary
vocally demonstrate the existence of claudin-low tumors that Fig. 4), further strengthening the characterization of the three
develop in the absence of gross CIN. claudin-low subgroups.

Genomic diversity of claudin-low tumors. Although most Distinct gene expression signatures of claudin-low subgroups.
claudin-low tumors are diploid (Fig. 2a), the extent of FGA was Gene-set enrichment analysis (GSEA) was next used to identify
highly variable across tumors (Fig. 2b), confirming their intrinsic pathways differentially regulated among the three claudin-low
diversity24,25. The use of Gaussian finite mixture models revealed subgroups, by comparing one with the other two (CL1 vs. CL2/3,
a trimodal distribution, reflecting the existence of three subgroups CL2 vs. CL1/3, and CL3 vs. CL1/2). More than 15,000 pathways,
of claudin-low tumors relative to their level of FGA (Fig. 2c, d). available in H (Hallmarks), C2 (encompassing curated gene sets
The subgroup 1 of claudin-low tumors (CL1), defined by a low from literature and canonical pathways such as KEGG, REAC-
FGA (<10%), was mostly composed of ER-negative tumors that TOME, or BIOCARTA), and C5 (comprising GO pathways) gene
were all stratified into integrative cluster 4 (Fig. 2d–f). The sub- sets from Molecular Signature Database (MSigDB), were tested.
group 2 (CL2) showed an intermediate level of FGA (>10% and These analyses highlighted as follows: (i) an enrichment in stem
<30%), similar to that of luminal A breast tumors. Consistent cell-related signatures in CL1, (ii) an enrichment in luminal-
with this finding, CL2 was mostly composed of ER-positive related signatures in CL2, and (iii) an enrichment in cell cycle-
tumors mainly stratified in two luminal-related clusters, whereas related pathways in CL3 tumors (Fig. 3a). Of note, global gene
35% of CL2 tumors belonged to cluster 4. The subgroup 3 (CL3) expression signatures demonstrated that the CL2 and
displayed a high FGA (>30%), similar to the one found in basal- CL3 subgroups showed lower luminal- and basal-related sig-
like breast cancers. Consistent with this notion, 33% of CL3 nature scores, respectively, compared with luminal- and basal-like
tumors were classified into the genomically unstable cluster 10. breast tumors, while exhibiting a significant enrichment in
However, this subgroup was heterogeneous, with both ER- invasiveness-related signatures (Supplementary Fig. 5a, b). We
negative or ER-positive tumors, and a significant fraction of CL3 then performed single-sample GSEA (ssGSEA) analyses to
tumors were found in luminal-related clusters and in cluster 4. Of determine the differentiation profile of the three claudin-low

NATURE COMMUNICATIONS | (2020)11:3431 | https://doi.org/10.1038/s41467-020-17249-7 | www.nature.com/naturecommunications 3


ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-17249-7

a b c
6 100 0.150 CL1 tumors 10
90 CL2 tumors
0.125 CL3 tumors
5 80 8
70
0.100

Frequency
Density
4 60 6

FGA (%)
Ploidy

Density
50 0.075

3 40 4
0.050
30
2
2 20 0.025

10
0.000 0
1 0
CL Basal Her2 LumA LumB CL Basal Her2 LumA LumB 0 10 20 30 40 50 60 70 80
FGA (%)

d e Luminal-related clusters Basal-related cluster 10 f


Claudin-low-related cluster 4 Her2-related cluster 5 TN ER/PR+ HER2+
100 100 100
6%
12% 11% 12%
90
80 80 80
Percentage of tumors

Percentage of tumors
70 33.3% 22%
60 60 60
FGA (%)

50 41%
40 40 53% 33.3% 40
30
76%
20 20 20
10
100% 35% 33.3% 66% 18% 47%
0 0 0
CL1 CL2 CL3 Basal Her2 LumA LumB CL1 CL2 CL3 CL1 CL2 CL3

Fig. 2 Claudin-low tumors form a genomically and molecularly heterogeneous subgroup. a Ploidy for each molecular subtype of the METABRIC cohort.
Claudin-low tumors are mainly diploid. b FGA% for each molecular subtype. Claudin-low tumors display overall low levels of genomic alterations. c
Gaussian mixture model application on FGA distribution across claudin-low tumors reveals three CNA-related subgroups of tumors. Each bar on the x axis
corresponds to one claudin-low tumor. d FGA% in claudin-low subgroups compared with other molecular subtypes. e Integrative clusters and f breast
cancer receptor status distribution in each claudin-low subgroup. The CNA-devoid CL1 subgroup is associated with IntClust4 and TN status, whereas CL2
and CL3 are respectively related to luminal (luminal clusters and hormone receptor expression) and basal-like (IntClust10 and TN tumor enrichment)
subtypes. Respective colors of luminal-related clusters are shown in Fig. 1. Boxplot: center line, median; box limits, upper and lower quartiles; whiskers,
minimum to maximum; all data points are shown. Claudin-low tumors (n = 45); basal tumors (n = 129); HER2 tumors (n = 102); luminal A tumors (n =
461); luminal B tumors (n = 429); CL1 tumors (n = 10); CL2 tumors (n = 17); CL3 tumors (n = 18).CL: claudin-low; TN: triple negative.

subgroups. Gene expression signatures for MaSCs, LPs, and mL lines showed low, moderate, and elevated FGA levels, respectively
cells were generated based on the transcription profiles of sub- (Supplementary Fig. 6e, f). Of note, we performed a pathway
populations of normal human mammary epithelial cells isolated analysis of CL1 discriminant genes that revealed an enrichment in
from reduction mammoplasty tissues, as shown in Morel et al.18 stem cell and pediatric cancer markers (Supplementary Fig. 6g).
(Supplementary Data 1). Consistent with previous GSEA analysis,
CL1 tumors showed a strong enrichment in the MaSC signature,
whereas CL2 tumors exhibited a mL signature (Fig. 3b). Again, Distinct gene methylation profiles of claudin-low subgroups.
CL3 tumors showed a significant heterogeneity with a trend Differentially methylated gene analysis was next performed in
toward the LP signature. Notably, very similar data were obtained claudin-low subgroups followed by pathway-enrichment analysis,
when using an alternative set of MaSC, LP, and mL signatures14 using over 15,000 pathways available in MSigDB (HALLMARKS,
(Fig. 3c). C2, and C5 gene sets) (Supplementary Fig. 7). When compared
with CL2 and CL3, the CL1 subgroup presented 40 differentially
methylated genes, and among them 75% were hypomethylated
Identification of a gene expression-based classifier. We next and enriched in stemness and EMT markers (Fig. 4a). The
generated a gene expression-based classifier by using the nearest CL2 subgroup encompassed 68 differentially methylated genes
shrunken centroid method26, with the objective of discriminating compared with CL1 and CL3, and among them 88% were
claudin-low subgroups. A claudin-low gene list of 137 genes hypermethylated and enriched in basal-like and EMT pathways
discriminating the 3 claudin-low subgroups was thus created (Fig. 4b). Finally, the CL3 subgroup was characterized by 219
(Supplementary Fig. 6a, b). This expression-based classifier differentially methylated genes compared with CL1 and CL2;
showed an accuracy of 91% when compared with the FGA-based among them, 58% were hypermethylated and related to luminal
classification using Gaussian finite mixture models (Supplemen- pathways and 42% presented a hypomethylation profile and were
tary Fig. 5c, d). The classifier was then applied to claudin-low associated with basal-like pathways (Fig. 4c). To correlate these
samples from The Cancer Genome Atlas Network (TCGA; distinct gene methylation profiles with gene expression, we per-
https://www.cancer.gov/about-nci/organization/ccg/research/ formed a gene expression analysis of stemness and EMT gene
structural-genomics/tcga) and the Cancer Cell Line Encyclopedia markers in all three claudin-low subgroups and in the intrinsic
(CCLE27), and the level of FGA was used as a readout to verify subtypes of breast cancers from METABRIC, TCGA, and CCLE
the characteristics of each claudin-low subgroup. Validating the cohorts. The CL1 subgroup had the highest score for stemness-
accuracy of our classifier, CL1, CL2, and CL3 tumors and cell (ALDH1A1, PROCR) and EMT- (mesenchymal markers: ZEB1,

4 NATURE COMMUNICATIONS | (2020)11:3431 | https://doi.org/10.1038/s41467-020-17249-7 | www.nature.com/naturecommunications


NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-17249-7 ARTICLE

a b 1.0 MaSC1/2/3 signature (from Morel et al. data)


c 1.0 MaSC signature (Lim et al.)
CL1 vs CL2/CL3 - NES ranked top 3 pathways UP: stemness/DOWN: proliferation
Pathway UP Gene ranks NES pval padj
0.5 0.5
SMID_BREAST_CANCER_NORMAL_LIKE_UP 3.53 4.1e 04 3.2e 03

ssGSEA scores

ssGSEA scores
BOQUEST_STEM_CELL_UP 3.26 3.4e 04 2.8e 03
LIM_MAMMARY_STEM_CELL_UP 3.20 4.2e 04 3.2e 03 0.0 0.0

DUTERTRE_ESTRADIOL_RESPONSE_24HR_UP 3.46 1.4e 04 2.1e 03


P = 9.9e-3 P = 1.5e-3
ROSTY_CERVICAL_CANCER_PROLIFERATION_CLUSTER 3.67 1.5e 04 2.1e 03 -0.5 -0.5
SOTIRIOU_BREAST_CANCER_GRADE_1_VS_3_UP 3.74 1.5e 04 2.1e 03
0 5000 10,000 15,000
DOWN -1.0 -1.0
CL1 CL2 CL3 Basal Her2 LumA LumB CL1 CL2 CL3 Basal Her2 LumA LumB

1.0 mL1/2 signature (from Morel et al. data) 1.0 mL signature (Lim et al.)
CL2 vs CL1/CL3 - NES ranked top 3 pathways UP: luminal breast subtype/DOWN: stemness
Pathway UP Gene ranks NES pval padj
0.5 0.5
VANTVEER_BREAST_CANCER_ESR1_UP 3.27 1.8e 04 3.4e 03 P = 8.8e-5 P = 1.1e-3

ssGSEA scores
ssGSEA scores
SMID_BREAST_CANCER_LUMINAL_B_UP 3.11 1.9e 04 3.4e 03
DOANE_BREAST_CANCER_ESR1_UP 3.09 1.9e 04 3.4e 03 0.0 0.0

LIEN_BREAST_CARCINOMA_METAPLASTIC_VS_DUCTAL_UP 3.05 2.1e 04 3.4e 03


SMID_BREAST_CANCER_RELAPSE_IN_BONE_DN 3.07 2.2e 04 3.4e 03 -0.5 -0.5
LIM_MAMMARY_STEM_CELL_UP 3.12 2.3e 04 3.4e 03
0 5000 10,000 1,5000
DOWN
-1.0 -1.0
CL1 CL2 CL3 Basal Her2 LumA LumB CL1 CL2 CL3 Basal Her2 LumA LumB

1.0 LP signature (from Morel et al. data) 1.0 LP signature (Lim et al.)
CL3 vs CL1/CL2 - NES ranked top 3 pathways UP: proliferation/DOWN: translation n.s. n.s.
Pathway UP Gene ranks NES pval padj
0.5 0.5
SOTIRIOU_BREAST_CANCER_GRADE_1_VS_3_UP 3.52 2.1e 04 4.2e 03

ssGSEA scores

ssGSEA scores
ROSTY_CERVICAL_CANCER_PROLIFERATION_CLUSTER 3.43 2.1e 04 4.2e 03
DUTERTRE_ESTRADIOL_RESPONSE_24HR_UP 3.24 2.1e 04 4.2e 03 0.0 0.0

GO_COTRANSLATIONAL_PROTEIN_TARGETING_TO_MEMBRANE 3.10 1.9e 04 4.2e 03


SMID_BREAST_CANCER_NORMAL_LIKE_UP 3.24 1.9e 04 4.2e 03 -0.5 -0.5
KEGG_RIBOSOME 3.24 1.9e 04 4.2e 03
0 5000 10,000 15,000
DOWN -1.0 -1.0
CL1 CL2 CL3 Basal Her2 LumA LumB CL1 CL2 CL3 Basal Her2 LumA LumB

Fig. 3 Claudin-low subgroups show distinct gene expression signatures relative to their differentiation status. a GSEA comparing global gene
expression of each claudin-low subgroup from the METABRIC cohort to the two others. The three pathways with the highest and lowest enrichment scores
are represented (>15,000 tested pathways—NES ranking). b, c Gene expression analysis for each molecular subtype of (b) MaSC1/2/3, LP, and mL1/2
transcriptomic signatures (generated from Morel et al.18 data) and (c) previously published MaSC, LP, and mL transcriptomic signatures (from Lim et al.14
publication). The CL1 subgroup displays strong stemness features, whereas CL2 and CL3 present luminal- and basal-like transcriptomic characteristics,
respectively. Wilcoxon tests. Boxplot: center line, median; box limits, upper and lower quartiles; whiskers, minimum to maximum; all data points are shown.
Basal tumors (n = 129); HER2 tumors (n = 102); luminal A tumors (n = 461); luminal B tumors (n = 429); CL1 tumors (n = 10); CL2 tumors (n = 17); CL3
tumors (n = 18). GSEA: gene-set enrichment analysis; LP: luminal progenitor; MaSC: mammary stem cell; mL: mature luminal; NES: normalized
enrichment score.

VIM, CDH2; epithelial markers: EPCAM, CDH1) related genes, cohorts using two different cellular signature deconvolution
whereas CL2 and CL3 displayed an intermediate score compared algorithms confirmed that these tumors were highly infiltrated by
with CL1 tumors and with their relative luminal/basal counter- immune cells (Supplementary Fig. 9a–d). Nevertheless, unsu-
parts (Supplementary Fig. 8). pervised clustering of microenvironment signatures (immune and
non-immune cell subsets) did not allow to discriminate the dif-
ferent claudin-low subgroups (Supplementary Fig. 9e–h). The
Distinct oncogenic pathways in claudin-low subgroups. To additional pathways specifically enriched in all claudin-low sub-
characterize the biological pathways driving the development of groups included the TP53-dependent pathway, the mitogen-
the three subgroups of claudin-low tumors, we determined, for activated protein kinase (RASMAPK) signaling pathway and a
each breast tumor sample from METABRIC and TCGA cohorts, differentiation/EMT-related pathway (Fig. 5). CL1 presented the
ssGSEA scores for all MSigDB hallmark gene-set signatures. We highest enrichment in claudin-low-related pathways, when
then conducted an unsupervised clustering based on the median compared with all other subtypes. Of note, the CL2 subgroup
score of each of the 48 biological processes computed per breast displayed a concomitant enrichment in claudin-low and luminal
molecular subtype (Fig. 5). Interestingly, in both datasets, the pathways, whereas the CL3 subgroup was enriched in claudin-low
three claudin-low subgroups clustered together in a specific and basal pathways (Fig. 5).
branch of the dendogram, which further discriminated CL1 from To validate these observations, we generated ssGSEA scores for
the CL2/CL3 tumors, highlighting the specific biological processes the pathways previously identified in Fig. 5 (EMT, RAS/MAPK,
distinguishing claudin-low from non-claudin-low and CL1 from proliferation, estrogen, and DNA damage response (DDR)), using
the CL2/CL3 tumors. Furthermore, unsupervised pathway clus- MSigDB C2 curated gene sets (KEGG, GO, REACTOME…). The
tering identified specific biological functions differentially enri- same pathways were also analyzed at the protein level using
ched in claudin-low, basal, and luminal breast tumors. Among reverse-phase protein array (RPPA) data from TCGA28. Support-
the pathways specifically enriched in all claudin-low subgroups, ing our previous results, CL2 and CL3 showed, among the three
some of them were linked to immunity function, supporting the claudin-low subgroups, the highest score for luminal- (estrogen-
hypothesis of privileged infiltration by immune cells within the related response) and basal-related (proliferation and DDR)
microenvironment of claudin-low tumors4,19. Additional gene signatures, respectively (Supplementary Fig. 10). Nevertheless,
expression analyses performed on METABRIC and TCGA statistical analyses revealed a significantly attenuated phenotype

NATURE COMMUNICATIONS | (2020)11:3431 | https://doi.org/10.1038/s41467-020-17249-7 | www.nature.com/naturecommunications 5


ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-17249-7

a
40 differentially methylated genes in CL1vs CL2/CL3 - Welch's t test FDR P-value < 0.001
Hypermethylated genes in CL1 (n = 10/40)
No significantly enriched pathway
Methylation level
(z score)
2
1
0
1
2
CL subgroup Hypomethylated genes in CL1 (n = 30/40) Stemness
CL1 FDR P-value ranked top 3 enriched pathways
CL2
CL3
LIM_MAMMARY_STEM_CELL_UP P = 2.1e-07

GO_CELL_SUBSTRATE_JUNCTION P = 3.2e-05

GO_ANCHORING_JUNCTION P = 6.5e-05

0 2 4 6 8
–log10 (FDR P-value)

b
68 differentially methylated genes in CL2 vs CL1/CL3 - Welch's t test FDR P-value < 0.001
Hypermethylated genes in CL2 (n = 60/68) Basal-like
FDR P-value ranked top 3 enriched pathways

Methylation level SMID_BREAST_CANCER_BASAL_UP P = 4.7e-11


(z score)
2 VANTVEER_BREAST_CANCER_ESR1_DN P = 4.0e-09

1 GO_ANCHORING_JUNCTION P = 5.8e-09
0 0 5 10 15
1 –log10 (FDR P-value)
2
CL subgroup
Hypomethylated genes in CL2 (n = 8/68)
CL1
CL2 no significantly enriched pathway
CL3

c
219 differentially methylated genes in CL3 vs CL1/CL2 - Welch's t test FDR P-value < 0.001
Hypermethylated genes in CL3 (n = 128/219) Luminal
FDR P-value ranked top 3 enriched pathways

Methylation level SMID_BREAST_CANCER_BASAL_DN P = 1.4e-21


(z score)
2 CHARAFE_BREAST_CANCER_LUMINAL_VS_BASAL_UP P = 3.8e-16

1 FARMER_BREAST_CANCER_BASAL_VS_LUMINAL P = 1.1e-14
0 0 5 10 15 20 25
1 –log10 (FDR P-value)
2
CL subgroup Hypomethylated genes in CL3 (n = 91/219) Basal-like
CL1 FDR P-value ranked top 3 enriched pathways
CL2
CL3
SMID_BREAST_CANCER_BASAL_UP P = 2.7e-40

VANTVEER_BREAST_CANCER_ESR1_DN P = 7.9e-20

SMID_BREAST_CANCER_LUMINAL_B_DN P = 1.8e-17

0 10 20 30 40 50
–log10 (FDR P-value)

Fig. 4 Claudin-low subgroups show distinct gene methylation profiles relative to their differentiation status. Heatmaps and pathway-enrichment
analysis of differentially methylated genes between a CL1 vs. CL2 and CL3 tumors from TCGA, b CL2 vs. CL1 and CL3 tumors from TCGA, and c CL3 vs. CL1
and CL2 tumors from TCGA. CL1 tumors mainly display hypomethylated genes that are related to a stemness phenotype, CL2 tumors are overall
hypermethylated for genes associated with basal-like features, and CL3 tumors show more heterogeneous methylation profiles with both hypermethylated
genes (linked to luminal phenotype) and hypomethylated genes (relative to basal-like characteristics) (see Supplementary Fig. 5 for methodology).
Clustering method: Ward’s; distance: Spearman. FDR: false discovery rate.

compared with their related molecular subtype. Furthermore, Consistent with these findings, claudin-low cell lines were
both CL2 and CL3 showed an activation of the EMT process significantly more sensitive to MEK inhibitors (MEKi) than
(Fig. 6a, b, g), coherent with the downregulation of hormone (in non-claudin-low cell lines (Fig. 6h).
CL2) and proliferation/DDR (in CL3) signatures, compared with Altogether, our data strongly support the notion that the
luminal and basal tumors, respectively (Supplementary Fig. 10 CL1 subgroup of claudin-low tumors was related to normal
and Fig. 6g). Moreover, CL1 displayed a low proliferation activity MaSCs, CL2 to normal mL cells and to luminal breast cancers,
and a low DDR (Supplementary Fig. 10a, b, e, f), as well as a and CL3 to basal-like breast cancers. Unstratified claudin-low
strong TP53 signaling pathway signature associated with a low tumors have been previously associated with poor prognosis4,29.
frequency of TP53 mutations (Fig. 6c, d, g). Finally, CL1 exhibited When stratified by CL subgroups, variations in survival outcome
a high activation of the RAS-MAPK signaling pathway, a feature were found, consistent with their presumed origin (Supplemen-
also found in CL2 and CL3, although at a lower level (Fig. 6e–g). tary Fig. 11). Indeed, the basal-like-related CL3 subgroup of

6 NATURE COMMUNICATIONS | (2020)11:3431 | https://doi.org/10.1038/s41467-020-17249-7 | www.nature.com/naturecommunications


NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-17249-7 ARTICLE

a METABRIC Purity Pathway scores b TCGA Purity Pathway scores

0.2 0.4 0.6 0.8 1 2 1 0 1 2 0.2 0.4 0.6 0.8 1 2 1 0 1 2

CL1 CL3 CL2 Basal Her2 LumB LumA Classification CL3 CL2 CL1 Her2 LumB LumA Basal Classification
Purity Purity
HALLMARK_WNT_BETA_CATENIN_SIGNALING HALLMARK_MITOTIC_SPINDLE
HALLMARK_HYPOXIA HALLMARK_PI3K_AKT_MTOR_SIGNALING
HALLMARK_APICAL_JUNCTION HALLMARK_REACTIVE_OXYGEN_SPECIES_PATHWAY
HALLMARK_HEDGEHOG_SIGNALING HALLMARK_CHOLESTEROL_HOMEOSTASIS

Basal pathways
HALLMARK_NOTCH_SIGNALING HALLMARK_DNA_REPAIR

Proliferation/
DNA repair
HALLMARK_INTERFERON_GAMMA_RESPONSE HALLMARK_OXIDATIVE_PHOSPHORYLATION

RAS signaling
HALLMARK_INTERFERON_ALPHA_RESPONSE HALLMARK_UNFOLDED_PROTEIN_RESPONSE

Immunity
HALLMARK_APOPTOSIS HALLMARK_G2M_CHECKPOINT
HALLMARK_INFLAMMATORY_RESPONSE HALLMARK_E2F_TARGETS
HALLMARK_IL2_STAT5_SIGNALING HALLMARK_MTORC1_SIGNALING
HALLMARK_TNFA_SIGNALING_VIA_NFKB HALLMARK_GLYCOLYSIS
CL pathways

HALLMARK_ALLOGRAFT_REJECTION HALLMARK_UV_RESPONSE
HALLMARK_COMPLEMENT HALLMARK_MYC_TARGETS_V2
HALLMARK_KRAS_SIGNALING HALLMARK_MYC_TARGETS_V1

Differentiation/EMT
HALLMARK_IL6_JAK_STAT3_SIGNALING HALLMARK_SPERMATOGENESIS

TP53-related
HALLMARK_HEME_METABOLISM HALLMARK_PROTEIN_SECRETION

Luminal pathways
HALLMARK_MYOGENESIS HALLMARK_ANDROGEN_RESPONSE
HALLMARK_APICAL_SURFACE HALLMARK_XENOBIOTIC_METABOLISM

metabolism
Hormone/
HALLMARK_TGF_BETA_SIGNALING HALLMARK_PEROXISOME
HALLMARK_COAGULATION HALLMARK_FATTY_ACID_METABOLISM
HALLMARK_ANGIOGENESIS HALLMARK_ESTROGEN_RESPONSE_LATE
HALLMARK_EPITHELIAL_MESENCHYMAL_TRANSITION HALLMARK_ESTROGEN_RESPONSE_EARLY
HALLMARK_P53_PATHWAY HALLMARK_ADIPOGENESIS
HALLMARK_ANDROGEN_RESPONSE HALLMARK_BILE_ACID_METABOLISM
HALLMARK_XENOBIOTIC_METABOLISM HALLMARK_PANCREAS_BETA_CELLS
HALLMARK_CHOLESTEROL_HOMEOSTASIS HALLMARK_P53_PATHWAY

RAS signaling TP53-related


HALLMARK_GLYCOLYSIS HALLMARK_MYOGENESIS

Immunity differentiation/EMT
HALLMARK_UNFOLDED_PROTEIN_RESPONSE HALLMARK_APICAL_SURFACE
HALLMARK_MYC_TARGETS_V2 HALLMARK_IL2_STAT5_SIGNALING
Basal pathways

Proliferation/
HALLMARK_DNA_REPAIR HALLMARK_KRAS_SIGNALING

DNA repair
HALLMARK_MTORC1_SIGNALING HALLMARK_ANGIOGENESIS

CL pathways
HALLMARK_G2M_CHECKPOINT HALLMARK_EPITHELIAL_MESENCHYMAL_TRANSITION
HALLMARK_UV_RESPONSE HALLMARK_COAGULATION
HALLMARK_E2F_TARGETS HALLMARK_TGF_BETA_SIGNALING
HALLMARK_MYC_TARGETS_V1 HALLMARK_APOPTOSIS
HALLMARK_SPERMATOGENESIS HALLMARK_HEME_METABOLISM
HALLMARK_MITOTIC_SPINDLE HALLMARK_INTERFERON_GAMMA_RESPONSE
HALLMARK_REACTIVE_OXYGEN_SPECIES_PATHWAY HALLMARK_INTERFERON_ALPHA_RESPONSE
HALLMARK_PI3K_AKT_MTOR_SIGNALING HALLMARK_IL6_JAK_STAT3_SIGNALING
HALLMARK_ESTROGEN_RESPONSE_EARLY HALLMARK_ALLOGRAFT_REJECTION
Luminal pathways

HALLMARK_ESTROGEN_RESPONSE_LATE metabolism HALLMARK_COMPLEMENT


HALLMARK_FATTY_ACID_METABOLISM Hormone/ HALLMARK_INFLAMMATORY_RESPONSE
HALLMARK_PEROXISOME HALLMARK_HYPOXIA
HALLMARK_OXIDATIVE_PHOSPHORYLATION HALLMARK_NOTCH_SIGNALING
HALLMARK_PROTEIN_SECRETION HALLMARK_HEDGEHOG_SIGNALING
HALLMARK_PANCREAS_BETA_CELLS HALLMARK_TNFA_SIGNALING_VIA_NFKB
HALLMARK_BILE_ACID_METABOLISM HALLMARK_APICAL_JUNCTION
HALLMARK_ADIPOGENESIS HALLMARK_WNT_BETA_CATENIN_SIGNALING

Fig. 5 Claudin-low subgroups show distinct activated biological pathways. Heatmaps of ssGSEA scores for each MSigDB hallmark gene-set signature
(median by molecular subtype) for a METABRIC and b TCGA datasets. ASCAT median purity score of each subtype is indicated on the top heat map
annotation. Claudin-low tumors cluster together in a specific branch of the dendrogram downstream distinguishing CL1 from CL2/CL3 tumors. Hallmark
gene-set cluster in three main groups of pathways related to luminal, basal, and claudin-low tumors. ASCAT: allele-specific copy number analysis of
tumors; MSigDB: molecular signature database; ssGSEA: single-sample GSEA. Clustering method: Complete; distance: Euclidean.

claudin-low tumors was associated with low overall and disease- transcriptomic features with the luminal A intrinsic subtype.
free survivals, as compared with the luminal-related Although unexpected, this finding is reminiscent of the experi-
CL2 subgroup. In line with their low proliferation index and mental observation that EMT induction in luminal breast cancer
their low frequency of TP53 mutations, tumors of the cell lines confers them with a claudin-low expression pattern32. A
CL1 subgroup were associated with a favorable prognosis. second degree of heterogeneity is shown by the CL1 subgroup of
claudin-low breast cancers. Among the three subgroups of
Discussion claudin-low tumors, CL1 appears as the most distinct, with dif-
Here we have characterized the intrinsic diversity within the ferential transcriptomic, epigenetic, and genetic traits that com-
claudin-low breast cancers by demonstrating the existence of prise a prominent stemness signature and a paucity of genomic
three main molecular subgroups. Consistent with the data from a aberrations. Although TNBCs with negligible CNAs were pre-
recent study30, published during the revision process of our viously reported21,22, their existence was questioned due to the
manuscript, we have shown that these subgroups are associated marked immune and stromal cell infiltration attributed to
with distinct survival outcomes. Moreover, our study has delved claudin-low tumors11. Here, the identification of focal genetic
into the underlying reasons of the observed heterogeneity, alterations in tumors selected for their purity unequivocally
revealing that claudin-low tumors exhibit different developmental demonstrates the existence of CNA-devoid claudin-low tumors.
origins. Beyond their stemness characteristics and their low CIN, CL1
Supporting the prevailing hypothesis that some of these tumors tumors have additional characteristic features when compared to
are generated from basal-like breast cancers, the comprehensive all other breast cancers. These traits include (i) a high expression
characterization of METABRIC and TCGA databases have of the ZEB1 EMT-inducing transcription factor and of its target,
revealed the existence of claudin-low breast cancers the methionine sulfoxide reductase MSRB3; (ii) a frequent acti-
(CL3 subgroup) with characteristics of basal mammary epithelial vation of the RAS-MAPK signaling pathway; (iii) a low activation
cells and of basal-like breast cancers. These traits include the of the DDR; and (iv) a low frequency of TP53 mutations (Sup-
hypomethylation of genes related to basal-related pathways, the plementary Figs. 8 and 10, and Fig. 6). These findings are highly
hypermethylation of genes related to luminal-related pathways, a consistent with the stemness features of CL1 tumors. Indeed, we
high level of expression of proliferation-related genes and a demonstrated recently that normal human MaSCs exhibit a pre-
strongly disturbed genomic landscape. Several experimental data emptive antioxidant program driven by ZEB1 and MSRB3, pro-
substantiate the hypothesis of the basal origin of claudin-low viding them with the unique capacity to readily adapt to an
breast cancers, including the demonstration that the oncogenic aberrant activation of the RAS-MAPK pathway18. As a con-
transformation of basal mammary epithelial cells and of LPs can sequence, RAS-driven malignant transformation of MaSCs occurs
generate malignant cells with mesenchymal characteristics in the absence of DNA damage, thereby alleviating the selective
through the activation of an EMT process17,31. However, high- pressure on the inactivation of the TP53-dependent failsafe
lighting a first degree of heterogeneity, a significant fraction of program8,18. Altogether, these findings led us to propose a model
claudin-low tumors (CL2 subgroup) displays a gene expression with two alternative paths leading to the development of claudin-
signature of mL cells and shares common genetic, epigenetic and low tumors (Fig. 7). A direct path relies upon the malignant

NATURE COMMUNICATIONS | (2020)11:3431 | https://doi.org/10.1038/s41467-020-17249-7 | www.nature.com/naturecommunications 7


ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-17249-7

a PAN-CANCER EMT SIGNATURE (Tan et al., 2014) b EMT PROTEIN SCORE (RPPA) c TP53 SIGNALING PATHWAY SIGNATURE (GO PATHWAY)

P = 6.5e-9 P = 1.1e-7 P = 2.3e-2 P = 1.3e-3 P = 0.11

1.0 P = 1.4e-7 P = 1.0e-7 12 P = 0.19 P = 7.8e-4 P = 3.1e-2


10 0.5 0.5
0.5
8

RPPA pathway scores


0.5

ssGSEA scores

ssGSEA scores
6

EMT scores
EMT scores

4
0.0 0.0
2 0.0
0.0
P = 2.5e-3 0
-0.5 P = 0.35 -2 P = 0.18
-0.5 P = 1.1e-2 P = 4.3e-5
-4
METABRIC TCGA TCGA - RPPA METABRIC TCGA
-1.0 -6 -0.5
CL1 CL2 CL3 Basal Her2 LumA LumB CL1 CL2 CL3 Basal Her2 LumA LumB CL1 CL2 CL3 Basal Her2 LumA LumB CL1 CL2 CL3 Basal Her2 LumA LumB CL1 CL2 CL3 Basal Her2 LumA LumB

P = 5.6e-23 P = 1.6e-28 P = 9.4-6 P = 1.6e-4 P = 4.3e-4

d TP53 SOMATIC MUTATION e MAPK SIGNALING SIGNATURE (KEGG PATHWAY) f RAS/MAPK PROTEIN SCORE (RPPA)

P = 2.1e-5 P = 5.3e-7 P = 0.2


METABRIC TCGA
WT TP53 Mutated TP53 WT TP53 Mutated TP53 P = 2.0e-4 P = 2.6e-10 8
100 100 P = 0.87

90 n = 14 90 0.5 6
n = 115

RPPA pathway scores


80 80 n = 28
n = 26 4

ssGSEA scores
ssGSEA scores
Percentage of tumors
Percentage of tumors

n = 76 n = 87
70 70 n = 26 0.5
n=7 n = 50
60 n = 11 60 2
n = 11
50 50 n = 12 0.0
P = 1.7e-4 0
40 40
n = 11
P = 0.053 -2
30 30 n=5
n = 417 n = 235 0.0
20 n = 14 20 n = 67 -4
n = 112
n = 19 P = 2.3e-3
n=9
n=3
n=3 n = 228 METABRIC
-0.5 TCGA
10 n = 432 10 TCGA - RPPA
n=1
n = 29
n = 21 -6
0 0 CL1 CL2 CL3 Basal Her2 LumA LumB CL1 CL2 CL3 Basal Her2 LumA LumB CL1 CL2 CL3 Basal Her2 LumA LumB
CL1 CL2 CL3 Basal Her2 LumA LumB CL1 CL2 CL3 Basal Her2 LumA LumB

P = 3.1e-12 P = 1.3e-22 P = 4.3-4

g ONCONGENIC PATHWAYS – PRINCIPAL COMPONENT ANALYSIS h SENSITIVITY TO MEK INHIBITORS (IC50)


HORMONE.PROT
HORMONE.PROT d=2

P = 9.8e-3 P = 5.4e-3 P = 3.1e-2

80 200 350

300
HORMONE.RNA 60
HORMONE.RNA 150 250

REFAMETINIB IC50 (micromolar)


SELUMETINIB IC50 (micromolar)
TRAMETINIB IC50 (micromolar)

LumA
LumA 200
40
100
150
LumB
LumB
MAPK.PROT
MAPK.RNA
MAPK.PROT MAPK.RNA 20 100
CL2
CL2
DDR.PROT
DDR.PROT CL1 50 CL1 CL1
CL1
CL1 CL2 CL2 50 CL2
CL3 CL3 CL3
Her2
Her2
DDR.RNA
DDR.RNA 15 20
2
EMT.PROT
EMT.PROT CL3
CL3
15
Basal
Basal CYCLE.RNA
CYCLE.RNA 10
PCA2 (17.89%)

1 10
EMT.RNA
EMT.RNA
5
5
CYCLE.PROT
CYCLE.PROT
0 0 0
CL Basal Her2 LumA LumB CL Basal Her2 LumA LumB CL Basal Her2 LumA LumB

PCA1 (28.96%)

Fig. 6 Claudin-low subgroups jointly display high EMT features and MAPK pathway activation. a, c, e ssGSEA score distribution for each molecular
subtype, from METABRIC and TCGA cohorts, related to a EMT, c TP53, and e RAS/MAPK pathways. d TP53 somatic mutation distribution across breast
molecular subtypes of METABRIC and TCGA cohorts. b, f Protein-based pathway scores (RPPA data) for each molecular subtype from TCGA breast
tumors related to b EMT and f RAS/MAPK. g Principal component analysis (PCA) of TCGA breast tumors according to EMT, RAS/MAPK, cell cycle/
proliferation, hormone-related and DDR pathways at RNA (ssGSEA scores), and protein (RPPA scores) levels. For each sample, molecular subtype is
indicated in color and variables (pathways) are highlighted in black squares. h IC50 distribution according to molecular subtype for three different MEK
inhibitors available in the GDSC database. Claudin-low cell lines commonly exhibit higher sensitivity to MAPK inhibitors than other breast molecular
subgroups. Wilcoxon tests. Boxplot: center line, median; box limits, upper and lower quartiles; whiskers, minimum to maximum; all data points are shown.
TP53 signaling pathway signature: GO positive regulation of signal transduction by p53 class mediator; MAPK signaling signature: KEGG MAPK signaling
pathway. DDR: DNA damage response; EMT: epithelial-mesenchymal transition; RPPA: reverse-phase protein array.

transformation of a normal MaSC, leading to undifferentiated thus reflect the high pliancy of human MaSCs for RAS activation.
tumors with a low genomic instability and a low frequency of Interestingly, although CL1 tumors show the most frequent
TP53 mutations. An indirect path relies upon the activation of an activation of the RAS-MAPK pathway, it is noteworthy that the
EMT process, over the course of tumor progression. Beyond a activation of this mitogenic pathway is also a common event in
certain extent of transdifferentiation, the gain of mesenchymal CL2 and CL3 tumors, compared with other breast cancer sub-
features triggers the conversion into a gene expression signature types. This latter finding may reflect the capacity of the RAS
of claudin-low tumor subtype. When it occurs in a basal-like signaling pathway to induce EMT in permissive conditions34–36.
tumor, EMT promotes the evolution toward a claudin-low tumor Implicit in this notion is that the activation of the RAS-MAPK
with extensive genomic aberrations as the reflect of previous pathway may be a primary oncogenic event in the tumorigenic
periods of exacerbated CIN. When this process occurs in a process leading to CL1 tumors and a secondary event in the
luminal breast cancer, it leads gradually to a claudin-low tumor course of tumor progression in a fraction of basal-like and
with a moderate level of genomic aberrations. luminal breast cancers, eventually leading to CL2 and CL3
Overall, this model illustrates the emerging concept of cellular tumors.
pliancy8. The level of pliancy is defined by the intrinsic suscept- Although this hypothesis remains to be tested, our data may
ibility of a discrete cell state to undergo malignant transformation have significant clinical implications in light of the previously
or degeneration after sustaining a particular oncogenic insult8,33. described resistance of EMT-related tumors to a variety of anti-
In this regard, the low genomic instability of CL1 tumors might cancer therapies37–39. Indeed, the identification of the recurrent

8 NATURE COMMUNICATIONS | (2020)11:3431 | https://doi.org/10.1038/s41467-020-17249-7 | www.nature.com/naturecommunications


NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-17249-7 ARTICLE

Normal Breast cancer


differentiation development
Claudin-low 1
MaSC
Transformation

FGA: LOW
STEMNESS: HIGH
MAPK: HIGH

Basal-like Claudin-low 3

LP
Transformation EMT

FGA: high FGA: high


Stemness: low Stemness: medium
MAPK: low MAPK: medium

Luminal Claudin-low 2

mL
Transformation EMT

FGA: medium FGA: medium


Stemness: low Stemness: medium
MAPK: low MAPK: medium

Fig. 7 The diversity of claudin-low tumors reflects the impact of the cell-of-origin on the evolutionary process of breast tumorigenesis. The malignant
transformation of a normal MaSC leads to undifferentiated tumors with a low genomic instability and a low frequency of TP53 mutations. These features
are characteristic of the CL1 subgroup. The activation of an EMT transdifferentiation process and the gain of mesenchymal features in a basal-like tumor
promotes the development of a CL3 tumor with extensive genomic aberrations. EMT commitment in a luminal-like breast cancer leads gradually to a CL2
tumor with a moderate level of genomic aberrations.

activation of the RAS-MAPK pathway in claudin-low breast g ∈ G and each sample s ∈ S,


cancers may lead to new therapeutic opportunities that need to be 0 1
tested in preclinical models. B FPKMðg; sÞ C
B C
TPMðg; sÞ ¼ B G C ´ 106
@P A
Methods FPKMði; sÞ
i¼1
Samples. Breast tumor samples used in this study were from METABRIC21 and
TCGA Research Network (https://www.cancer.gov/tcga) cohorts, and breast cancer RNASeq expression data from the CCLE breast cell lines were extracted as
cell lines were from CCLE database27. RPKM values from the CCLE data portal (https://portals.broadinstitute.org/ccle).
RPKM data by gene were converted to TPM as follows: for each gene g ∈ G and
each sample s ∈ S,
Statistics. All analyses and statistical tests were carried out with the R software 0 1
(version 3.6.1)40. Heatmaps were generated with ComplexeHeatmap, principal
component analysis was completed with ade4, Gaussian finite models were per- B RPKMðg; sÞ C
B C
formed with mclust and Rmixmod, and survival analyses were conducted with TPMðg; sÞ ¼ B G C ´ 106
@P A
survival R packages25,41–43. All statistical tests were two-tailed. Figures were created RPKMði; sÞ
using either the R software or GraphPad Prism 8.0 (GraphPad Software, Inc., San i¼1

Diego, USA).
Molecular breast cancer subtype assignment. Triple-negative (TN) status was
Expression data processing. METABRIC microarray expression data from dis- determined from clinical data obtained through the Synapse platform (https://
covery and validation sets were extracted from the EMBL–EBI archive (EGA, www.synapse.org/) (syn1757053) for METABRIC dataset and the GDC data portal
http://www.ebi.ac.uk/ega/; accession number: EGAS00000000083) (Normalized (https://portal.gdc.cancer.gov/) for TCGA dataset. Expression value for estrogen
expression data files)21. Normalized expression data per probe of the discovery set and progesterone receptors were combined with the SNP6 state for ERBB2 gene, to
and the validation set were combined after normalization of each set independently define the TN status of each available tumor.
with a median Z-score calculation for each probe. The expression levels of different Integrative cluster and breast cancer molecular subtype attribution were
probes associated with the same Entrez Gene ID were averaged for each sample in performed using the R package genefu44. Basal-like, luminal A, luminal B, Her2,
order to obtain a single expression value by gene. and normal-like subtype assignments were computed from five different
The Cancer Genome Atlas Breast Invasive Carcinoma (TCGA BRCA) RNA algorithms (PAM50, AIMS, SCMGENE, SSP2006, and SCMOD2)45–49. An
sequencing (RNASeq) expression data were extracted as FPKM (fragments per assignment was considered final if defined by at least three different algorithms. In
kilobase of transcript per million mapped reads) values from the Genomic Data case of divergence between classifiers, PAM50 subtype attribution was used.
Commons (GDC) data portal (https://portal.gdc.cancer.gov/). FPKM data by gene The claudin-low subtype classification was defined by the nearest centroid
were converted to transcripts per kilobase million (TPM) as follows: for each gene method. For that, the Euclidean distance between each tumor sample (from

NATURE COMMUNICATIONS | (2020)11:3431 | https://doi.org/10.1038/s41467-020-17249-7 | www.nature.com/naturecommunications 9


ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-17249-7

METABRIC and TCGA cohorts) and the previously described claudin-low and RPPA data processing. RPPA level 4 data were extracted from the cancer pro-
non-claudin-low centroids for tumor samples were determined, using the 1667 teome atlas portal (https://tcpaportal.org/tcpa/download.html)62. RPPA pathway
genes defined by Prat et al.4 as significantly differentially expressed between lists, defined by Akbani et al.28, were used to compute protein pathway scores for
claudin-low tumors and all other molecular subtypes. The tumor-related centroids each sample.
(differentially expressed genes between claudin-low and non-claudin-low tumors
using significance analysis of microarrays) were used rather than the cell-line-
Somatic mutation data. METABRIC somatic mutation data from targeted
related centroids (nine cell lines), as cell culture dramatically changes tumor
sequencing were obtained from Pereira et al.63 (https://github.com/cclab-brca/
phenotype and thus transcriptomic signature of primary samples. The use of a
mutationalProfiles/tree/master/Data) and TCGA somatic mutation data from
high-purity threshold for tumor selection allowed to circumvent the contamination
whole-exome sequencing were obtained from Ellrott et al.64. Only somatic muta-
bias related to the tumor-related centroid (Supplementary Fig. 1). For CCLE breast
tions annotated as exonic, in-frame, and non-silent were used for mutation
cancer cell lines, claudin-low status assignment was performed using the nine-cell-
analysis.
line predictor via the R package genefu44.

Drug response data. Predicted IC50 data for MEKi (Trametinib, Selumetinib,
Pathway enrichment analysis. All pathway-enrichment analyses were conducted
Refametinib) were download from Genomics of Drug Sensitivity in Can-
using MSigDB gene sets H, C2, and C5 from msigdbr R package50. GSEAs were cer (GDSC) database (https://www.cancerrxgene.org/)65.
carried out using fgsea R package51. Gene lists were pre-ranked using Signal2Noise
metric. ssGSEA scores were computed through gsva R package52. Pan-cancer
transcriptomic EMT signature, defined by Tan et al.53, was used to compute EMT Clinicopathological analysis. Complete clinical data were obtained through the
score for each sample. Synapse platform (https://www.synapse.org/) (syn1757053) for METABRIC
dataset.
For TCGA dataset, survival data were extracted from cBioPortal (TCGA
Normal mammary cell transcriptomic signatures. Previously published micro-
BRCA, PanCancer Atlas) (http://www.cbioportal.org/) and other clinical data were
array expression data were used to generate lists of differentially expressed genes
obtained from the GDC data portal (https://portal.gdc.cancer.gov/)59,60.
along the mammary epithelial cell hierarchy (MaSC1/2/3, LP, and mL1/2)18.
Microarray data were robust multiarray average-normalized through oligo R
package and differential expression analysis was performed using limma R Reporting summary. Further information on research design is available in
package54,55. The top 500 false discovery rate (FDR) p-value ranked genes differ- the Nature Research Reporting Summary linked to this article.
entially expressed between one subpopulation (MaSC or LP, or mL cells) vs. the
two others (FDR p-value < 0.05) were used as transcriptomic signature for each of
the three cell subpopulations (Supplementary Data 1).
Data availability
The complete set of CEL files from Morel et al.18 is available in the GEO database under
accession number GSE56031 and the ArrayExpress database under accession number E-
Claudin-low subgroups classifier. Expression-based classifier for the three MTAB-4145. METABRIC data are available in the EMBL–EBI archive (accession
claudin-low subgroups was identified using shrunken nearest centroid method number: EGAS00000000083) and from Supplementary Information in Curtis et al.21 and
through pamr R package26. A 1.87 threshold for centroid shrinkage was defined Pereira et al.63. TCGA BRCA RNAseq expression data were extracted as FPKM values
after examination of training errors and the cross-validation results. Finally, the from the GDC data portal (https://portal.gdc.cancer.gov/). RNAseq expression data from
nearest shrunken centroid classifier encompassed 137 genes whose expression the CCLE breast cell lines were extracted as RPKM values from the CCLE data portal
discriminate the three FGA-related claudin-low subgroups in METABRIC cohort (https://portals.broadinstitute.org/ccle). Triple-negative (TN) status was determined
(Supplementary Fig. 6). from clinical data obtained through the Synapse platform (https://www.synapse.org/)
(syn1757053) for METABRIC dataset and through the GDC data portal (https://portal.
Microenvironment analysis. Estimation of immune and non-immune cell frac- gdc.cancer.gov/) for TCGA dataset. METABRIC segmented copy number data from
tions from tumor microenvironment were determined through gene expression discovery and validation sets were extracted from the EGA (http://www.ebi.ac.uk/ega/;
analysis using Immunedeconv R package56 together with Xcell and MCPCounter accession number: EGAS00000000083) (Segmented-CBS copy number aberration (CNA)
deconvolution methods57,58. files). TCGA BRCA segmented copy number data were extracted from the GDC data
portal (https://portal.gdc.cancer.gov/). ASCAT ploidy and purity estimates were
Copy number data processing. METABRIC segmented copy number data from extracted from COSMIC data repository (https://cancer.sanger.ac.uk/ cosmic/).
discovery and validation sets were extracted from the EGA (http://www.ebi.ac.uk/ Normalized methylation β-values per gene were extracted from cBioPortal (http://www.
ega/; accession number: EGAS00000000083) (Segmented (CBS) copy number cbioportal.org/). RPPA level 4 data were extracted from The Cancer Proteome Atlas
aberrations (CNA) files)21. TCGA BRCA segmented copy number data were (TCPA) portal (https://tcpaportal.org/tcpa/download.html). METABRIC somatic
extracted from the GDC data portal repository (files corresponding to alignments mutation data from targeted sequencing were obtained from Pereira et al.63 (https://
on the hg19 version of the human genome without germline CNV were chosen). github.com/cclab-brca/mutationalProfiles/tree/master/Data) and TCGA somatic
FGAs was evaluated from TCGA and METABRIC segmented copy number data mutation data from whole-exome sequencing were obtained from Ellrott et al.64.
(both generated from Affymetrix SNP6.0 arrays) as follows: Predicted IC50 data for MEKi (Trametinib, Selumetinib, Refametinib) were download
from Genomics of Drug Sensitivity in Cancer (GDSC) database (https://www.
P P cancerrxgene.org/). Complete clinical data were obtained through the Synapse platform
Lð i Þ Lð i Þ (https://www.synapse.org/) (syn1757053) for METABRIC dataset. For TCGA dataset,
CNi>WMþT
FGA ¼ P þ P
CNi<WMT survival data were extracted from cBioPortal (Breast Invasive Carcinoma, TCGA,
ð LðiÞÞ ð Lð i Þ Þ PanCancer Atlas) (http://www.cbioportal.org/) and other clinical data were obtained
from the GDC data portal (https://portal.gdc.cancer.gov/).
For each segment i, CNi is the mean LRR along segment i, L(i) is the length of
segment i, WM is the weighted median of CNi by L(i) for each sample I, and T is Received: 19 December 2019; Accepted: 22 June 2020;
the threshold value of the CNi above which the segments are considered to be
altered. In other words, FGA is the ratio of the sum of the lengths of all segments
with signal above the threshold to the sum of all segment lengths. For CCLE cell
lines analysis, T was set as 0.2, whereas for METABRIC and TCGA tumors ana-
lysis, T was set as 0.1.
ASCAT ploidy and purity estimates were extracted from COSMIC data
repository (https://cancer.sanger.ac.uk/cosmic/)20. To avoid any substantial bias References
due to non-tumor cell contamination, tumors without available estimation of 1. Perou, C. M. et al. Molecular portraits of human breast tumours. Nature 406,
ASCAT aberrant tumor cell fraction were removed from the whole cohort. 747–752 (2000).
Furthermore, a stringent purity threshold (ASCAT purity > 0.38) was determined, 2. Cancer Genome Atlas Network. Comprehensive molecular portraits of human
by applying Wilcoxon test, to select purest tumor samples from TCGA and breast tumours. Nature 490, 61–70 (2012).
METABRIC databases when applicable. 3. Perou, C. M. Molecular stratification of triple-negative breast cancers.
Oncologist 15(Suppl 5), 39–48 (2010).
4. Prat, A. et al. Phenotypic and molecular characterization of the claudin-low
Methylation data processing. Normalized methylation β-values per gene were intrinsic subtype of breast cancer. Breast Cancer Res. 12, R68 (2010).
extracted from cBioPortal (http://www.cbioportal.org/)59,60. As previously descri- 5. Prat, A. et al. Clinical implications of the intrinsic molecular subtypes of breast
bed for genome-wide expression–methylation quantitative trait loci analysis61,
cancer. Breast 24(Suppl 2), S26–S35 (2015).
genes for which methylation β-values were negatively correlated with expression
6. Van Keymeulen, A. et al. Reactivation of multipotency by oncogenic PIK3CA
values (Spearman’s ρ < 0 and FDR p-value < 0.05) were selected for differential
induces breast tumour heterogeneity. Nature 525, 119–123 (2015).
methylation analysis (Supplementary Fig. 7).

10 NATURE COMMUNICATIONS | (2020)11:3431 | https://doi.org/10.1038/s41467-020-17249-7 | www.nature.com/naturecommunications


NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-17249-7 ARTICLE

7. Koren, S. et al. PIK3CA(H1047R) induces multipotency and multi-lineage 39. Drápela, S., Bouchal, J., Jolly, M. K., Culig, Z. & Souček, K. ZEB1: a critical
mammary tumours. Nature 525, 114–118 (2015). regulator of cell plasticity, DNA damage response, and therapy resistance.
8. Puisieux, A., Pommier, R. M., Morel, A.-P. & Lavial, F. Cellular pliancy and Front. Mol. Biosci. 7, 36 (2020).
the multistep process of tumorigenesis. Cancer Cell 33, 164–172 (2018). 40. R Core Team. R: A Language and Environment for Statistical Computing (R
9. Badve, S. et al. Basal-like and triple-negative breast cancers: a critical review Foundation for Statistical Computing, 2019).
with an emphasis on the implications for pathologists and oncologists. Mod. 41. Gu, Z., Eils, R. & Schlesner, M. Complex heatmaps reveal patterns and
Pathol. 24, 157–167 (2011). correlations in multidimensional genomic data. Bioinformatics 32, 2847–2849
10. Chin, S. F. et al. High-resolution aCGH and expression profiling identifies a (2016).
novel genomic subtype of ER negative breast cancer. Genome Biol. 8, R215 42. Dray, S. & Dufour, A. -B. The ade4 Package: implementing the duality
(2007). diagram for ecologists. J. Stat. Softw. 22 (2007).
11. Russnes, H. G., Lingjærde, O. C., Børresen-Dale, A.-L. & Caldas, C. Breast 43. Lebret, R. et al. Rmixmod: The R package of the model-based unsupervised,
cancer molecular stratification: from intrinsic subtypes to integrative clusters. supervised, and semi-supervised classification mixmod library. J. Stat. Softw.
Am. J. Pathol. 187, 2152–2162 (2017). 67, (2015).
12. Herschkowitz, J. I. et al. Identification of conserved gene expression features 44. Gendoo, D. M. A. et al. Genefu: an R/Bioconductor package for computation
between murine mammary carcinoma models and human breast tumors. of gene expression-based signatures in breast cancer. Bioinformatics 32,
Genome Biol. 8, R76 (2007). 1097–1099 (2016).
13. Prat, A. & Perou, C. M. Deconstructing the molecular portraits of breast 45. Haibe-Kains, B. et al. A three-gene model to robustly identify breast cancer
cancer. Mol. Oncol. 5, 5–23 (2011). molecular subtypes. J. Natl Cancer Inst. 104, 311–325 (2012).
14. Lim, E. et al. Aberrant luminal progenitors as the candidate target population 46. Wirapati, P. et al. Meta-analysis of gene expression profiles in breast cancer:
for basal tumor development in BRCA1 mutation carriers. Nat. Med. 15, toward a unified understanding of breast cancer subtyping and prognosis
907–913 (2009). signatures. Breast Cancer Res. 10, R65 (2008).
15. Morel, A.-P. et al. Generation of breast cancer stem cells through epithelial- 47. Parker, J. S. et al. Supervised risk predictor of breast cancer based on intrinsic
mesenchymal transition. PLoS ONE 3, e2888 (2008). subtypes. J. Clin. Oncol. 27, 1160–1167 (2009).
16. Mani, S. A. et al. The epithelial-mesenchymal transition generates cells with 48. Hu, Z. et al. The molecular portraits of breast tumors are conserved across
properties of stem cells. Cell 133, 704–715 (2008). microarray platforms. BMC Genomics 7, 96 (2006).
17. Morel, A.-P. et al. EMT inducers catalyze malignant transformation of 49. Paquet, E. R. & Hallett, M. T. Absolute assignment of breast cancer intrinsic
mammary epithelial cells and drive tumorigenesis towards claudin-low tumors molecular subtype. J. Natl Cancer Inst. 107, 357 (2015).
in transgenic mice. PLoS Genet. 8, e1002723 (2012). 50. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based
18. Morel, A.-P. et al. A stemness-related ZEB1-MSRB3 axis governs cellular approach for interpreting genome-wide expression profiles. Proc. Natl Acad.
pliancy and breast cancer genome stability. Nat. Med. 23, 568–578 (2017). Sci. USA 102, 15545–15550 (2005).
19. Dias, K. et al. Claudin-low breast cancer; clinical & pathological 51. Sergushichev, A. A. An algorithm for fast preranked gene set enrichment
characteristics. PLoS ONE 12, e0168669 (2017). analysis using cumulative statistic calculation. bioRxiv 60012. https://doi.org/
20. Van Loo, P. et al. Allele-specific copy number analysis of tumors. Proc. Natl 10.1101/060012 (2016).
Acad. Sci. USA 107, 16910–16915 (2010). 52. Hänzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis
21. Curtis, C. et al. The genomic and transcriptomic architecture of 2,000 breast for microarray and RNA-Seq data. BMC Bioinformatics 14, 7 (2013).
tumours reveals novel subgroups. Nature 486, 346–352 (2012). 53. Tan, T. Z. et al. Epithelial-mesenchymal transition spectrum quantification
22. Dawson, S.-J., Rueda, O. M., Aparicio, S. & Caldas, C. A new genome-driven and its efficacy in deciphering survival and drug responses of cancer patients.
integrated classification of breast cancer and its implications. EMBO J. 32, EMBO Mol. Med. 6, 1279–1293 (2014).
617–628 (2013). 54. Carvalho, B. S. & Irizarry, R. A. A framework for oligonucleotide microarray
23. Song, S. et al. qpure: A tool to estimate tumor cellularity from genome-wide preprocessing. Bioinformatics 26, 2363–2367 (2010).
single-nucleotide polymorphism profiles. PLoS ONE 7, e45835 (2012). 55. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-
24. Biernacki, C., Celeux, G., Govaert, G. & Langrognet, F. Model-based cluster sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).
and discriminant analysis with the MIXMOD software. Comput. Stat. Data 56. Sturm, G. et al. Comprehensive evaluation of transcriptome-based cell-type
Anal. 51, 587–600 (2006). quantification methods for immuno-oncology. Bioinformatics 35, i436–i445
25. Scrucca, L., Fop, M., Murphy, T. B. & Raftery, A. E. mclust 5: clustering, (2019).
classification and density estimation using Gaussian finite mixture models. R. 57. Aran, D., Hu, Z. & Butte, A. J. xCell: digitally portraying the tissue cellular
J. 8, 289–317 (2016). heterogeneity landscape. Genome Biol. 18, 220 (2017).
26. Tibshirani, R., Hastie, T., Narasimhan, B. & Chu, G. Diagnosis of multiple 58. Becht, E. et al. Estimating the population abundance of tissue-infiltrating
cancer types by shrunken centroids of gene expression. Proc. Natl Acad. Sci. immune and stromal cell populations using gene expression. Genome Biol. 17,
USA 99, 6567–6572 (2002). 218 (2016).
27. Barretina, J. et al. The Cancer Cell Line Encyclopedia enables predictive 59. Cerami, E. et al. The cBio cancer genomics portal: an open platform for exploring
modelling of anticancer drug sensitivity. Nature 483, 603–607 (2012). multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).
28. Akbani, R. et al. A pan-cancer proteomic perspective on The Cancer Genome 60. Gao, J. et al. Integrative analysis of complex cancer genomics and clinical
Atlas. Nat. Commun. 5, 3887 (2014). profiles using the cBioPortal. Sci. Signal. 6, pl1 (2013).
29. Sabatier, R. et al. Claudin-low breast cancers: clinical, pathological, molecular 61. Fleischer, T. et al. DNA methylation at enhancers identifies distinct breast
and prognostic characterization. Mol. Cancer 13, 228 (2014). cancer lineages. Nat. Commun. 8, 1379 (2017).
30. Fougner, C., Bergholtz, H., Norum, J. H. & Sørlie, T. Re-definition of claudin- 62. Li, J. et al. TCPA: a resource for cancer functional proteomics data. Nat.
low as a breast cancer phenotype. Nat. Commun. 11, 1787 (2020). Methods 10, 1046–1047 (2013).
31. Nguyen, L. V. et al. Barcoding reveals complex clonal dynamics of de novo 63. Pereira, B. et al. The somatic mutation profiles of 2,433 breast cancers refine
transformed human mammary cells. Nature 528, 267–271 (2015). their genomic and transcriptomic landscapes. Nat. Commun. 7, 11479
32. Dhasarathy, A., Phadke, D., Mav, D., Shah, R. R. & Wade, P. A. The (2016).
transcription factors Snail and Slug activate the transforming growth factor- 64. Ellrott, K. et al. Scalable open science approach for mutation calling of tumor
beta signaling pathway in breast cancer. PLoS ONE 6, e26514 (2011). exomes using multiple genomic pipelines. Cell Syst. 6, 271–281.e7 (2018).
33. Chen, X., Pappo, A. & Dyer, M. A. Pediatric solid tumor genomics and 65. Yang, W. et al. Genomics of Drug Sensitivity in Cancer (GDSC): a resource for
developmental pliancy. Oncogene 34, 5207–5215 (2015). therapeutic biomarker discovery in cancer cells. Nucleic Acids Res. 41,
34. Zhang, K. et al. Oncogenic K-Ras upregulates ITGA6 expression via FOSL1 to D955–D961 (2013).
induce anoikis resistance and synergizes with αV-Class integrins to promote
EMT. Oncogene 36, 5681–5694 (2017).
35. Zhang, X., Cheng, Q., Yin, H. & Yang, G. Regulation of autophagy and EMT
by the interplay between p53 and RAS during cancer progression (Review). Acknowledgements
Int. J. Oncol. 51, 18–24 (2017). We thank B. Manship for proofreading the manuscript. This work was supported by
36. Voon, D. C.-C. et al. EMT-induced stemness and tumorigenicity are fueled by funding from the Ligue Nationale contre le Cancer (EL2016.LNCC/AIP and EL2019.
the EGFR/Ras pathway. PLoS ONE 8, e70427 (2013). LNCC/AIP) and SIRIC LYriCAN (INCa-DGOS-Inserm_12563). This work was addi-
37. Zhang, P. et al. ATM-mediated stabilization of ZEB1 promotes DNA damage tionally supported by funding from Inserm in the framework of the International
response and radioresistance through CHK1. Nat. Cell Biol. 16, 864–875 Associated Laboratory between Cancer Research Centre of Lyon, France, and the Vic-
(2014). torian Comprehensive Cancer Centre of Melbourne, Australia (Appel à projets LIA/LEA
38. Farmer, P. et al. A stroma-related gene signature predicts resistance to 2016, ASC17019CSA). This work was additionally supported by funding from Le Cancer
neoadjuvant chemotherapy in breast cancer. Nat. Med. 15, 68–74 (2009). du sein. Parlons-en! association.

NATURE COMMUNICATIONS | (2020)11:3431 | https://doi.org/10.1038/s41467-020-17249-7 | www.nature.com/naturecommunications 11


ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-17249-7

Author contributions Reprints and permission information is available at http://www.nature.com/reprints


R.M.P. performed all bioinformatics and statistical analyses, prepared figures and
methods, and assisted in data interpretation. M.O. and A.S. assisted in preparation of Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in
figures and manuscript writing. L.T., J.K., E.T., A.F., A.-S.S., F.H., and P.M. assisted in published maps and institutional affiliations.
data interpretation. A.T. and A.-P.M. provided scientific content. F.H. provided critical
evaluation of the manuscript and scientific content. A.P. conceived the project, designed
analyses, interpreted data, and wrote the manuscript. All authors read and approved the Open Access This article is licensed under a Creative Commons
final version of the manuscript. Attribution 4.0 International License, which permits use, sharing,
adaptation, distribution and reproduction in any medium or format, as long as you give
Competing interests appropriate credit to the original author(s) and the source, provide a link to the Creative
The authors declare no competing interests. Commons license, and indicate if changes were made. The images or other third party
material in this article are included in the article’s Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the
Additional information article’s Creative Commons license and your intended use is not permitted by statutory
Supplementary information is available for this paper at https://doi.org/10.1038/s41467-
regulation or exceeds the permitted use, you will need to obtain permission directly from
020-17249-7.
the copyright holder. To view a copy of this license, visit http://creativecommons.org/
licenses/by/4.0/.
Correspondence and requests for materials should be addressed to A.P.

Peer review information Nature Communications thanks Brian Lehmannand and © The Author(s) 2020
Vessela Kristensen for their contribution to the peer review of this work. Peer reviewer
reports are available.

12 NATURE COMMUNICATIONS | (2020)11:3431 | https://doi.org/10.1038/s41467-020-17249-7 | www.nature.com/naturecommunications

You might also like